Neuropathy 1
Neuropathy 1
REVIEW OF
NEUROBIOLOGY
VOLUME 127
SERIES EDITORS
R. ADRON HARRIS
Waggoner Center for Alcohol and Drug Addiction Research
The University of Texas at Austin
Austin, Texas, USA
PETER JENNER
Division of Pharmacology and Therapeutics
GKT School of Biomedical Sciences
King's College, London, UK
EDITORIAL BOARD
ERIC AAMODT HUDA AKIL
PHILIPPE ASCHER MATTHEW J. DURING
DONARD S. DWYER DAVID FINK
MARTIN GIURFA BARRY HALLIWELL
PAUL GREENGARD JON KAAS
NOBU HATTORI LEAH KRUBITZER
DARCY KELLEY KEVIN MCNAUGHT
BEAU LOTTO A. OBESO
JOSE
MICAELA MORELLI CATHY J. PRICE
JUDITH PRATT SOLOMON H. SNYDER
EVAN SNYDER STEPHEN G. WAXMAN
JOHN WADDINGTON
Academic Press is an imprint of Elsevier
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, USA
525 B Street, Suite 1800, San Diego, CA 92101-4495, USA
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, UK
125 London Wall, London, EC2Y 5AS, UK
No part of this publication may be reproduced or transmitted in any form or by any means,
electronic or mechanical, including photocopying, recording, or any information storage and
retrieval system, without permission in writing from the publisher. Details on how to seek
permission, further information about the Publisher’s permissions policies and our
arrangements with organizations such as the Copyright Clearance Center and the Copyright
Licensing Agency, can be found at our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by
the Publisher (other than as may be noted herein).
Notices
Knowledge and best practice in this field are constantly changing. As new research and
experience broaden our understanding, changes in research methods, professional practices,
or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in
evaluating and using any information, methods, compounds, or experiments described
herein. In using such information or methods they should be mindful of their own safety and
the safety of others, including parties for whom they have a professional responsibility.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of
products liability, negligence or otherwise, or from any use or operation of any methods,
products, instructions, or ideas contained in the material herein.
ISBN: 978-0-12-803915-1
ISSN: 0074-7742
V. Bril
Ellen and Martin Prosserman Centre for Neuromuscular Diseases, University Health
Network, University of Toronto, Toronto, ON, Canada
N.A. Calcutt
University of California, San Diego, La Jolla, CA, United States
C. Casellini
Eastern Virginia Medical School, Strelitz Diabetes and Neuroendocrine Center, Norfolk,
VA, United States
R.T. Dobrowsky
The University of Kansas, Lawrence, KS, United States
S.M. Emery
The University of Kansas, Lawrence, KS, United States
P. Fernyhough
University of Manitoba; St. Boniface Hospital Research Centre, Winnipeg, MB, Canada
O.J. Freeman
University of Manchester, Manchester, United Kingdom
N.J. Gardiner
University of Manchester, Manchester, United Kingdom
R.A. Malik
Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
C. Martin
University of Michigan Medical School, Ann Arbor, MI, United States
M.-L. Névoret
Impeto Medical Inc., San Diego, CA, United States
R. Pop-Busui
University of Michigan Medical School, Ann Arbor, MI, United States
S.M. Todorovic
School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO,
United States
A.I. Vinik
Eastern Virginia Medical School, Strelitz Diabetes and Neuroendocrine Center, Norfolk,
VA, United States
S. Yagihashi*
Hirosaki University Graduate School of Medicine, Hirosaki, Japan
*Present address: The Nukada Institute of Medical and Biological Research, 4-16 Inage-machi,
Inage-ku, Chiba, Japan. E-mail: [email protected].
xi
xii Contributors
M.A. Yorek
Iowa City Health Care System; Fraternal Order of Eagles Diabetes Research Center,
University of Iowa, Iowa City, IA, United States
D.W. Zochodne
Neuroscience and Mental Health Institute and Alberta Diabetes Institute, University of
Alberta, Edmonton, AB, Canada
PREFACE
xiii
xiv Preface
clinical trials, but we need to understand the mechanism behind it. If the
factors underlying the pain were common to those precipitating the later
neurodegeneration, then it would be a valuable therapeutic focus. An inter
vention that had a powerful effect on neuropathic pain via its underlying
cause, rather than nonspecific analgesia, might well be useful in trials and
it would have the added advantage of ameliorating symptoms, rather than
impacting on signs, such as electrophysiology, which might be unrelated
to the condition or develop too late, when the pathology has become irre
versible. Thus, the chapter by Slobodan Todorovic, dissecting the pain
mechanisms, is of great value to our understanding.
This new book returns to the consideration of the value of modeling the
disease and/or its components. Nigel Calcutt suggests that clinicians, as super
intendents of failed clinical trials, are not entitled to blame inaccuracies of ani
mal models for their failures. We are long past the notion that animal models
can holistically represent human diabetic neuropathy. Instead the focus must
be to use animals to model specific components of the dysmetabolisms inher
ent in diabetes and to gauge their impact on the nervous system. In this
context, Mark Yorek has introduced a rat model with the defects associated
with Type II diabetes. This will give a metabolic profile that differs from the
conventional streptozotocin-diabetic rat and may include or rule out some of
the candidate metabolic precursors for neuropathy.
The same questions and constraints should be applied to experiments
based on neurons in culture. These are entirely valid as a means of tracking
subcellular changes related to glucose and its sequelae. Indeed, as Natalie
Gardiner and Ollie Freeman illustrate in their chapter, studies on nerve cul
tures can give a uniquely clean identification of neuronal targets for glucose
toxicity. That done, the requirement is to show that these targets are features
of the human condition.
Interpretation of findings from such models demands two questions.
(1) Does the model register a deviant or exaggerated metabolic anomaly that
is relevant to human diabetes? (2) Are the direct consequences of this
dysmetabolism present in or relevant to human diabetes? The polyol path
way serves as an excellent example. Widely studied in the rat model, the
answer to (1) is yes, but the answer to (2) is maybe, which explains the uncer
tainty that still surrounds the efficacy of aldose reductase inhibitors. Thus,
in vitro and in vivo models are acceptable approaches to mechanisms, but
that is as far as their predictive values go. I suggest, therefore, that debates
of the “rat vs mouse” type are best left to back-stage discussions and not
dragged out in front of the audience.
xvi Preface
It follows from the above that, unless these two questions have positive
answers for an end point in a model, efficacy for a potential treatment in
animals cannot be a certain springboard for clinical trials. Again, aldose
reductase inhibitors authenticate this assertion. Thus, for many clinical trials
that have been run, their failure might be blamed on interpretation of data
from the model, at least in part.
So, we come to the issue of clinical trials. Both the old and the new
volumes have addressed this. A systematic and scientific approach to under
standing diabetic neuropathy was kick-started in 1979 by a review published
by Rex Clements entitled “Diabetic neuropathy: new concepts of its
etiology” (Diabetes 28: 604–611), so we are not far from 50 years of research
with no therapeutic outcomes. It would not be unreasonable for the finger
of blame to be looking for targets. It might hover over clinical trials. I do not
know how many compounds have been through clinical trials for diabetic
neuropathy, but it is well into double figures, and the sad fact is that not only
have no successes emerged, but we cannot be certain that the agents tested
were failures. To generalize, the trials have been too short and we cannot be
sure that the right end points were selected. As Vera Bril points out in this
volume, trials lasting 5–8 years will be required to demonstrate clinical ben
efits. Rayaz Malik reviews potential end points for identification of patients
at risk or for inclusion in trials. None of these is entirely satisfactory for either
purpose. Clear signs of true neuropathy probably indicate the condition is
irreversible.
There are additional problems in finding suitable treatment. A successful
agent may have to be given prophylactically for neuropathy, perhaps starting
with the first signs of background retinopathy as an indicator of susceptibility
to diabetes complications. This would mean that significant side effects
might make the treatment unacceptable for asymptomatic patients. Thus,
I can think of no more difficult challenges to pharmacologists and clinical
trial designers than diabetic neuropathy, especially when duration of trials
and numbers of patients are constrained by finance and the acceptable pace
of the return on investment. Let us hope that this book spurs the scientists to
reveal the appropriate drug targets and that the industry keeps faith enough
to rise to the challenge.
D. TOMLINSON
University of Manchester, Manchester, United Kingdom
CHAPTER ONE
The earliest clinical trials in subjects with diabetic neuropathy targeted pain
as the primary end point (Kastrup, Angelo, Petersen, et al., 1986; Kvinesdal,
Molin, Froland, et al., 1984; Rull, Quibrera, Gonzalez-Millan, et al., 1969;
Saudek, Werns, & Reidenberg, 1977). This approach recognized that pain
was a particularly disruptive feature of diabetic neuropathy and took the
pragmatic view that, in the absence of known pathogenic mechanisms, drugs
that produced pain relief in other conditions might show benefit in diabetic
patients. Sadly, the limited advances made in understanding the pathogenesis
of pain in diabetes and difficulties inherent in translating preclinical research
to therapeutic use mean that this speculative approach of almost half a cen-
tury ago is not dissimilar to many current drug development programs for
painful diabetic neuropathy. Recent progress in identifying the pathogenesis
of painful diabetic neuropathy is discussed in detail elsewhere in this volume.
It is also noteworthy that other aspects of nerve function, such as large fiber
conduction velocity and action potential amplitudes, were initially included
in clinical trials primarily to establish that any pain relief achieved by ther-
apeutic intervention was not due to general neurotoxicity of the drugs under
investigation. However, reports that glucose metabolism through the polyol
pathway contributed to large fiber nerve conduction velocity (NCV)
slowing in diabetic rodents (Tomlinson, Holmes, & Mayer, 1982) supported
the evolution of large fiber electrophysiological dysfunction into the de
facto biomarker for diabetic peripheral neuropathy. A statistically signifi-
cant improvement of conduction velocity slowing persists as the primary
#
International Review of Neurobiology, Volume 127 2016 Elsevier Inc. 3
ISSN 0074-7742 All rights reserved.
http://dx.doi.org/10.1016/bs.irn.2016.03.014
4 N.A. Calcutt and P. Fernyhough
along with low numbers of subjects, recruitment of subjects with severe and
probably irreversible neuropathy, and an inability to assess efficacy against
the proposed pathogenic mechanism (nerve polyol pathway flux, oxidative
damage, etc.), have been widely acknowledged as weaknesses that could
have limited the success of early clinical trials. Improving glycemic control
allows for a clear and well-validated readout of subject compliance and treat-
ment efficacy via HbA1C levels. The initial successes against NCV slowing
led to instigation of large multicenter clinical trials of intensive glycemic
control such as the Oslo (Amthor, Dahl-Jorgensen, Berg, et al., 1994),
DCCT (1988), and EDIC studies. These studies were careful to address trial
design concerns and emphasized standardized objective measures of nerve
function. Their influence on clinical trial design has persisted to the present.
The DCCT and EDIC trials codified many good practices for clinical
trials. These include long durations of treatment, use of multiple trial sites,
recruitment of large numbers of subjects with mild to moderate neuropathy,
implementation of standardized protocols with objective measurements, and
the tracking of subject compliance. However, their very success may also
have introduced an unhelpful rigidity to subsequent decades of preclinical
and clinical research in diabetic neuropathy, specifically in their focus on
hyperglycemia as the primary pathogenic mechanism and large fiber
NCV slowing as the primary readout of efficacy. Many of the current con-
troversies in diabetic neuropathy arise from an evolving appreciation that the
prolonged intensive insulin therapy of the DCCT was only partially effective
and that risk factors such as hypertension and dyslipidemia may contribute
independently to neuropathy. The capacity of insulin to act as a direct tro-
phic factor for peripheral nerve and the absence of C-peptide replacement
during insulin therapy may also contribute to both the apparent efficacy of
intensive glycemic control and its incomplete efficacy in studies such as the
DCCT. Further, the focus on large fiber NCV slowing as the primary read-
out of treatment efficacy may constrain use of technological advances in the
objective quantification of small fiber neuropathy to preferentially deter-
mine efficacy of agents that specifically target this aspect of diabetic
neuropathy.
The persistent failure of clinical trials of agents to treat diabetic neurop-
athy is frequently attributed to poor drug design and inappropriate animal
models that identify pathogenic mechanisms that are not pertinent to
humans. However, at the other end of the pipeline, recognition that one
size (of trial design) may not fit all (therapeutic approaches) should allow
a flexibility that embraces advances in how diabetic neuropathy is assessed
History and Controversies of Clinical Trials in Diabetic Neuropathy 7
so that both new and old therapies can be evaluated in a scientifically rational
and clinically meaningful manner.
REFERENCES
Factors in development of diabetic neuropathy. Baseline analysis of neuropathy in feasibility
phase of Diabetes Control and Complications Trial (DCCT). The DCCT Research
Group. (1988). Diabetes, 37, 476–481.
Abraham, R. R., Abraham, R. M., & Wynn, V. (1984). A double blind placebo controlled
trial of mixed gangliosides in diabetic peripheral and autonomic neuropathy. Advances in
Experimental Medicine & Biology, 174, 607–624.
Amthor, K. F., Dahl-Jorgensen, K., Berg, T. J., et al. (1994). The effect of 8 years of strict
glycaemic control on peripheral nerve function in IDDM patients: The Oslo Study.
Diabetologia, 37, 579–584.
Calcutt, N. A., Jolivalt, C. G., & Fernyhough, P. (2008). Growth factors as therapeutics for
diabetic neuropathy. Current Drug Targets, 9, 47–59.
Calcutt, N. A., Willars, G. B., & Tomlinson, D. R. (1988). Statil-sensitive polyol formation
in nerve of galactose-fed mice. Metabolism, 37, 450–453.
Chen, X., Graham, J., Dabbah, M. A., et al. (2015). Small nerve fiber quantification in the
diagnosis of diabetic sensorimotor polyneuropathy: Comparing corneal confocal micros-
copy with intraepidermal nerve fiber density. Diabetes Care, 38, 1138–1144.
Crepaldi, G., Fedele, D., Tiengo, A., et al. (1983). Ganglioside treatment in diabetic periph-
eral neuropathy: A multicenter trial. Acta Diabetologica Latina, 20, 265–276.
Dahl-Jorgensen, K., Brinchmann-Hansen, O., Hanssen, K. F., et al. (1986). Effect of near
normoglycaemia for two years on progression of early diabetic retinopathy, nephropa-
thy, and neuropathy: The Oslo study. British Medical Journal (Clinical Research Ed), 293,
1195–1199.
Dyck, P. J., & O’Brien, P. C. (1989). Meaningful degrees of prevention or improvement of
nerve conduction in controlled clinical trials of diabetic neuropathy. Diabetes Care, 12,
649–652.
Dyck, P. J., Sherman, W. R., Hallcher, L. M., et al. (1980). Human diabetic endoneurial
sorbitol, fructose, and myo-inositol related to sural nerve morphometry. Annals of Neu-
rology, 8, 590–596.
Fagius, J., & Jameson, S. (1981). Effects of aldose reductase inhibitor treatment in
diabetic polyneuropathy—A clinical and neurophysiological study. Journal of Neurology,
Neurosurgery & Psychiatry, 44, 991–1001.
Fukuda, Y., Fukui, T., Hikichi, C., et al. (2015). Neurotropin promotes NGF signaling
through interaction of GM1 ganglioside with Trk neurotrophin receptor in PC12 cells.
Brain Research, 1596, 13–21.
Greene, D. A., Brown, M. J., Braunstein, S. N., et al. (1981). Comparison of clinical course
and sequential electrophysiological tests in diabetics with symptomatic polyneuropathy
and its implications for clinical trials. Diabetes, 30, 139–147.
Greene, D. A., De Jesus, P. V., Jr., & Winegrad, A. I. (1975). Effects of insulin and dietary
myoinositol on impaired peripheral motor nerve conduction velocity in acute
streptozotocin diabetes. Journal of Clinical Investigation, 55, 1326–1336.
Gregersen, G., Bertelsen, B., Harbo, H., et al. (1983). Oral supplementation of myoinositol:
Effects on peripheral nerve function in human diabetics and on the concentration in
plasma, erythrocytes, urine and muscle tissue in human diabetics and normals. Acta Neu-
rologica Scandinavica, 67, 164–172.
Jaspan, J., Maselli, R., Herold, K., et al. (1983). Treatment of severely painful diabetic neu-
ropathy with an aldose reductase inhibitor: Relief of pain and improved somatic and
autonomic nerve function. Lancet, 2, 758–762.
8 N.A. Calcutt and P. Fernyhough
Judzewitsch, R. G., Jaspan, J. B., Polonsky, K. S., et al. (1983). Aldose reductase inhibition
improves nerve conduction velocity in diabetic patients. New England Journal of Medicine,
308, 119–125.
Kastrup, J., Angelo, H., Petersen, P., et al. (1986). Treatment of chronic painful diabetic neu-
ropathy with intravenous lidocaine infusion. British Medical Journal (Clinical Research Ed.),
292, 173.
Kessler, J. A., Smith, A. G., Cha, B. S., et al. (2015). Double-blind, placebo-controlled study
of HGF gene therapy in diabetic neuropathy. Annals of Clinical and Translational Neurol-
ogy, 2, 465–478.
Kvinesdal, B., Molin, J., Froland, A., et al. (1984). Imipramine treatment of painful diabetic
neuropathy. JAMA, 251, 1727–1730.
Levin, E. R., Hanscom, T. A., Fisher, M., et al. (1981). The influence of pyridoxine in dia-
betic peripheral neuropathy. Diabetes Care, 4, 606–609.
Malik, R. A. (2014). Which test for diagnosing early human diabetic neuropathy? Diabetes,
63, 2206–2208.
Miwa, I., Kanbara, M., & Okuda, J. (1989). Improvement of nerve conduction velocity in
mutant diabetic mice by aldose reductase inhibitor without affecting nerve myo-inositol
content. Chemical & Pharmaceutical Bulletin (Tokyo), 37, 1581–1582.
Nishio, M., Fukumoto, S., Furukawa, K., et al. (2004). Overexpressed GM1 suppresses nerve
growth factor (NGF) signals by modulating the intracellular localization of NGF recep-
tors and membrane fluidity in PC12 cells. Journal of Biological Chemistry, 279,
33368–33378.
Oates, P. J. (2008). Aldose reductase, still a compelling target for diabetic neuropathy. Current
Drug Targets, 9, 14–36.
Papanas, N., & Ziegler, D. (2014). Efficacy of alpha-lipoic acid in diabetic neuropathy. Expert
Opinion on Pharmacotherapy, 15, 2721–2731.
Polydefkis, M., Arezzo, J., Nash, M., et al. (2015). Safety and efficacy of ranirestat in patients
with mild-to-moderate diabetic sensorimotor polyneuropathy. Journal of the Peripheral
Nervous System, 20, 363–371.
Rull, J. A., Quibrera, R., Gonzalez-Millan, H., et al. (1969). Symptomatic treatment of
peripheral diabetic neuropathy with carbamazepine (Tegretol): Double blind crossover
trial. Diabetologia, 5, 215–218.
Sachse, G., & Willms, B. (1980). Efficacy of thioctic acid in the therapy of peripheral diabetic
neuropathy. Hormone and Metabolic Research Supplement Series, 9, 105–107.
Salway, J. G., Whitehead, L., Finnegan, J. A., et al. (1978). Effect of myo-inositol on
peripheral-nerve function in diabetes. Lancet, 2, 1282–1284.
Saudek, C. D., Werns, S., & Reidenberg, M. M. (1977). Phenytoin in the treatment of dia-
betic symmetrical polyneuropathy. Clinical Pharmacology and Therapeutics, 22, 196–199.
Service, F. J., Rizza, R. A., Daube, J. R., et al. (1985). Near normoglycaemia improved
nerve conduction and vibration sensation in diabetic neuropathy. Diabetologia, 28,
722–727.
Tomlinson, D. R., Holmes, P. R., & Mayer, J. H. (1982). Reversal, by treatment with an
aldose reductase inhibitor, of impaired axonal transport and motor nerve conduction
velocity in experimental diabetes mellitus. Neuroscience Letters, 31, 189–193.
Vieira, K. P., de Almeida e Silva Lima Zollner, A. R., Malaguti, C., et al. (2008). Ganglioside
GM1 effects on the expression of nerve growth factor (NGF), Trk-A receptor,
proinflammatory cytokines and on autoimmune diabetes onset in non-obese diabetic
(NOD) mice. Cytokine, 42, 92–104.
Young, R. J., Ewing, D. J., & Clarke, B. F. (1983). A controlled trial of sorbinil, an aldose
reductase inhibitor, in chronic painful diabetic neuropathy. Diabetes, 32, 938–942.
Ziegler, D., Low, P. A., Freeman, R., et al. (2016). Predictors of improvement and progres-
sion of diabetic polyneuropathy following treatment with alpha-lipoic acid for 4 years in
the NATHAN 1 trial. Journal of Diabetes and Its Complications, 30, 350–356.
CHAPTER TWO
Contents
1. Introduction 10
2. Neuropathy Outcomes Assessments 11
2.1 DCCT and EDIC Design 11
3. Complementary Assessments in EDIC 16
3.1 Evaluation of Urologic Complications 16
3.2 Gastroparesis 16
3.3 Other Evaluations in EDIC 17
4. DCCT/EDIC Findings 17
4.1 DSPN and CAN Outcomes in the DCCT and EDIC Study 17
5. Discussion 19
5.1 DCCT/EDIC and Contemporary Neuropathy Trials Design 21
References 23
Abstract
The Diabetes Control and Complications Trial (DCCT) and its epidemiological follow-up,
the Epidemiology of Diabetes Interventions and Complications (EDIC) provide impor-
tant insight on the natural history of distal symmetrical polyneuropathy and cardiovas-
cular autonomic neuropathy in patients with type 1 diabetes and on the impact of
intensive treatment of hyperglycemia on disease progression. This chapter summarizes
the design and methods used for neuropathy evaluations both in the DCCT and in
EDIC, the characteristics of the DCCT/EDIC patient population, and summarizes the
findings of the DCCT/EDIC relative to neuropathic complications of type 1 diabetes.
Lessons learned from the DCCT and EDIC experiences of longitudinal assessments of
neuropathic complications are also reviewed.
1. INTRODUCTION
Diabetic neuropathies are among the most prevalent of the chronic
complications of diabetes (Boulton et al., 2005; Tesfaye et al., 2010), with
a broad spectrum of forms and clinical manifestations. The most studied in
clinical trials are distal (length-dependent) symmetrical sensorimotor poly-
neuropathy (DSPN) and cardiovascular autonomic neuropathy (CAN).
Outcome measures specific to DSPN and CAN forms were included in
the Diabetes Control and Complications Trial (DCCT) design and will
be discussed in this chapter.
The DCCT enrolled 1441 patients with type 1 diabetes between 1983
and 1989 at 28 clinical sites across United States and Canada (DCCT, 1993).
The DCCT included a primary prevention cohort and a secondary inter-
vention cohort. The primary prevention cohort had diabetes for 1–5 years
(mean 2.6 years) and no retinopathy at baseline. The secondary intervention
cohort had diabetes for 1–15 years (mean 8.7 years) and mild-to-moderate
retinopathy at baseline (DCCT, 1993). Patients were randomly assigned
to intensive (INT) or conventional (CONV) insulin therapy. Briefly, the
intensive therapy group used either insulin pumps, or three or more daily
injections of insulin, together with frequent daily self-blood glucose
monitoring in order to achieve blood glucose levels as close as possible to
normal. The conventional therapy group used one or two injections of insu-
lin daily, without routine blood glucose monitoring with a goal of minimal
symptoms related to hyper- or hypoglycemia. In 1993, after an average of
6.5 years of follow-up, HbA1c was 7.4% in the intensive group and 9.1%
in the conventional group (DCCT, 1993). The HbA1c improvement in
the intensive group was associated with significantly reduced incidence of
diabetic retinopathy, nephropathy, and neuropathy and the randomized,
clinical trial was therefore stopped (DCCT, 1993, 1995, 1998). Participants
from both the INT and CONV groups were encouraged to continue or to
adopt an intensive diabetes treatment regimen and DCCT CONV group
subjects were instructed in intensive therapy by the DCCT treatment teams.
By the end of 1993, after an average of 6.5 years of follow-up, all DCCT
participants were returned to their prior health care providers for
ongoing care.
The DCCT finding that intensive insulin therapy of type 1 diabetes to
improve glycemia profoundly reduced complications of type 1 diabetes trig-
gered fundamental changes in the management of diabetes in the United
Neuropathy in the DCCT/EDIC 11
MNSI
DSPN DSPN Urologic
Urologic yearly
CAN CAN (ED, LUTS,
(ED, LUTS, DSPN: 1186 FSD, UI)
CAN CAN FSD, UI) CAN: 1226 CAN
(1214)
EDIC Start
1375 (94%)
Entire cohort
DCCT EDIC
Fig. 1 Timeline of main and other neuropathy evaluations in the DCCT/EDIC. CAN,
cardiovascular autonomic neuropathy; DSPN, distal symmetrical polyneuropathy;
ED, erectile dysfunction; FSD, female sexual dysfunction; LUTS, lower urinary tract
symptoms; MNSI, Michigan Neuropathy Screening Instrument; UI, urinary incontinence.
presence of DSPN (Feldman et al., 1994; Herman et al., 2012). Using the
MNSI, DSPN was defined as either an MNSI questionnaire score of 7 or
an MNSI exam score of 2.5; these scores being based on the initial vali-
dation of the MNSI instrument (Feldman et al., 1994). The MNSI evalua-
tions through EDIC year 8 suggested that the metabolic memory
phenomenon observed for retinal and renal complications, also applied to
new-onset (incident) neuropathy (Martin et al., 2006). The MNSI however,
was not designed to evaluate DSPN severity nor was it initially conceived as
a tool for longitudinal assessment of neuropathy. Further, it lacked the sen-
sitivity and specificity of the original DCCT evaluations and therefore could
not be used as a surrogate for the robust DCCT primary outcome of con-
firmed clinical neuropathy.
3.2 Gastroparesis
The DCCT did not assess gastroparesis, arguably one of the most debilitating
autonomic complications of diabetes and certainly one of the most frustrat-
ing from a clinical management perspective. In EDIC, the assessment of
gastroparesis in the entire cohort is entirely based on self-report of symptoms
obtained at annual EDIC visits. However, a formal assessment of gastric
emptying as a measure of gastroparesis was performed in 78 DCCT/EDIC
participants at EDIC year 20 using the 13C-Spirulina gastric emptying
breath test (GEBT, 223 kcal with 19.2 g carbohydrates, 12 g protein, and
10.9 g fat; AB Diagnostics, Brentwood, TN) under an investigator-initiated
Neuropathy in the DCCT/EDIC 17
investigational new drug (IND) from the Food and Drug Administration
(FDA) as described (Bharucha et al., 2015).
4. DCCT/EDIC FINDINGS
4.1 DSPN and CAN Outcomes in the DCCT and EDIC Study
DSPN (confirmed clinical neuropathy) and CAN were uncommon at
DCCT start, in part owing to the intentional exclusion of persons with
symptomatic neuropathy (DCCT, 1993; Martin et al., 2014; Pop-Busui
et al., 2010). The prevalence of confirmed clinical neuropathy increased
substantially in the conventional subjects (from 5% to 17%, P < 0.001)
and only slightly among the intensive group participants (from 7% to 9%;
DCCT, 1993, 1995; Martin et al., 2014; Pop-Busui et al., 2010). Adjusting
for the presence of confirmed DSPN at baseline, the risk reduction for inci-
dent DSPN with intensive glucose control during DCCT was 64% (95% CI,
45–76) (DCCT, 1993, 1995; Martin et al., 2014).The prevalence of CAN
almost doubled in the conventional group by DCCT-end, while remaining
static in the intensive group (DCCT, 1993, 1998). The risk reduction in
incident CAN with intensive therapy during DCCT was 45% (DCCT,
1993, 1998; Martin et al., 2014; Pop-Busui et al., 2010).
During EDIC, the HbA1c separation between former intensive and con-
ventional treatment groups observed through the DCCT quickly waned and
by the fifth year of EDIC follow-up, the HbA1c separation between treat-
ment groups completely disappeared (Martin et al., 2014; Pop-Busui et al.,
2009; Writing Team for DCCT/EDIC, 2003). Prevalence of DSPN
increased during EDIC in both groups and, despite no measureable differ-
ence in glucose control from EDIC year 5 through EDIC year 13/14, a 30%
reduction in the risk of developing confirmed DSPN by EDIC year 13/14
with prior intensive glucose control persisted (Martin et al., 2014; Pop-
Busui et al., 2010). Similar trends were observed on several NCS measures
(Albers et al., 2010; Martin et al., 2014; Pop-Busui et al., 2010). After
adjusting for NCS results at DCCT closeout, the risk reduction attenuated
to 17% (not significant), suggesting that subclinical DSPN (as documented
18 R. Pop-Busui and C. Martin
5. DISCUSSION
The DCCT/EDIC has furthered the understanding of the role of glu-
cose control on development and progression of DSPN and CAN (Martin
et al., 2014; Pop-Busui et al., 2010). Differences in the incidence and prev-
alence of both DSPN and CAN reflect differences in glucose control, favor-
ing HbA1c levels that are closer to nondiabetic levels.
Confirmed clinical neuropathy increased in both intensive and conven-
tional participants by EDIC year 13–14 but treatment-group differences
were eliminated by adjusting for continuous nerve conduction variables
at the end of the DCCT (Martin et al., 2014). Among subjects without con-
firmed clinical neuropathy, former conventional subjects had worse (less
normal) electrophysiologic results at the end of the DCCT (Albers et al.,
2010, 2007; Martin et al., 2014). These subjects were already closer to
the “tipping point” for meeting confirmed clinical neuropathy criteria by
DCCT-end and this could partially explain the findings of continued differ-
ence in the development of DSPN in the intensive and conventional groups
during EDIC (Albers et al., 2010, 2007; Martin et al., 2014). Whether an
additional influence of early intensive glucose control might have been
apparent earlier during EDIC is unknown, but a persistent metabolic effect
was not required to explain the durable beneficial effects on confirmed
DSPN at EDIC year 13–14 (Albers et al., 2010, 2007; Martin et al., 2014).
In contrast, the durable benefit of intensive glucose control first observed
for retinopathy and nephropathy was also observed for CAN at EDIC year
20 R. Pop-Busui and C. Martin
13–14 and modeling that adjusted for R-R variation at DCCT closeout did
not negate the INT-associated risk reduction for development of CAN
(Martin et al., 2014; Pop-Busui et al., 2009). These findings parallel the
reported long-term benefits of prior intensive glycemic control on retinop-
athy, nephropathy (DCCT, 2002; The Diabetes Control and Complications
Trial/Epidemiology of Diabetes Interventions and Complications Research
Group, 2000; Writing Team for DCCT/EDIC, 2003), and cardiovascular
disease (Nathan et al., 2005), and are consistent with the phenomenon ter-
med “metabolic memory” (Martin et al., 2014; Pop-Busui et al., 2009;
Writing Team for DCCT/EDIC, 2003). This discordance of the impact
of DCCT treatment-group effect on longer term outcomes for DSPN
and CAN may reflect differences in susceptibility of small and large nerve
fibers to glycemic exposure (Martin et al., 2014; Pop-Busui et al., 2010).
Although both DCCT and EDIC confirmed that glycemic control is a
significant and robust predictor of neuropathy in T1D, they also show that
for most patients with T1D, current strategies for optimizing glucose control
are insufficient to fully prevent or delay the development of neuropathic
complications, as 25% of subjects in the former intensive treatment group
and 35% of subjects in the former conventional treatment group had con-
firmed DSPN at EDIC year 13–14.
Genitourinary problems associated with diabetes, with the likely excep-
tion of ED, are frequently overlooked in clinical practice and are rarely con-
sidered in the context of diabetic neuropathies. Including evaluations for the
urologic complications both in men and women as part of the UroEDIC
study thus afforded an opportunity to define the extent of genitourinary
complications of diabetes and to explore any relationships to well-defined
complications, including neuropathy (Pop-Busui et al., 2015). Our recent
findings reporting strong association between CAN, ED, and LUTS in a
large cohort of more than 600 male DCCT/EDIC participants with
T1DM demonstrate a link between CAN and more generalized autonomic
neuropathy and also suggest that CAN may predict the development of ED
and LUTS in men with long-standing T1DM (Pop-Busui et al., 2015).
Although prior experimental evidence suggested that peripheral and auto-
nomic dysfunction may be an important factor in the etiology of ED and
possibly LUTS, these findings are the first to systematically demonstrate a
link between CAN and DSPN, and ED/LUTS in a large cohort of well-
characterized men with T1DM, owing perhaps to shared or overlapping
mechanisms of neuronal damage (Pop-Busui et al., 2015).
Neuropathy in the DCCT/EDIC 21
Lastly, at the time the DCCT was conducted, the limitations of exoge-
nous insulin treatment, namely the significantly greater risk of hypoglycemia
associated with INT treatment during DCCT, resulted in overall levels of
glucose control that were still not optimal. It may be that, with the use of
continuous glucose monitoring and emerging closed-loop insulin delivery
systems, normoglycemia without hypoglycemia may be reached. This
would indeed offer the opportunity to not only improve our understanding
of the impact of glycemia on neurologic outcomes, but to also examine the
effects of other risk factors including lipids and obesity.
In summary, the DSPN and CAN measures selected for the DCCT and
EDIC represent a carefully considered balance between the aims of the
study, and test validity, reproducibility, tolerability, safety, availability,
and cost. The DCCT/EDIC participants have been unbelievably generous
in their commitment to these studies, now with over 30 years of follow-up.
The EDIC study continues to evaluate the incidence, long-term outcomes,
and risk factors for neuropathic complications in a large number of well-
characterized patients with T1D and continues to demonstrate the value
of optimizing glucose control as early as possible in the course of the disease
to ameliorate the long-term effects of hyperglycemia.
REFERENCES
Albers, J. W., Herman, W. H., Pop-Busui, R., Feldman, E. L., Martin, C. L., Cleary, P. A.,
et al. (2010). Effect of prior intensive insulin treatment during the Diabetes Control and
Complications Trial (DCCT) on peripheral neuropathy in type 1 diabetes during the
Epidemiology of Diabetes Interventions and Complications (EDIC) Study. Diabetes
Care, 33, 1090–1096.
Albers, J. W., Herman, W. H., Pop-Busui, R., Martin, C. L., Cleary, P., & Waberski, B.
(2007). Subclinical neuropathy among diabetes control and complications trial partici-
pants without diagnosable neuropathy at trial completion: Possible predictors of incident
neuropathy? Diabetes Care, 30, 2613–2618.
American Diabetes Association. (2015). Standards of medical care in diabetes-2015: Sum-
mary of revisions. Diabetes Care, 38(Suppl.), S4.
Ang, L., Jaiswal, M., Martin, C., & Pop-Busui, R. (2014). Glucose control and diabetic neu-
ropathy: Lessons from recent large clinical trials. Current Diabetes Reports, 14, 528.
Bharucha, A. E., Batey-Schaefer, B., Cleary, P. A., Murray, J. A., Cowie, C., Lorenzi, G.,
et al. (2015). Delayed gastric emptying is associated with early and long-term hypergly-
cemia in type 1 diabetes mellitus. Gastroenterology, 149, 330–339.
Boulton, A. J., Malik, R. A., Arezzo, J. C., & Sosenko, J. M. (2004). Diabetic somatic neu-
ropathies. Diabetes Care, 27, 1458–1486.
Boulton, A. J., Vinik, A. I., Arezzo, J. C., Bril, V., Feldman, E. L., Freeman, R., et al. (2005).
Diabetic neuropathies: A statement by the American diabetes association. Diabetes Care,
28, 956–962.
DCCT. (1993). The effect of intensive treatment of diabetes on the development
and progression of long-term complications in insulin-dependent diabetes mellitus.
24 R. Pop-Busui and C. Martin
The Diabetes Control and Complications Trial Research Group. The New England
Journal of Medicine, 329, 977–986.
DCCT. (1995). Effect of intensive diabetes treatment on nerve conduction in the Diabetes
Control and Complications Trial. Annals of Neurology, 38, 869–880.
DCCT. (1998). The effect of intensive diabetes therapy on measures of autonomic nervous
system function in the Diabetes Control and Complications Trial (DCCT). Diabetologia,
41, 416–423.
DCCT. (2002). Effect of intensive therapy on the microvascular complications of type 1 dia-
betes mellitus. JAMA, 287, 2563–2569.
Epidemiology of Diabetes Interventions and Complications (EDIC). (1999). Design, imple-
mentation, and preliminary results of a long-term follow-up of the Diabetes Control and
Complications Trial cohort. Diabetes Care, 22, 99–111.
Feldman, E. L., Stevens, M. J., Thomas, P. K., Brown, M. B., Canal, N., & Greene, D. A.
(1994). A practical two-step quantitative clinical and electrophysiological assessment for
the diagnosis and staging of diabetic neuropathy. Diabetes Care, 17, 1281–1289.
Herman, W. H., Pop-Busui, R., Braffett, B. H., Martin, C. L., Cleary, P. A., Albers, J. W.,
et al. (2012). Use of the Michigan Neuropathy Screening Instrument as a measure of
distal symmetrical peripheral neuropathy in type 1 diabetes: Results from the Diabetes
Control and Complications Trial/Epidemiology of Diabetes Interventions and Compli-
cations. Diabetic Medicine, 29, 937–944.
Hotaling, J., Sarma, A. V., Pop-Busui, R., Cleary, P., Dunn, R. L., & Wessels, H. (2013).
Autonomic neuropathy, sexual dysfunction and urinary incontinence in women with
type 1 diabetes: Findings from the DCCT/EDIC. Diabetologia, 56, S1–S566.
Low, P. A., Benrud-Larson, L. M., Sletten, D. M., Opfer-Gehrking, T. L., Weigand, S. D.,
O’Brien, P. C., et al. (2004). Autonomic symptoms and diabetic neuropathy:
A population-based study. Diabetes Care, 27, 2942–2947.
Low, P. A., Denq, J. C., Opfer-Gehrking, T. L., Dyck, P. J., O’Brien, P. C., & Slezak, J. M.
(1997). Effect of age and gender on sudomotor and cardiovagal function and blood pres-
sure response to tilt in normal subjects. Muscle & Nerve, 20, 1561–1568.
Malik, R., Veves, A., Tesfaye, S., Smith, G., Cameron, N., Zochodne, D., et al. (2011).
Small fiber neuropathy: Role in the diagnosis of diabetic sensorimotor polyneuropathy.
Diabetes/Metabolism Research and Reviews, 27, 678–684.
Martin, C. L., Albers, J., Herman, W. H., Cleary, P., Waberski, B., Greene, D. A., et al.
(2006). Neuropathy among the diabetes control and complications trial cohort 8 years
after trial completion. Diabetes Care, 29, 340–344.
Martin, C. L., Albers, J. W., & Pop-Busui, R. (2014). Neuropathy and related findings in the
diabetes control and complications trial/epidemiology of diabetes interventions and
complications study. Diabetes Care, 37, 31–38.
Martin, C. L., Waberski, B. H., Pop-Busui, R., Cleary, P. A., Catton, S., Albers, J. W., et al.
(2010). Vibration perception threshold as a measure of distal symmetrical peripheral neu-
ropathy in type 1 diabetes: Results from the DCCT/EDIC study. Diabetes Care, 33,
2635–2641.
Nathan, D. M., Cleary, P. A., Backlund, J. Y., Genuth, S. M., Lachin, J. M., Orchard, T. J.,
et al. (2005). Intensive diabetes treatment and cardiovascular disease in patients with type
1 diabetes. The New England Journal of Medicine, 353, 2643–2653.
Pop-Busui, R., Braffett, B. H., Hotaling, J., Cleary, P., Martin, C. L., Dunn, R. L., et al.
(2014). Diabetic neuropathy and urologic complications in men with type 1 diabetes
in the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interven-
tion and Complications Study (DCCT/EDIC). Diabetes, 63, A149–A580.
Pop-Busui, R., Herman, W. H., Feldman, E. L., Low, P. A., Martin, C. L., Cleary, P. A.,
et al. (2010). DCCT and EDIC studies in type 1 diabetes: Lessons for diabetic neuropathy
regarding metabolic memory and natural history. Current Diabetes Reports, 10, 276–282.
Neuropathy in the DCCT/EDIC 25
Pop-Busui, R., Hotaling, J., Braffett, B. H., Cleary, P. A., Dunn, R. L., Martin, C. L., et al.
(2015). Cardiovascular autonomic neuropathy, erectile dysfunction and lower urinary
tract symptoms in men with type 1 diabetes: Findings from the DCCT/EDIC. The
Journal of Urology, 193, 2045–2051.
Pop-Busui, R., Low, P. A., Waberski, B. H., Martin, C. L., Albers, J. W., Feldman, E. L.,
et al. (2009). Effects of prior intensive insulin therapy on cardiac autonomic nervous sys-
tem function in type 1 diabetes mellitus: The Diabetes Control and Complications Trial/
Epidemiology of Diabetes Interventions and Complications study (DCCT/EDIC).
Circulation, 119, 2886–2893.
Sarma, A. V., Kanaya, A. M., Nyberg, L. M., Kusek, J. W., Vittinghoff, E., Rutledge, B.,
et al. (2009). Urinary incontinence among women with type 1 diabetes—How common
is it? The Journal of Urology, 181, 1224–1230. discussion 1230.
Smith, A. G., & Singleton, J. R. (2012). Diabetic neuropathy. Continuum (Minneapolis, Minn),
18, 60–84.
Tesfaye, S., Boulton, A. J., Dyck, P. J., Freeman, R., Horowitz, M., Kempler, P., et al.
(2010). Diabetic neuropathies: Update on definitions, diagnostic criteria, estimation of
severity, and treatments. Diabetes Care, 33, 2285–2293.
The Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and
Complications Research Group. (2000). Retinopathy and nephropathy in patients with
type 1 diabetes four years after a trial of intensive therapy. The New England Journal of
Medicine, 342, 381–389.
Vileikyte, L., Peyrot, M., Bundy, C., Rubin, R. R., Leventhal, H., Mora, P., et al. (2003).
The development and validation of a neuropathy- and foot ulcer-specific quality of life
instrument. Diabetes Care, 26, 2549–2555.
Wessells, H., Penson, D. F., Cleary, P., Rutledge, B. N., Lachin, J. M., McVary, K. T., et al.
(2011). Effect of intensive glycemic therapy on erectile function in men with type 1 dia-
betes. The Journal of Urology, 185, 1828–1834.
Writing Team for DCCT/EDIC. (2003). Sustained effect of intensive treatment of type 1
diabetes mellitus on development and progression of diabetic nephropathy: The Epide-
miology of Diabetes Interventions and Complications (EDIC) study. JAMA, 290,
2159–2167.
CHAPTER THREE
Contents
1. Introduction 27
2. Study Aims: Prevent DSP, Prevent Progression, or Reverse DSP? 28
3. Selection of Study Participants 29
4. Trial Duration 31
5. Study End Points 33
6. Core Labs and Training 35
7. Intervention 35
8. Study Conduct 36
9. Streamlined Ethics and Contracts Process 37
10. Summary 37
References 38
Abstract
Multiple phase III clinical trials have failed to show disease-modifying benefits for dia-
betic sensorimotor polyneuropathy (DSP) and this may be due to the design of the clin-
ical trials. The perfect clinical trial in DSP would enroll sufficiently large numbers of
patients having early or minimal disease, as demonstrated by nerve conduction studies
(NCS). These patients would be treated with an intervention given at an effective and
well-tolerated dose for a sufficient duration of time to show change in the end points
selected. For objective or surrogate measures such as NCS and for some small fiber mea-
sures, the duration needed to show positive change may be as brief as 6–12 months, but
subsequently, trials lasting 5–8 years will be required to demonstrate clinical benefits.
1. INTRODUCTION
Multiple phase III clinical trials have lacked success in identifying
disease-modifying therapy for diabetic sensorimotor polyneuropathy
(DSP). Although numerous pharmacological agents such as neurotrophic fac-
tors, aldose reductase inhibitors, protein kinase C inhibitors, and antioxidant
Table 1 Pharmacological Agents Tested in Phase III Trials for Disease Modification in
Diabetic Sensorimotor Polyneuropathy
symptoms, physical signs, or both, helps satisfy regulatory and diabetes com-
munity preferences for investigation of disease that is considered to be relevant
to patients. Simple clinical measures, such as the TCNS or mTCNS (Bril &
Perkins, 2002; Bril, Tomioka, et al., 2009) or the Michigan Neuropathy Score
(Herman et al., 2012) can provide a summary method of compiling the clinical
features of disease and help identify the presence of neuropathy reliably
(Zilliox, Ruby, Singh, Zhan, & Russell, 2015). Assessment of progression
may be improved with scales that are more granular and likely more sensitive
such as the mTCNS (Bril, Tomioka, et al., 2009).
Objective criteria, such as nerve conduction studies (NCS), have higher
reliability and are preferable in clinical trials (Bril et al., 1998; Dyck et al.,
2010). As progressive nerve fiber loss is the hallmark of DSP, loss of the
lower-limb sensory nerve responses shows advanced disease and selecting
patients who have a recordable sural sensory nerve response ensures that they
will have less severe neuropathy and may respond to interventions (Vinik
et al., 2005). So, all patients in clinical trials should have NCS to help select
patients and to show response to therapy when large fiber function is
expected to improve. However, DSP may be too far advanced even with
NCS in the normal range, as the healthy values for NCS parameters in
any given patient are unknown. The reference range is broad and the results
in any given patient may have declined significantly but remain in the lower
range of normal. Given unlimited resources, one might suggest that every-
one in the population have baseline NCS, so that when diabetes is diagnosed
and the NCS are repeated, one could determine the magnitude of changes
from baseline and then document the presence and severity of DSP at
an earlier stage. Naturally, a cohort of healthy volunteers would need to
have matched NCS at time points similar to those in diabetes patients to
determine the natural history of NCS in a broad population and show
the differences in diabetes subjects. Given that this approach is unrealistic
to achieve, additional means of accurately identifying disease presence
and status are required.
Identification of small fiber impairments are thought to precede
large nerve fiber loss and can help select patients for clinical trials. These
methods include evaluation of intraepidermal nerve fiber density (IENFD)
(Lauria et al., 1999), quantitative thermal thresholds (QTT) (Arezzo,
Schaumburg, & Laudadio, 1986; Yarnitsky, 1997), laser Doppler flare area
(Krishnan & Rayman, 2004), and corneal confocal nerve fiber microscopy
(CCM) (Ahmed et al., 2012; Malik et al., 2003). Tests of sudomotor
function can help supplement these studies (Casellini, Parson, Richardson,
The Perfect Clinical Trial 31
4. TRIAL DURATION
The most successful clinical trial on neuropathy in diabetes to date was
the DCCT (The effect of intensive diabetes therapy on the development and
progression of neuropathy. The Diabetes Control and Complications Trial
Research Group, 1995). In this study of 1441 subjects with Type 1 diabetes,
intensive glycemic control for 5 years reduced the prevalence of neuropathy
as measured by clinical, electrophysiological, and cardiac autonomic tests, by
60% (The effect of intensive diabetes therapy on the development and
progression of neuropathy. The Diabetes Control and Complications
Trial Research Group, 1995). This study showed that the end points of
clinical evaluation, NCS, and autonomic system testing could reflect
changes in DSP and are thus valid for use in clinical trials. Also, this costly
study was funded by US governmental agencies, was planned for a duration
of 7 years, and had a very large number of subjects who were evaluated com-
prehensively with clinical examination by neurologists, autonomic system
testing, and also NCS. The DCCT showed at 5 years the benefits on
DSP of intensive compared to conventional glycemic control in Type 1
patients, so the minimum duration of any study in Type 1 patients should
32 V. Bril
be 5 years when this array of tests are the key clinical endpoints. Assessments
at intermediate time-points were not available and so shorter durations of
studies cannot be recommended. Large-scale epidemiological studies or
intervention studies without similar comprehensive evaluations of neurop-
athy did not provide strong evidence of benefits with respect to DSP, partly
because the neuropathy was not evaluated in a comprehensive manner or
because the separation of treatment groups of regular and improved control
was suboptimal (U. P. D. S. Group, 1998). Nonetheless, a separation of
vibration perception was noted at 8 years in the UKPDS study in Type 2
patients, even with these caveats. Reduction in autonomic neuropathy
has been observed with multifactorial interventions (Gaede, Vedel,
Parving, & Pedersen, 1999, Greene, Arezzo, Brown, & Zenarestat Study
Group, 1999). The experience with Type 2 patients may have differed if
different study protocols were employed. However, these studies suggest
that durations in this population need to be lengthy and perhaps as long
as 8 years or more.
The longest study funded by the pharmacology industry was a 4-year
treatment trial of an oral medication (the NATHAN study) that was not effec-
tive and did not show benefits in the main end points of the study (Ziegler et al.,
2011). The end points were similar to those in the DCCT. This outcome is
challenging to understand because evaluations in the DCCT were done at base-
line and at 5 years, but not at 4 years. We cannot know whether the reduction in
prevalence of DSP in the DCCT was linear over 5 years, or not. If the neurop-
athy assessments had been done at 4 years, then the outcome of the NATHAN
study could be compared directly to the DCCT results, but that is not the case.
Also, the study populations were different in that the NATHAN study had a
predominance of patients with Type 2 diabetes. When factors that might
have influenced the outcome were evaluated, those with a normal BMI,
normal blood pressure, and higher burden related to cardiovascular disease, dia-
betes, and neuropathy were more responsive to treatment (Ziegler, Low,
Freeman, Tritschler, & Vinik, 2016), suggesting that a multifactorial approach
when treating DSP will be required. Nonetheless, it is likely that the DCCT
would have shown some trend to less neuropathy in Type 1 diabetes subjects
at 4 years.
In summary, given the fact that DSP is a chronic, insidiously progressive
axonal neuropathy, and the results of the DCCT and UKPDS, the perfect
clinical trial would last at least 5 years in Type 1 patients and 8 years or more
in Type 2 patients.
The Perfect Clinical Trial 33
not as objective as might be thought (Perry & Bril, 1994) without standard-
ized training of those evaluating the biopsy (Kalichman, Chalk, & Mizisin,
1999), so that large nerve fiber biopsy is not advocated at this time.
End points should include evaluation of small nerve fibers as these can be
demonstrated to be affected before NCS falls into the abnormal range
(Breiner et al., 2014; Smith, Ramachandran, Tripp, & Singleton, 2001).
The use of IENFD and other measures of epidermal and dermal nerve fibers
(such as axonal swellings and varicosities) can provide direct structural evi-
dence of nerve status (Mellgren, Nolano, & Sommer, 2013). Nerve regen-
eration with the capsaicin IENFD system can be another way to assess nerve
recovery (Polydefkis et al., 2004). So, clinical trials should include IENFD at
baseline and then at end of treatment. Sampling from the distal lower leg and
distal thigh are suggested. The procedure is invasive, but minimally so with
minor potential side effects of infection and bleeding.
Noninvasive measures of small nerve fiber function are preferable to
invasive ones, but should also supplement the invasive measure. Corneal
confocal microscopy to directly visualize small terminal trigeminal nerve
fibers in Bowman’s layer of the cornea may be useful to assess nerve regen-
eration (Ahmed et al., 2012; Malik et al., 2003). Nerve fiber length on CCM
is a surrogate measure for the longest nerve fibers, correlates with longer
nerve fiber measures (IENFD, NCS, QST), and may show changes earlier
than would be apparent in longer fibers (Hertz et al., 2011; Sivaskandarajah
et al., 2013). This end point should be measured at baseline and then every 6
months during a study lasting for a minimum of 5 years (Mehra et al., 2007).
If no change in CCM parameters is observed at earlier time points, then the
study can be terminated earlier, but if changes are apparent, then the study
should be continued for 5 years to demonstrate corresponding changes in the
longest nerve fibers and provide a convincing study result.
Quantitative sensory threshold measurements provide a numerical eval-
uation of sensory function, with thermal thresholds (QTT) indicating small
nerve fiber function and VPTs reflecting large nerve fiber function. In QTT,
cold detection thresholds (CDT) are more reliable than warming thresholds,
and so are preferable (Zinman, Bril, & Perkins, 2004). These measures
should be done in clinical trials to help evaluate how sensory nerves are func-
tioning in those with DSP. There is great variation between assessments, but
VPT and CDT are valuable end points and should be done at study start and
end (Young, Every, & Boulton, 1993; Zinman et al., 2004).
Laser Doppler flare area imaging in response to heat-mediated vasodila-
tion can provide a measure of cutaneous nerve fiber function (Krishnan &
The Perfect Clinical Trial 35
Rayman, 2004). This end point may help provide supplementary evidence
of small nerve fiber function (Krishnan, Quattrini, Jeziorska, Malik, &
Rayman, 2009).
Autonomic function tests such as sudomotor function, heart rate vari-
ability, and others can provide separate evidence of small nerve fiber
responsiveness.
Generally, all end points at baseline and end of study should have triplicate
measures to improve the accuracy of the results. The median value is used so
that one unusual value does not skew the “real” value of the parameter.
7. INTERVENTION
Only therapies that are effective in appropriate preclinical models at
doses that can sensibly be used in humans, allowing for interspecies allome-
tric scaling (Freireich, Gehan, Rall, Schmidt, & Skipper, 1966; Huang &
Riviere, 2014) should be advanced to human trials. In many unsuccessful
clinical trials, a study drug has been tested and found effective in preclinical
and phase II studies and then failed in phase III. This has sometimes been
related to the use of doses in the phase III study that were lower than the
phase II and subsequently proved to be less effective, as discussed for trials
of NGF in the previous edition of this volume (Apfel, 2002). The use of
36 V. Bril
8. STUDY CONDUCT
For a 5-year or longer clinical trial, retention of study subjects
becomes a major concern. The DCCT was successful in large part due to
the motivated population under study and the initiatives put in place by
the study coordinators: morale and team building items and activities that
engaged study participants proved key to the success of this study. For
any long duration study, such activities are necessary and should be allowed
despite the current ethical climate that focuses on perceived “coercion” of
study subjects. It is more unethical to run a study that cannot be completed
with the requisite number of subjects without constant positive reinforce-
ment and encouragement by the study staff. Small study perks (mugs,
t-shirts, baseball caps, and other items) might convince study participants
to stay the course. Adequate remuneration for study subjects’ time, coverage
of all patient study-related costs, and proper funding for sufficient numbers
of study staff so that they can spend the time necessary for subject retention
are essential. The ability to enroll subjects who live far from a study center
should be enabled by proper budgeting of participant expenses related to the
study. Newer communication tools such as videoconferencing with each
study participant using personal electronic devices might help with long-
term retention. All of the items identified in this section are part of a study
done with unlimited resources, but need to be stated explicitly to ensure that
The Perfect Clinical Trial 37
they are funded in the current research climate where all costs tend to be
minimized as much as possible, if not eliminated entirely, to the detriment
of any study.
10. SUMMARY
The perfect clinical trial in DSP would enroll sufficiently large num-
bers of patients having early or minimal disease, as demonstrated by NCS.
These patients would be treated with an intervention at an effective and
well-tolerated dose for a sufficient duration of time to show change in
the end points selected. For objective or surrogate measures such as NCS
and for some small fiber measure, this duration can be as brief as 6–12
months, but eventually longer duration trials of 5–8 years will be required
to show clinical improvement.
38 V. Bril
REFERENCES
Ahmed, A., Bril, V., Orszag, A., Paulson, J., Yeung, E., Ngo, M., et al. (2012). Detection of
diabetic sensorimotor polyneuropathy by corneal confocal microscopy in type 1 diabetes:
A concurrent validity study. Diabetes Care, 35(4), 821–828. http://doi.org/10.2337/
dc11-1396.
Apfel, S. C. (2002). Nerve growth factor for the treatment of diabetic neuropathy: What
went wrong, what went right, and what does the future hold? International Review of Neu-
robiology, 50, 393–413.
Arezzo, J. C., Schaumburg, H. H., & Laudadio, C. (1986). Thermal sensitivity tester. Device
for quantitative assessment of thermal sense in diabetic neuropathy. Diabetes, 35(5),
590–592.
Breiner, A., Lovblom, L. E., Perkins, B. A., & Bril, V. (2014). Does the prevailing hypothesis
that small-fiber dysfunction precedes large-fiber dysfunction apply to type 1 diabetic
patients? Diabetes Care, 37(5), 1418–1424. http://doi.org/10.2337/dc13-2005.
Bril, V., Ellison, R., Ngo, M., Bergstrom, B., Raynard, D., & Gin, H. (1998). Electrophys-
iological monitoring in clinical trials. Roche Neuropathy Study Group. Muscle & Nerve,
21(11), 1368–1373.
Bril, V., Katzberg, H., Donofrio, P., Banach, M., Dalakas, M. C., Deng, C., et al. (2009).
Electrophysiology in chronic inflammatory demyelinating polyneuropathy with IGIV.
Muscle & Nerve, 39(4), 448–455. http://doi.org/10.1002/mus.21236.
Bril, V., & Perkins, B. A. (2002). Validation of the Toronto Clinical Scoring System for dia-
betic polyneuropathy. Diabetes Care, 25(11), 2048–2052.
Bril, V., Tomioka, S., Buchanan, R. A., Perkins, B. A., & mTCNS Study Group (2009).
Reliability and validity of the modified Toronto Clinical Neuropathy Score in diabetic
sensorimotor polyneuropathy. Diabetic Medicine: A Journal of the British Diabetic Association,
26(3), 240–246. http://doi.org/10.1111/j.1464-5491.2009.02667.x.
Casellini, C. M., Parson, H. K., Richardson, M. S., Nevoret, M. L., & Vinik, A. I. (2013).
Sudoscan, a noninvasive tool for detecting diabetic small fiber neuropathy and auto-
nomic dysfunction. Diabetes Technology & Therapeutics, 15(11), 948–953. http://doi.
org/10.1089/dia.2013.0129.
D. C. A. C. T. D. R. Group (1995). Effect of intensive diabetes treatment on nerve conduc-
tion in the diabetes control and complications trial. Annals of Neurology, 38(6), 869–880.
Dunnigan, S. K., Ebadi, H., Breiner, A., Katzberg, H. D., Lovblom, L. E., Perkins, B. A.,
et al. (2013). Conduction slowing in diabetic sensorimotor polyneuropathy. Diabetes
Care, 36(11), 3684–3690. http://doi.org/10.2337/dc13-0746.
Dyck, P. J., Overland, C. J., Low, P. A., Litchy, W. J., Davies, J. L., Dyck, P. J. B., et al.
(2010). Signs and symptoms versus nerve conduction studies to diagnose diabetic senso-
rimotor polyneuropathy: Cl vs. NPhys trial. Muscle & Nerve, 42(2), 157–164. http://doi.
org/10.1002/mus.21661.
Freireich, E. J., Gehan, E. A., Rall, D. P., Schmidt, L. H., & Skipper, H. E. (1966). Quan-
titative comparison of toxicity of anticancer agents in mouse, rat, hamster, dog, monkey,
and man. Cancer Chemotherapy Reports Part 1, 50(4), 219–244.
Gaede, P., Vedel, P., Parving, H. H., & Pedersen, O. (1999). Intensified multifactorial inter-
vention in patients with type 2 diabetes mellitus and microalbuminuria: The Steno type 2
randomised study. Lancet, 353(9153), 617–622. http://doi.org/10.1016/S0140-6736(
98)07368-1.
Gibbons, C. H., Freeman, R., Tecilazich, F., Dinh, T., Lyons, T. E., Gnardellis, C., et al.
(2013). The evolving natural history of neurophysiologic function in patients with
well-controlled diabetes. Journal of the Peripheral Nervous System: JPNS, 18(2),
153–161. http://doi.org/10.1111/jns5.12021.
The Perfect Clinical Trial 39
Greene, D. A., Arezzo, J. C., Brown, M. B., & Zenarestat Study Group (1999). Effect of
aldose reductase inhibition on nerve conduction and morphometry in diabetic neurop-
athy. Zenarestat Study Group. Neurology, 53(3), 580–591.
Herman, W. H., Pop-Busui, R., Braffett, B. H., Martin, C. L., Cleary, P. A., Albers, J. W.,
et al. (2012). Use of the Michigan neuropathy screening instrument as a measure of distal
symmetrical peripheral neuropathy in type 1 diabetes: Results from the diabetes control
and complications trial/epidemiology of diabetes interventions and complications. Dia-
betic Medicine: A Journal of the British Diabetic Association, 29(7), 937–944. http://doi.org/
10.1111/j.1464-5491.2012.03644.x.
Hertz, P., Bril, V., Orszag, A., Ahmed, A., Ng, E., Nwe, P., et al. (2011). Reproducibility of
in vivo corneal confocal microscopy as a novel screening test for early diabetic sensori-
motor polyneuropathy. Diabetic Medicine: A Journal of the British Diabetic Association,
28(10), 1253–1260. http://doi.org/10.1111/j.1464-5491.2011.03299.x.
Hotta, N., Kawamori, R., Fukuda, M., Shigeta, Y., & Aldose Reductase Inhibitor-Diabetes
Complications Trial Study Group (2012). Long-term clinical effects of epalrestat, an
aldose reductase inhibitor, on progression of diabetic neuropathy and other microvascu-
lar complications: Multivariate epidemiological analysis based on patient background fac-
tors and severity of diabetic neuropathy. Diabetic Medicine: A Journal of the British Diabetic
Association, 29(12), 1529–1533. http://doi.org/10.1111/j.1464-5491.2012.03684.x.
Huang, Q., & Riviere, J. E. (2014). The application of allometric scaling principles to predict
pharmacokinetic parameters across species. Expert Opinion on Drug Metabolism & Toxicol-
ogy, 10(9), 1241–1253. http://doi.org/10.1517/17425255.2014.934671.
Kalichman, M. W., Chalk, C. H., & Mizisin, A. P. (1999). Classification of teased nerve fibers
for multicenter clinical trials. Journal of the Peripheral Nervous System: JPNS, 4(3–4),
233–244.
Krishnan, S. T. M., Quattrini, C., Jeziorska, M., Malik, R. A., & Rayman, G. (2009). Abnor-
mal LDIflare but normal quantitative sensory testing and dermal nerve fiber density in
patients with painful diabetic neuropathy. Diabetes Care, 32(3), 451–455. http://doi.
org/10.2337/dc08-1453.
Krishnan, S. T. M., & Rayman, G. (2004). The LDIflare: A novel test of C-fiber function
demonstrates early neuropathy in type 2 diabetes. Diabetes Care, 27(12), 2930–2935.
Lauria, G., Holland, N., Hauer, P., Cornblath, D. R., Griffin, J. W., & McArthur, J. C.
(1999). Epidermal innervation: Changes with aging, topographic location, and in sensory
neuropathy. Journal of the Neurological Sciences, 164(2), 172–178.
Malik, R. A., Kallinikos, P., Abbott, C. A., van Schie, C. H. M., Morgan, P., Efron, N., et al.
(2003). Corneal confocal microscopy: A non-invasive surrogate of nerve fibre damage
and repair in diabetic patients. . Diabetologia, 46(5), 683–688. http://doi.org/10.1007/
s00125-003-1086-8.
Mehra, S., Tavakoli, M., Kallinikos, P. A., Efron, N., Boulton, A. J. M., Augustine, T., et al.
(2007). Corneal confocal microscopy detects early nerve regeneration after pancreas
transplantation in patients with type 1 diabetes. Diabetes Care, 30(10), 2608–2612.
http://doi.org/10.2337/dc07-0870.
Mellgren, S. I., Nolano, M., & Sommer, C. (2013). The cutaneous nerve biopsy: Technical
aspects, indications, and contribution. Handbook of Clinical Neurology, 115, 171–188.
http://doi.org/10.1016/B978-0-444-52902-2.00010-2.
Partanen, J., Niskanen, L., Lehtinen, J., Mervaala, E., Siitonen, O., & Uusitupa, M. (1995).
Natural history of peripheral neuropathy in patients with non-insulin-dependent diabe-
tes mellitus. The New England Journal of Medicine, 333(2), 89–94. http://doi.org/10.
1056/NEJM199507133330203.
Perkins, B. A., Dholasania, A., Buchanan, R. A., & Bril, V. (2010). Short-term metabolic
change is associated with improvement in measures of diabetic neuropathy: A 1-year
40 V. Bril
placebo cohort analysis. Diabetic Medicine: A Journal of the British Diabetic Association,
27(11), 1271–1279. http://doi.org/10.1111/j.1464-5491.2010.03110.x.
Perkins, B. A., Olaleye, D., Zinman, B., & Bril, V. (2001). Simple screening tests for periph-
eral neuropathy in the diabetes clinic. Diabetes Care, 24(2), 250–256.
Perry, J. R., & Bril, V. (1994). Complications of sural nerve biopsy in diabetic versus non-
diabetic patients. The Canadian Journal of Neurological Sciences Le Journal Canadien Des Sci-
ences Neurologiques, 21(1), 34–37.
Pirart, J. (1978). Diabetes mellitus and its degenerative complications: A prospective study of
4,400 patients observed between 1947 and 1973. Diabetes Care, 1(168–188), 252–263.
Polydefkis, M., Hauer, P., Sheth, S., Sirdofsky, M., Griffin, J. W., & McArthur, J. C. (2004).
The time course of epidermal nerve fibre regeneration: Studies in normal controls and in
people with diabetes, with and without neuropathy. Brain: A Journal of Neurology, 127(Pt.
7), 1606–1615. http://doi.org/10.1093/brain/awh175.
Singleton, J. R., Smith, A. G., & Bromberg, M. B. (2001). Increased prevalence of impaired
glucose tolerance in patients with painful sensory neuropathy. Diabetes Care, 24(8),
1448–1453.
Sivaskandarajah, G. A., Halpern, E. M., Lovblom, L. E., Weisman, A., Orlov, S., Bril, V.,
et al. (2013). Structure-function relationship between corneal nerves and conventional
small-fiber tests in type 1 diabetes. Diabetes Care, 36(9), 2748–2755. http://doi.org/10.
2337/dc12-2075.
Smith, A. G., Ramachandran, P., Tripp, S., & Singleton, J. R. (2001). Epidermal nerve
innervation in impaired glucose tolerance and diabetes-associated neuropathy.
Neurology, 57, 1701–1704.
Sumner, C. J., Sheth, S., Griffin, J. W., Cornblath, D. R., & Polydefkis, M. (2003). The
spectrum of neuropathy in diabetes and impaired glucose tolerance. Neurology, 60(1),
108–111.
Tesfaye, S., Chaturvedi, N., Eaton, S. E. M., Ward, J. D., Manes, C., Ionescu-Tirgoviste, C.,
et al. (2005). Vascular risk factors and diabetic neuropathy. The New England Journal of
Medicine, 352(4), 341–350. http://doi.org/10.1056/NEJMoa032782.
Tesfaye, S., Tandan, R., Bastyr, E. J., Kles, K. A., Skljarevski, V., Price, K. L., et al. (2007).
Factors that impact symptomatic diabetic peripheral neuropathy in placebo-administered
patients from two 1-year clinical trials. Diabetes Care, 30(10), 2626–2632. http://doi.
org/10.2337/dc07-0608.
The effect of intensive diabetes therapy on the development and progression of neuropathy.
The Diabetes Control and Complications Trial Research Group. (1995). Annals of Inter-
nal Medicine, 122(8), 561–568.
U. P. D. S. Group (1998). Tight blood pressure control and risk of macrovascular and micro-
vascular complications in type 2 diabetes: UKPDS 38. UK Prospective Diabetes Study
Group. BMJ, 317(7160), 703–713.
Vinik, A. I., Bril, V., Litchy, W. J., Price, K. L., Bastyr, E. J., & MBBQ Study Group (2005).
Sural sensory action potential identifies diabetic peripheral neuropathy responders to
therapy. Muscle & Nerve, 32(5), 619–625. http://doi.org/10.1002/mus.20423.
Yarnitsky, D. (1997). Quantitative sensory testing. Muscle & Nerve, 20(2), 198–204.
Young, M. J., Every, N., & Boulton, A. J. (1993). A comparison of the neurothesiometer and
biothesiometer for measuring vibration perception in diabetic patients. Diabetes Research
and Clinical Practice, 20(2), 129–131.
Ziegler, D., Low, P. A., Freeman, R., Tritschler, H., & Vinik, A. I. (2016). Predictors of
improvement and progression of diabetic polyneuropathy following treatment with
α-lipoic acid for 4 years in the NATHAN 1 trial. Journal of Diabetes and its Complications,
30(2), 350–356. http://dx.doi.org/10.1016/j.jdiacomp.2015.10.018.
Ziegler, D., Low, P. A., Litchy, W. J., Boulton, A. J. M., Vinik, A. I., Freeman, R., et al.
(2011). Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in
The Perfect Clinical Trial 41
Contents
1. Why Use Animal Models? 45
2. What Species? 46
3. What Diabetogenic Insult? 48
4. STZ Toxicity 49
5. Novel Models 50
References 50
#
International Review of Neurobiology, Volume 127 2016 Elsevier Inc. 45
ISSN 0074-7742 All rights reserved.
http://dx.doi.org/10.1016/bs.irn.2016.03.011
46 N.A. Calcutt and P. Fernyhough
which are their short life span (and thus exposure time to diabetes) and short
hairy legs (and thus short distance from cell body to nerve terminal).
Whether there are interspecies differences in physiological and cellular
responses to diabetes that invalidate the common animal models is more
debatable. Support for the pertinence of animal models of diabetic neurop-
athy comes from recognition that diabetic animals develop many disorders in
common with diabetic patients, including nerve conduction slowing, resis-
tance to ischemic conduction block, allodynia, loss of thermal sensation, and
reduced small sensory fiber density in the skin and cornea (Biessels et al.,
2014). Additional practical benefits of using animal models include the abil-
ity to test hypotheses by methods not permissible or practical in humans, the
lower cost of preclinical research, and the accelerated temporal progression
of neuropathy that allows longitudinal studies over months rather than years
or decades. Unfortunately, as with many model systems, the literature is
cluttered with the inappropriate use of specialized models or overly exuber-
ant interpretation of positive findings. The fidelity of animal models of
diabetic neuropathy remains an important issue that requires frequent con-
sideration, revision, and refinement.
2. WHAT SPECIES?
Diabetes is not an exclusively human disease. There are reports of
spontaneous diabetes in many long-lived species that live in close contact
with humans, including nonhuman primates (Kuhar, Fuller, & Dennis,
2013), elephants (van der Kolk et al., 2011), camelids (Cebra, 2009), dogs
(Fleeman, Rand, & Morton, 2009), and cats (O’Brien, 2002). In many cases,
the provision of unnatural diets to wild animals kept in captivity and to spe-
cies domesticated for farming or companionship is suspected as causing dia-
betes. Indeed, the rise of type 2 diabetes in the human population is being
paralleled by a similar epidemic in the domestic cat population, which is also
exposed to poor diet and limited exercise. Diabetic cats are particularly
prone to developing a peripheral neuropathy that is strikingly similar to
the human condition with nerve conduction slowing accompanied by struc-
tural damage to blood vessels, Schwann cells, and axons (Estrella et al., 2008;
Mizisin et al., 2007). These observations support use of the diabetic cat pop-
ulation as a bridge between traditional preclinical drug efficacy studies and
clinical trials so that drug efficacy can be evaluated in nerves exhibiting the
full spectrum of structural pathology. Recruitment of diabetic cats and
screening for neuropathy by veterinarians is as viable as similar recruitment
Animal Models of Diabetic Neuropathy 47
exercises for clinical trials but, in our experience, also requires the same
attention to recruitment practices such as investigator training and incentives
for both investigator and the subject (or their owner). Given the huge cost of
clinical trials, efficacy testing in diabetic cats may be useful as a cost-effective
final preclinical evaluation point in the drug development pipeline.
Some species show normal physiological features that would indicate
diabetes in humans. For example, plasma glucose levels of birds, while vary-
ing between species, are much higher (10–40 mM range) than in most mam-
mals. This is not a permanent disease condition so much as a reflection of
different physiological goals, with glucagon, not insulin, being the dominant
pancreatic hormone in chickens (Braun & Sweazea, 2008). While hypergly-
cemia and relative insulin deficiency are the hallmarks of type 1 diabetes and
implicated in most current pathogenic hypotheses, they do not appear to
adversely impact the nervous system of birds. It has been argued that such
interspecies variations may prove useful for studying how organs resist dam-
age by hyperglycemia and/or insulinopenia (Singer, 2011). The first foray by
one of us into medical research was an attempt to make chickens diabetic in
order to study the effects of hyperglycemia on the enticingly long vagus
nerve (Calcutt & Tomlinson, 1985). The approach, along with many
chickens, was ultimately doomed not only because birds are inherently
hyperglycemic, but also because it became clear that the chicken pancreas
is relatively resistant to the diabetogenic drug streptozotocin (STZ)
(Modak, Datar, Bhonde, & Ghaskadbi, 2007; Simon & Dubois, 1980).
A lifetime quest for a diabetic giraffe continues.
Experimental requirements for an adequate number of observations
to move beyond the anecdotal have restricted research in larger species to
occasional observations of the effects of diabetes on nerve from primates
(Pare et al., 2007), pigs ( Juranek et al., 2010), and dogs (Walker et al.,
2001), all of which are expensive to maintain and have yet to reveal tangible
benefits as models. By far the vast majority of studies of experimental diabetic
neuropathy have therefore been performed in rats and mice. Rodents can be
bred to consistently develop spontaneous type 1 or type 2 diabetes or else can
have diabetes induced by dietary manipulation or chemicals that ablate pan-
creatic beta cells. For many years, rats were the most widely studied species.
This was in part due to their size, which allows surgical manipulations and
yields enough tissue for crude biochemical assays. Rats were also preferred
because the dominant pathogenic mechanism under evaluation during the
1960s to 1990s was glucose flux through the polyol pathway and polyol
pathway metabolites accumulated in the nerve of diabetic rats. Mice were
48 N.A. Calcutt and P. Fernyhough
not widely studied as models for diabetic neuropathy during this period,
other than basic characterization of the db/db and ob/ob mouse models
of spontaneous type 2 diabetes, largely because it was erroneously thought
that they lacked polyol pathway activity in their nerves. Recognition that
lack of accumulation of polyol pathway intermediates in nerve was due
to distinctive enzyme kinetics that makes mice similar to humans in this
respect (Calcutt, Willars, & Tomlinson, 1988; Tomlinson, 1989) and that
polyol pathway flux can drive nerve dysfunction in mice (Calcutt,
Tomlinson, & Biswas, 1990; Ho et al., 2006; Ng et al., 1998), along with
the emergence of new technology that favors use of mice for genetic manip-
ulations, has caused the use of mouse models of both type 1 and type 2
diabetes to greatly expand over the last 10–20 years.
Sliding scales of STZ dose have been developed for rats of different ages and
weights (Calcutt, 2004). In mice, the severity of insulinopenia and its practical
consequences such as conversion rate, survival duration, and frequency of
reversal of diabetes arising from beta cell regeneration can be tempered by
varying both STZ dose and dose frequency. A variety of protocols have been
developed that employ between 2 and 5 low doses of STZ given on consec-
utive days (Biessels et al., 2014). As STZ is inexpensive and relatively easy to
use when following established protocols, the STZ-diabetic rat and mouse
have become the dominant models of diabetic neuropathy.
4. STZ TOXICITY
Concerns are intermittently raised that the cachexia that can occur in
rodents treated with high doses of STZ makes them poor models, particu-
larly for behavioral studies such as those using nociceptive testing to assess
sensory function. Such concerns can be mitigated by appropriate dose titra-
tion of STZ that produces hyperglycemia without the extreme
insulinopenia that underlies cachexia, or by trace insulin replacement
(Calcutt, 2004), along with reporting of performance in rotarod or other
tests. High doses of STZ can also produce direct and acute nephrotoxicity
in mice (Breyer et al., 2005) and direct STZ neurotoxicity has been evoked
to explain tactile allodynia in STZ-injected animals that did not develop
overt hyperglycemia. However, STZ-induced insulinopenia that was not
sufficient to produce overt hyperglycemia but contributes to the pathogen-
esis of allodynia is a more likely explanation (Romanovsky, Cruz, Dienel, &
Dobretsov, 2006). A number of other observations also appear to argue
against meaningful neurotoxic effects of STZ:
• STZ enters cells via the GLUT-2 transporter, which is not found in
peripheral nerve.
• STZ is rapidly cleared from the body, disassociating it from the indices of
neuropathy that can take weeks to emerge.
• Insulin replacement therapy to normalize blood glucose levels and
started days after STZ delivery prevents onset of indices of neuropathy.
• Coinjection of STZ and 6-O-methyl glucose, which competitively
inhibits STZ-uptake by GLUT-2 transporters, produces neither diabetes
nor neuropathy as measured by NCV slowing and loss of heat-induced
pain sensation (Davidson et al., 2009).
• Indices of neuropathy in STZ-injected animals are replicated in other
models of type 1 and type 2 diabetes.
50 N.A. Calcutt and P. Fernyhough
5. NOVEL MODELS
The neuropathy phenotype of commonly used rodent models of dia-
betes has recently been evaluated under the aegis of Neurodiab and the NIH.
The resulting consensus statement is comprehensive and may be viewed
elsewhere (Biessels et al., 2014) so need not be replicated in this volume.
We have therefore taken the opportunity to invite colleagues to instead con-
sider alternative approaches to modeling diabetic neuropathy. A number of
such models, particularly models of type 2 diabetes and metabolic syndrome,
are currently undergoing characterization. The review “Alternatives to the
STZ-Diabetic Rodent” by Yorek is therefore a particularly opportune con-
tribution. Moreover, the pertinence of neuronal and glial cell cultures to
model the toxic impact of hyperglycemia, insulinopenia, or other diabetes-
associated insults is perhaps even more controversial than use of animal
models of diabetic neuropathy, with the advantage of selective manipulation
of the local environment offset by the disadvantage of lack of intercellular
and physiological interactions. The chapter “Can Diabetic Neuropathy
Be Modeled In Vitro?” by Gardiner eloquently addresses these trade-offs
to make a case that, within reason, we can indeed “do it in a dish.”
REFERENCES
Biessels, G. J., Bril, V., Calcutt, N. A., Cameron, N. E., Cotter, M. A., Dobrowsky, R., et al.
(2014). Phenotyping animal models of diabetic neuropathy: A consensus statement of the
diabetic neuropathy study group of the EASD (neurodiab). Journal of the Peripheral Nervous
System, 19, 77–87.
Braun, E. J., & Sweazea, K. L. (2008). Glucose regulation in birds. Comparative Biochemistry
and Physiology. Part B, Biochemistry & Molecular Biology, 151, 1–9.
Breyer, M. D., Bottinger, E., Brosius, F. C., 3rd, Coffman, T. M., Harris, R. C.,
Heilig, C. W., et al. (2005). Mouse models of diabetic nephropathy. Journal of the American
Society of Nephrology, 16, 27–45.
Animal Models of Diabetic Neuropathy 51
O’Brien, T. D. (2002). Pathogenesis of feline diabetes mellitus. Molecular and Cellular Endo-
crinology, 197, 213–219.
Pare, M., Albrecht, P. J., Noto, C. J., Bodkin, N. L., Pittenger, G. L., Schreyer, D. J., et al.
(2007). Differential hypertrophy and atrophy among all types of cutaneous innervation in
the glabrous skin of the monkey hand during aging and naturally occurring type 2 dia-
betes. The Journal of Comparative Neurology, 501, 543–567.
Pradhan Nabzdyk, L., Kuchibhotla, S., Guthrie, P., Chun, M., Auster, M. E., Nabzdyk, C.,
et al. (2013). Expression of neuropeptides and cytokines in a rabbit model of diabetic
neuroischemic wound healing. Journal of Vascular Surgery, 58, 766–775. e712.
Romanovsky, D., Cruz, N. F., Dienel, G. A., & Dobretsov, M. (2006). Mechanical
hyperalgesia correlates with insulin deficiency in normoglycemic streptozotocin-treated
rats. Neurobiology of Disease, 24, 384–394.
Simon, J., & Dubois, M. P. (1980). Failure of a sublethal streptozotocin dose to induce dia-
betes in the chicken. Hormone and Metabolic Research, 12, 631–632.
Singer, M. A. (2011). Insights into biomedicine from animal adaptations. Comprehensive Phys-
iology, 1, 2063–2081.
Tomlinson, D. R. (1989). Polyols and myo-inositol in diabetic neuropathy—Of mice and
men. Mayo Clinic Proceedings, 64, 1030–1033.
van der Kolk, J. H., Hoyer, M. J., Verstappen, F. A., Wolters, M. S., Treskes, M.,
Grinwis, G. C., et al. (2011). Diabetes mellitus in a 50-year-old captive Asian elephant
(Elaphas maximus) bull. The Veterinary Quarterly, 31, 99–101.
Walker, D., Siddique, I., Anderson, H., Gardiner, T. A., Archer, D. B., Boulton, A. J. M.,
et al. (2001). Nerve pathology in the type 1 diabetic dog: Effects of treatment with sul-
indac. Journal of the Peripheral Nervous System, 6, 219–226.
CHAPTER FIVE
Contents
1. Introduction 54
2. Diabetic Neuropathy 56
3. The Somatosensory Nervous System 56
4. Can We Model Diabetic Neuropathy In Vitro? 62
4.1 Choice of Cells 63
4.2 Choice of Stimuli 68
4.3 Choice of Output Measure 69
4.4 Looking Forward to a Better In Vitro Model of Diabetic Neuropathy 76
5. Conclusions 79
Acknowledgments 79
References 79
Abstract
Diabetic neuropathy is a common secondary complication of diabetes that impacts on
patient's health and well-being. Distal axon degeneration is a key feature of diabetic
neuropathy, but the pathological changes which underlie axonal die-back are incom-
pletely understood; despite decades of research a treatment has not yet been identified.
Basic research must focus on understanding the complex mechanisms underlying
changes that occur in the nervous system during diabetes. To this end, tissue culture
techniques are invaluable as they enable researchers to examine the intricate mecha-
nistic responses of cells to high glucose or other factors in order to better understand
the pathogenesis of nerve dysfunction. This chapter describes the use of in vitro models
to study a wide range of specific cellular effects pertaining to diabetic neuropathy
including apoptosis, neurite outgrowth, neurodegeneration, activity, and bioenergetics.
We consider problems associated with in vitro modeling and future refinement such as
use of induced pluripotent stem cells and microfluidic technology.
1. INTRODUCTION
Diabetic neuropathy affects approximately half of all patients with dia-
betes mellitus (Dyck et al., 1993). It can be a debilitating condition that
impacts on patient’s health and quality of life and presents a substantial
expense for health-care providers (Hex, Bartlett, Wright, Taylor, &
Varley, 2012). Patients typically present with sensory symptoms arising in
a “glove and stocking” distribution, with feet commonly affected before
hands. Sensory symptoms are divided into the “positive”: neuropathic pain,
paraesthesia, and dysesthesia; and the “negative”: loss of sensation, decreased
nerve conduction velocity, and nerve dysfunction. Autonomic and motor
neuropathies also occur, and increasing evidence suggests that this secondary
complication of diabetes extends to the central nervous system (CNS). Neu-
rons show a distal–proximal pattern of injury, with the longest axons show-
ing the worst pathology.
Diabetic neuropathy is multifactorial in etiology and many pathogenic
mechanisms have been described (Fernyhough & Calcutt, 2010;
Fernyhough, Roy Chowdhury, & Schmidt, 2010; Freeman et al., 2016;
Obrosova, 2002, 2003; Zochodne, 2014). Despite this, treatment options
are currently limited to glycemic control and/or symptomatic treatment
and are inadequate. Experimental rodent models of diabetes have long been
utilized to assess behavioral, electrophysiological, and morphological deficits
in diabetic neuropathy and to test the efficacy of novel therapeutics. These
models will be considered elsewhere in this volume. The sad fact stands that,
despite many decades of research, a treatment for diabetic neuropathy that
significantly reverses or ameliorates the progression of the disease has not
been identified. The search for efficacious drugs to better treat neuropathic
pain, prevent axon degeneration, and/or promote nerve regeneration is
therefore an urgent clinical need.
Basic research in diabetic neuropathy must focus on understanding
the complex mechanisms underlying the changes that occur in the ner-
vous system. To this end, tissue and cell culture techniques are valuable
as they enable researchers to dissect out the intricate mechanistic responses
of cells to high glucose or other exogenous factors. The crucial benefit
afforded by an in vitro over an in vivo approach is the ability to precisely
control and manipulate the environment (Table 1), thereby allowing
study of a wide range of specific cellular events ranging from survival to
death; neurite outgrowth to neurodegeneration; excitation to inhibition;
Can Diabetic Neuropathy Be Modeled In Vitro? 55
2. DIABETIC NEUROPATHY
The first symptoms of diabetic neuropathy typically arise in the
somatosensory aspect of the peripheral nervous system (PNS). The skin is
innervated by peptidergic and nonpeptidergic unmyelinated afferents that
form free axon terminals in the epidermis, and myelinated afferents that ter-
minate at specialized mechanoreceptors (Fig. 1) (Hilliges, Wang, &
Johansson, 1995; Owens & Lumpkin, 2014). Distal processes of sensory
nerves degenerate in both clinical and experimental diabetic neuropathy
(Kennedy & Zochodne, 2000, 2005; Tomlinson & Gardiner, 2008;
Yasuda et al., 2003), and degeneration occurs at a faster rate than rein-
nervation, leading to loss of sensation. This degeneration, along with other
structural changes in the nerve such as axonal dystrophy, Schwann cell
pathology, paranodal demyelination, and endoneurial microangiopathy,
coincides with electrophysiological changes and sensory dysfunction
(Tomlinson & Gardiner, 2008). Autonomic and motor neuropathies are less
well characterized than sensory neuropathies, possibly reflective of the lower
incidence of motor and visceral autonomic neuropathy in patients with dia-
betes (Dyck et al., 1993; Spallone et al., 2011) and the comparative ease of
studying the somatosensory system in both humans and experimental
models of diabetes. Since the interpretation of in vitro experiments requires
knowledge of the context in which different cell populations function, this
chapter will open with a brief overview of the development, maturation,
and structure of the PNS.
Fig. 1 The peripheral nervous system. (A) The cell bodies of most peripheral sensory
neurons are located in the dorsal root ganglia (DRG). The motor neuron cell bodies
within the ventral horn of the spinal cord extend efferents via the ventral roots to join
the peripheral nerve and terminate in specialized neuromuscular junctions. (B) Sensory
neurons are a heterogeneous population, express different receptors and phenotypic
markers, and respond to different neurotrophic factors. Peripheral sensory afferents ter-
minate as free nerve endings or in specialized sensory receptors such as Pacinian cor-
puscles (PC) in the skin (C). Central branches are directed to the spinal cord and through
collateral branches to the dorsal column nuclei (DCN). The electron micrograph
(D; kindly provided by Prof. David Tomlinson, University of Manchester) shows the inter-
nal organization of a peripheral nerve. Large-diameter sensory and motor axons (AX) are
surrounded by compacted myelin (m), while groups of small-diameter unmyelinated
sensory axons (ax) are grouped in Remak bundles (arrows). Endoneurial ECM, collagen
fibrils (c) surround Schwann cell basement membranes (arrowheads) of both Remak
bundles and myelinated axon/Schwann cell units. Reprinted from Gardiner, N. J.
(2011). Integrins and the extracellular matrix: Key mediators of development and regener-
ation of the sensory nervous system. Developmental Neurobiology, 71, 1054–1072, with
permission from John Wiley and Sons © 2011.
E9.5
1st wave 2nd wave 3rd wave
Premigratory
Multipotent
Sox10+
Sensory
biased Multipotent
Sox10+ Krox20+
Ngn2+ Sox10+ Sox10+
Migratory
SN committed
+
Sox10
+
Ngn2
+
Foxs1
Multipotent Glia
SN committed
Dorsoventral axis
−
Sox10
Postmigratory
+
Foxs1
+ Sox10 Glia?
SN committed?
Brn3a+ +
Sox10
+
Ngn1 SN committed
+
Brn3a −
Sox10
+
Ngn1
+
Brn3a
+
Foxs1
-E10.5–E11.5 Runx3
+
Runx1
+
+ +
TrkC TrkA
-E14.5 -P0–P30
Runx3
downregulation
+ + +
TrkB TrkB TrkC
+ + +
TrkC Ret Runx3 + Ret
+
TrkA −
Ret
+/– TrkA
+
Runx1
+ +
TrkB Ret
Peptidergic Nonpeptidergic
Mechanoreceptors Proprioceptors neurons neurons
No or low RUNX3 RUNX3 No RUNX1 RUNX1
Fig. 2 Genetic control of development of the somatosensory nervous system. In the
first wave of neurogenesis, SOX10+ cells migrate and begin to express neurogenin 2
(Ngn2). Ngn2+ cells subsequently commit to a sensory neuronal (SN) fate and express
forkhead transcription factor (Foxs1+) during migration. These postmigratory neurons
then begin to express Brn3a and form large sensory neurons (TrkC+ and Runx3+) at
an early developmental stage. In contrast, cells with little or no Ngn2 remain uncommit-
ted throughout migration. Postmigratory cells in the DRG then start to express Brn3a,
Ngn1, and Foxs1. They either produce TrkC+ or TrkA+ populations of sensory neurons by
(Continued)
60 N.J. Gardiner and O.J. Freeman
that ensheath axons. Fibronectin is more diffuse, being localized along exter-
nal surfaces of endoneurial tubes and the perineurium. This distinction high-
lights the need to optimize the environment of the culture systems to that of
the cell being studied.
Neurons do not naturally exist in simple two-dimensional monolayers,
but occupy a complex 3-dimensional space closely associated with multiple
cell types and ECM in an environment that is constantly adapting and
66 N.J. Gardiner and O.J. Freeman
Fig. 4 High glucose inhibits Schwann cell migration. Bright-field micrographs from
adult wild-type (WT) mouse DRG explants cultured for 8 days in media containing
NGF (20 ng/mL) with (A, B) 10 or 60 mM glucose (glc) or (C) 60 mM mannitol (man) show
evidence of reduced Schwann cell migration (demarcated with dashed line) in high glu-
cose. Reprinted from Gumy, L. F., Bampton, E. T., & Tolkovsky, A. M. (2008). Hyperglycaemia
inhibits Schwann cell proliferation and migration and restricts regeneration of axons and
Schwann cells from adult murine DRG. Molecular and Cellular Neurosciences, 37,
298–311, © 2008 with permission from Elsevier.
Can Diabetic Neuropathy Be Modeled In Vitro? 67
Hosmane, Thakor, & Hoke, 2009). We envisage that this system will soon
be employed to generate a “diabetes-induced” model of distal axonopathy
(see Section 4.4).
Immortalized cell lines, especially PC12 cells, have long been utilized in
regeneration studies and are useful tools to study neurite outgrowth since cells
differentiated with NGF extend neurite-like processes that can be easily mea-
sured. While neurite outgrowth appears reduced in cells cultured in high
glucose conditions, apoptosis is also often described (Koshimura, Tanaka,
Murakami, & Kato, 2002; Lelkes, Unsworth, & Lelkes, 2001; Sharifi,
Mousavi, Farhadi, & Larijani, 2007), meaning that caution should be exerted
in translating results to the in vivo situation. The ease of use of immortalized
Can Diabetic Neuropathy Be Modeled In Vitro? 75
cell lines has enabled the development of rapid high-throughput assays, pri-
marily exploited by the pharmaceutical industry (Radio, Breier, Shafer, &
Mundy, 2008), to quantify the effects of compounds on neuritogenesis.
High-throughput assay systems treat cells in multiwell plates with com-
pounds, then automatically capture images using fluorescent or bright-field
microscopy, and quantify neurite extension. This unbiased automated
approach to assess neurite outgrowth is useful for rapid drug screening and
toxicological assessment and may be used to identify compounds with regen-
erative properties using novel compounds or existing drug libraries
(Vincent & Feldman, 2008). However, using immortalized cell lines or
embryonic neurons for the study of regenerative pathways is simplistic,
due to the heterogeneous nature of adult neurons, as discussed above. There-
fore it is preferable to use dissociated adult neurons, with their mature com-
plement of neurotrophic factor receptors, for neurite outgrowth assays.
Following an initial quiescent period, adult sensory neurons in culture
exhibit two distinct forms of neurite outgrowth: an early arborizing form that
can be enhanced by addition of neurotrophic factors such as NGF, NT-3, or
GDNF (Fig. 3), and a later elongating regenerative growth mode which
depends on novel gene transcription (Diamond, Foerster, Holmes, &
Coughlin, 1992; Gavazzi et al., 1999; Kimpinski, Campenot, & Mearow,
1997; Smith & Skene, 1997). The regenerative responses of cells exposed
directly to high glucose and diabetes-associated cell stressors have been assessed
using a number of culture paradigms, including comparisons between cells
derived from either control or diabetic rodents. Indeed, reduced intrinsic
regenerative capacity of neurons may be most evident using tissue derived
from diabetic rats. Work from the Fernyhough laboratory clearly illustrates
a difference in susceptibility of neurons from STZ-diabetic rats to metabolic
stress (Akude et al., 2011; Akude, Zherebitskaya, Roy Chowdhury,
Girling, & Fernyhough, 2010; Zherebitskaya et al., 2009, 2012).
While treatment of dissociated sensory neurons from control or diabetic
rats with elevated glucose does not impair neurite outgrowth per se, neurites
of neurons from diabetic rats displayed abnormal swelling and beading along
the axon that immunostained for 4-HNE adducts indicative of lipid perox-
idation (Zherebitskaya et al., 2009). This suggests that high glucose induces
oxidative stress and dysmorphology in axons of adult sensory neurons, but
only those isolated from diabetic rats. Direct exposure of control adult rat
sensory neurons to 4-HNE at concentrations as low as 3 μM caused a sig-
nificant reduction in neurite outgrowth and similar changes to axonal mor-
phology (Akude et al., 2010). Similar studies have been performed using
dissociated CG/SMG neurons, which have a slower regenerative response
76 N.J. Gardiner and O.J. Freeman
in vitro than SCG neurons and were more sensitive to high glucose than
SCG neurons in that exposure to high glucose caused a significant decrease
in neurite-bearing cells from CG/SMG but not SCG (Semra et al., 2004).
While hyperglycemia can impact neurite initiation and morphology, neurite
length is not altered by up to 45 mM glucose in adult mouse DRG explants,
although supraphysiological glucose levels (60 mM) had some effect (Gumy
et al., 2008). There was also no effect on the density of growth cones at any
glucose level tested, indicating that exposure to supraphysiological glucose
did not reduce neurite initiation, but impaired rate of neurite extension.
These experiments also revealed a striking inhibition of Schwann cell migra-
tion from explants in high glucose (Fig. 4) that was confirmed using a
Matrigel™ drop assay of isolated neonatal Schwann cells in 30 mM glucose
(Gumy et al., 2008). The proliferation of Schwann cells thus appears partic-
ularly sensitive to elevated glucose concentrations, which could impact on
regenerative capacity in diabetic neuropathy.
Another key aspect to the regenerative deficit in diabetic neuropathy is an
unsupportive extracellular environment, both in terms of neurotrophic support
from other cells and the ECM (Bradley, King, Muddle, & Thomas, 2000;
Calcutt, Jolivalt, & Fernyhough, 2008; Gardiner, 2011; Kennedy &
Zochodne, 2005; King, Llewelyn, Thomas, Gilbey, & Watkins, 1989;
Yasuda et al., 2003). The ECM of peripheral nerve plays a key role in supporting
axonal regeneration, and attachment of neurons to ECM is an important phase
for successful neurite outgrowth in culture (Duran-Jimenez et al., 2009). Dis-
sociated sensory neurons from adult diabetic mice show reduced adhesion to
laminin, type I and IV collagens, and fibronectin (Sango, Horie, Okamura,
Inoue, & Takenaka, 1995), possibly through altered expression of ECM recep-
tors. ECM proteins are glycated in the peripheral nerve in diabetes (Duran-
Jimenez et al., 2009), and exogenous glycation of purified ECM proteins using
glucose, glyceraldehyde, or methylglyoxal inhibited neurite outgrowth from
neuroblastoma cells (Federoff, Lawrence, & Brownlee, 1993), dissociated neo-
natal (Luo, King, Lewin, & Thomas, 2002) and adult sensory neurons
(Duran-Jimenez et al., 2009; Radu et al., 2012), and adult DRG explants
(Ozturk, Sekeroglu, Erdogan, & Ozturk, 2006). Reducing AGE formation
in the peripheral nerve and target tissues may thereby diminish the inhibitory
environment and enable reinnervation of target tissue.
5. CONCLUSIONS
Can we model diabetic neuropathy in vitro? The answer is perhaps
“no, or at least not in its entirety.” However, we can now model many
aspects of neuropathy effectively. The in vitro models used to study the
pathophysiological responses to hyperglycemia, reduced insulin signaling,
hyperlipidemia, and other stressors, while simplistic, do enable us to predict
how neurons may behave in more complex in vivo environments. They also
enable us to investigate subcellular responses and dissect out mechanistic
changes that contribute to the pathogenesis of the disease, test neuro-
protective strategies, and develop high-throughput drug discovery screens
for drugs to promote axonal regeneration. As such, they will remain a
valuable component to the neurobiologists’ toolbox. Future refinement
of models, including use of iPSCs and the ability to challenge different
neuronal compartments using microfluidic devices, will hopefully enable
us to discover underlying mechanisms that predispose the distal axon to
degeneration in diabetic neuropathy and improve treatment options.
ACKNOWLEDGMENTS
The authors would like to extend thanks to the Juvenile Diabetes Research Foundation,
Diabetes UK and the Medical Research Council, UK for funding their research over the
past years. We apologize to the many authors whose work we were not able to cite due
to space constraints. We thank the editors Paul Fernyhough and Nigel Calcutt for their
helpful comments.
REFERENCES
Akude, E., Zherebitskaya, E., Chowdhury, S. K., Smith, D. R., Dobrowsky, R. T., &
Fernyhough, P. (2011). Diminished superoxide generation is associated with respiratory
chain dysfunction and changes in the mitochondrial proteome of sensory neurons from
diabetic rats. Diabetes, 60, 288–297.
Akude, E., Zherebitskaya, E., Roy Chowdhury, S. K., Girling, K., & Fernyhough, P. (2010).
4-Hydroxy-2-nonenal induces mitochondrial dysfunction and aberrant axonal out-
growth in adult sensory neurons that mimics features of diabetic neuropathy. Neurotox-
icity Research, 17, 28–38.
Allodi, I., Udina, E., & Navarro, X. (2012). Specificity of peripheral nerve regeneration:
Interactions at the axon level. Progress in Neurobiology, 98, 16–37.
Averill, S., McMahon, S. B., Clary, D. O., Reichardt, L. F., & Priestley, J. V. (1995). Immu-
nocytochemical localization of trkA receptors in chemically identified subgroups of adult
rat sensory neurons. The European Journal of Neuroscience, 7, 1484–1494.
Bennett, D. L., Averill, S., Clary, D. O., Priestley, J. V., & McMahon, S. B. (1996). Postnatal
changes in the expression of the trkA high-affinity NGF receptor in primary sensory neu-
rons. The European Journal of Neuroscience, 8, 2204–2208.
80 N.J. Gardiner and O.J. Freeman
Bhattacherjee, A., Liao, Z., & Smith, P. G. (2014). Trophic factor and hormonal regulation
of neurite outgrowth in sensory neuron-like 50B11 cells. Neuroscience Letters, 558,
120–125.
Bierhaus, A., Fleming, T., Stoyanov, S., Leffler, A., Babes, A., Neacsu, C., et al. (2012).
Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes
hyperalgesia in diabetic neuropathy. Nature Medicine, 18, 926–933.
Bisby, M. A. (1980). Axonal transport of labeled protein and regeneration rate in nerves of
streptozocin-diabetic rats. Experimental Neurology, 69, 74–84.
Bradley, J. L., King, R. H., Muddle, J. R., & Thomas, P. K. (2000). The extracellular matrix
of peripheral nerve in diabetic polyneuropathy. Acta Neuropathologica, 99, 539–546.
Bradley, J. L., Thomas, P. K., King, R. H., Muddle, J. R., Ward, J. D., Tesfaye, S., et al.
(1995). Myelinated nerve fibre regeneration in diabetic sensory polyneuropathy: Corre-
lation with type of diabetes. Acta Neuropathologica, 90, 403–410.
Bronner-Fraser, M., Stern, C. D., & Fraser, S. (1991). Analysis of neural crest cell lineage and
migration. Journal of Craniofacial Genetics and Developmental Biology, 11, 214–222.
Brownlee, M. (2001). Biochemistry and molecular cell biology of diabetic complications.
Nature, 414, 813–820.
Brownlee, M. (2005). The pathobiology of diabetic complications: A unifying mechanism.
Diabetes, 54, 1615–1625.
Calcutt, N. A., Jolivalt, C. G., & Fernyhough, P. (2008). Growth factors as therapeutics for
diabetic neuropathy. Current Drug Targets, 9, 47–59.
Calcutt, N. A., Muir, D., Powell, H. C., & Mizisin, A. P. (1992). Reduced ciliary neuro-
notrophic factor-like activity in nerves from diabetic or galactose-fed rats. Brain Research,
575, 320–324.
Campenot, R. B. (1977). Local control of neurite development by nerve growth factor.
Proceedings of the National Academy of Sciences of the United States of America, 74,
4516–4519.
Chambers, S. M., Qi, Y., Mica, Y., Lee, G., Zhang, X. J., Niu, L., et al. (2012). Combined
small-molecule inhibition accelerates developmental timing and converts human plurip-
otent stem cells into nociceptors. Nature Biotechnology, 30, 715–720.
Chen, C. L., Broom, D. C., Liu, Y., de Nooij, J. C., Li, Z., Cen, C., et al. (2006). Runx1
determines nociceptive sensory neuron phenotype and is required for thermal and neu-
ropathic pain. Neuron, 49, 365–377.
Chen, W., Mi, R., Haughey, N., Oz, M., & Hoke, A. (2007). Immortalization and charac-
terization of a nociceptive dorsal root ganglion sensory neuronal line. Journal of the Periph-
eral Nervous System: JPNS, 12, 121–130.
Chen, S., Rio, C., Ji, R. R., Dikkes, P., Coggeshall, R. E., Woolf, C. J., et al. (2003). Dis-
ruption of ErbB receptor signaling in adult non-myelinating Schwann cells causes pro-
gressive sensory loss. Nature Neuroscience, 6, 1186–1193.
Chen, Z. L., & Strickland, S. (2003). Laminin gamma1 is critical for Schwann cell differen-
tiation, axon myelination, and regeneration in the peripheral nerve. The Journal of Cell
Biology, 163, 889–899.
Cheng, C., & Zochodne, D. W. (2003). Sensory neurons with activated caspase-3 survive
long-term experimental diabetes. Diabetes, 52, 2363–2371.
Chowdhury, S. K., Smith, D. R., & Fernyhough, P. (2013). The role of aberrant mitochon-
drial bioenergetics in diabetic neuropathy. Neurobiology of Disease, 51, 56–65.
Chwalek, K., Tang-Schomer, M. D., Omenetto, F. G., & Kaplan, D. L. (2015). In vitro
bioengineered model of cortical brain tissue. Nature Protocols, 10, 1362–1373.
Court, F. A., Hendriks, W. T., MacGillavry, H. D., Alvarez, J., & van Minnen, J. (2008).
Schwann cell to axon transfer of ribosomes: Toward a novel understanding of the role
of glia in the nervous system. The Journal of Neuroscience: The Official Journal of the Society for
Neuroscience, 28, 11024–11029.
Can Diabetic Neuropathy Be Modeled In Vitro? 81
Craner, M. J., Klein, J. P., Renganathan, M., Black, J. A., & Waxman, S. G. (2002). Changes
of sodium channel expression in experimental painful diabetic neuropathy. Annals of
Neurology, 52, 786–792.
Delaney, C. L., Russell, J. W., Cheng, H. L., & Feldman, E. L. (2001). Insulin-like growth
factor-I and over-expression of Bcl-xL prevent glucose-mediated apoptosis in Schwann
cells. Journal of Neuropathology and Experimental Neurology, 60, 147–160.
Diamond, J., Foerster, A., Holmes, M., & Coughlin, M. (1992). Sensory nerves in adult rats
regenerate and restore sensory function to the skin independently of endogenous NGF.
The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 12, 1467–1476.
Dingle, Y. L., Boutin, M. E., Chirila, A. M., Livi, L. L., Labriola, N. R., Jakubek, L. M., et al.
(2015). 3D neural spheroid culture: An in vitro model for cortical studies. Tissue Engi-
neering Part C, Methods, 21, 1274–1283.
Duran-Jimenez, B., Dobler, D., Moffatt, S., Rabbani, N., Streuli, C. H., Thornalley, P. J.,
et al. (2009). Advanced glycation end products in extracellular matrix proteins contribute
to the failure of sensory nerve regeneration in diabetes. Diabetes, 58, 2893–2903.
Dyck, P. J., Kratz, K. M., Karnes, J. L., Litchy, W. J., Klein, R., Pach, J. M., et al. (1993). The
prevalence by staged severity of various types of diabetic neuropathy, retinopathy, and
nephropathy in a population-based cohort: The Rochester Diabetic Neuropathy Study.
Neurology, 43, 817–824.
Ekstrom, A. R., & Tomlinson, D. R. (1989). Impaired nerve regeneration in streptozotocin-
diabetic rats. Effects of treatment with an aldose reductase inhibitor. Journal of the Neu-
rological Sciences, 93, 231–237.
Eldridge, C. F., Bunge, M. B., Bunge, R. P., & Wood, P. M. (1987). Differentiation of axon-
related Schwann cells in vitro. I. Ascorbic acid regulates basal lamina assembly and myelin
formation. The Journal of Cell Biology, 105, 1023–1034.
Federoff, H. J., Lawrence, D., & Brownlee, M. (1993). Nonenzymatic glycosylation of lam-
inin and the laminin peptide CIKVAVS inhibits neurite outgrowth. Diabetes, 42,
509–513.
Feinberg, K., Eshed-Eisenbach, Y., Frechter, S., Amor, V., Salomon, D., Sabanay, H., et al.
(2010). A glial signal consisting of gliomedin and NrCAM clusters axonal Na + channels
during the formation of nodes of Ranvier. Neuron, 65, 490–502.
Fernyhough, P. (2015). Mitochondrial dysfunction in diabetic neuropathy: A series of unfor-
tunate metabolic events. Current Diabetes Reports, 15, 89.
Fernyhough, P., & Calcutt, N. A. (2010). Abnormal calcium homeostasis in peripheral neu-
ropathies. Cell Calcium, 47, 130–139.
Fernyhough, P., & McGavock, J. (2014). Mechanisms of disease: Mitochondrial dysfunction
in sensory neuropathy and other complications in diabetes. Handbook of Clinical Neurol-
ogy, 126, 353–377.
Fernyhough, P., Roy Chowdhury, S. K., & Schmidt, R. E. (2010). Mitochondrial stress and
the pathogenesis of diabetic neuropathy. Expert Review of Endocrinology & Metabolism, 5,
39–49.
Freeman, O. J., Unwin, R. D., Dowsey, A. W., Begley, P., Ali, S., Hollywood, K. A., et al.
(2016). Metabolic dysfunction is restricted to the sciatic nerve in experimental diabetic
neuropathy. Diabetes, 65, 228–238.
Fuchs, D., Birklein, F., Reeh, P. W., & Sauer, S. K. (2010). Sensitized peripheral nociception
in experimental diabetes of the rat. Pain, 151, 496–505.
Gardiner, N. J. (2011). Integrins and the extracellular matrix: Key mediators of development
and regeneration of the sensory nervous system. Developmental Neurobiology, 71,
1054–1072.
Gardiner, N. J., Fernyhough, P., Tomlinson, D. R., Mayer, U., von der Mark, H., &
Streuli, C. H. (2005). Alpha7 integrin mediates neurite outgrowth of distinct populations
of adult sensory neurons. Molecular and Cellular Neurosciences, 28, 229–240.
82 N.J. Gardiner and O.J. Freeman
Gardiner, N. J., Moffatt, S., Fernyhough, P., Humphries, M. J., Streuli, C. H., &
Tomlinson, D. R. (2007). Preconditioning injury-induced neurite outgrowth of adult
rat sensory neurons on fibronectin is mediated by mobilisation of axonal alpha5 integrin.
Molecular and Cellular Neurosciences, 35, 249–260.
Gavazzi, I., Kumar, R. D., McMahon, S. B., & Cohen, J. (1999). Growth responses of dif-
ferent subpopulations of adult sensory neurons to neurotrophic factors in vitro. The Euro-
pean Journal of Neuroscience, 11, 3405–3414.
Guan, W., Puthenveedu, M. A., & Condic, M. L. (2003). Sensory neuron subtypes have
unique substratum preference and receptor expression before target innervation. The
Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 23, 1781–1791.
Gumy, L. F., Bampton, E. T., & Tolkovsky, A. M. (2008). Hyperglycaemia inhibits
Schwann cell proliferation and migration and restricts regeneration of axons and
Schwann cells from adult murine DRG. Molecular and Cellular Neurosciences, 37, 298–311.
Harris, M. L., & Erickson, C. A. (2007). Lineage specification in neural crest cell pathfinding.
Developmental Dynamics: An Official Publication of the American Association of Anatomists,
236, 1–19.
Heng, L. J., Qi, R., Yang, R. H., & Xu, G. Z. (2015). Docosahexaenoic acid inhibits
mechanical allodynia and thermal hyperalgesia in diabetic rats by decreasing the excitabil-
ity of DRG neurons. Experimental Neurology, 271, 291–300.
Hex, N., Bartlett, C., Wright, D., Taylor, M., & Varley, D. (2012). Estimating the current
and future costs of Type 1 and Type 2 diabetes in the UK, including direct health costs
and indirect societal and productivity costs. Diabetic Medicine: A Journal of the British Dia-
betic Association, 29, 855–862.
Hilliges, M., Wang, L., & Johansson, O. (1995). Ultrastructural evidence for nerve fibers
within all vital layers of the human epidermis. The Journal of Investigative Dermatology,
104, 134–137.
Hinder, L. M., Figueroa-Romero, C., Pacut, C., Hong, Y., Vivekanandan-Giri, A.,
Pennathur, S., et al. (2014). Long-chain acyl coenzyme A synthetase 1 overexpression
in primary cultured Schwann cells prevents long chain fatty acid-induced oxidative stress
and mitochondrial dysfunction. Antioxidants & Redox Signaling, 21, 588–600.
Hopkins, A. M., DeSimone, E., Chwalek, K., & Kaplan, D. L. (2015). 3D in vitro modeling
of the central nervous system. Progress in Neurobiology, 125, 1–25.
Huang, T. J., Price, S. A., Chilton, L., Calcutt, N. A., Tomlinson, D. R., Verkhratsky, A.,
et al. (2003). Insulin prevents depolarization of the mitochondrial inner membrane in
sensory neurons of type 1 diabetic rats in the presence of sustained hyperglycemia.
Diabetes, 52, 2129–2136.
Huang, T. J., Verkhratsky, A., & Fernyhough, P. (2005). Insulin enhances mitochondrial
inner membrane potential and increases ATP levels through phosphoinositide 3-kinase
in adult sensory neurons. Molecular and Cellular Neurosciences, 28, 42–54.
Jessen, K. R., & Mirsky, R. (1997). Embryonic Schwann cell development: The biology of
Schwann cell precursors and early Schwann cells. Journal of Anatomy, 191(Pt. 4),
501–505.
Jessen, K. R., & Mirsky, R. (2005). The origin and development of glial cells in peripheral
nerves. Nature Reviews Neuroscience, 6, 671–682.
Kaewkhaw, R., Scutt, A. M., & Haycock, J. W. (2012). Integrated culture and purification of
rat Schwann cells from freshly isolated adult tissue. Nature Protocols, 7, 1996–2004.
Kamiya, H., Zhang, W., & Sima, A. A. (2006). Degeneration of the Golgi and neuronal loss
in dorsal root ganglia in diabetic BioBreeding/Worcester rats. Diabetologia, 49,
2763–2774.
Kamiya, H., Zhangm, W., & Sima, A. A. (2005). Apoptotic stress is counterbalanced by sur-
vival elements preventing programmed cell death of dorsal root ganglions in subacute
type 1 diabetic BB/Wor rats. Diabetes, 54, 3288–3295.
Can Diabetic Neuropathy Be Modeled In Vitro? 83
Karchewski, L. A., Kim, F. A., Johnston, J., McKnight, R. M., & Verge, V. M. (1999). Ana-
tomical evidence supporting the potential for modulation by multiple neurotrophins in
the majority of adult lumbar sensory neurons. The Journal of Comparative Neurology, 413,
327–341.
Kennedy, J. M., & Zochodne, D. W. (2000). The regenerative deficit of peripheral nerves in
experimental diabetes: Its extent, timing and possible mechanisms. Brain: A Journal of
Neurology, 123(Pt. 10), 2118–2129.
Kennedy, J. M., & Zochodne, D. W. (2005). Impaired peripheral nerve regeneration in dia-
betes mellitus. Journal of the Peripheral Nervous System: JPNS, 10, 144–157.
Kimpinski, K., Campenot, R. B., & Mearow, K. (1997). Effects of the neurotrophins nerve
growth factor, neurotrophin-3, and brain-derived neurotrophic factor (BDNF) on neu-
rite growth from adult sensory neurons in compartmented cultures. Journal of Neurobiol-
ogy, 33, 395–410.
King, R. H., Llewelyn, J. G., Thomas, P. K., Gilbey, S. G., & Watkins, P. J. (1989). Diabetic
neuropathy: Abnormalities of Schwann cell and perineurial basal laminae. Implications
for diabetic vasculopathy. Neuropathology and Applied Neurobiology, 15, 339–355.
Kirkpatrick, L. L., & Brady, S. T. (1994). Modulation of the axonal microtubule cytoskeleton
by myelinating Schwann cells. The Journal of Neuroscience: The Official Journal of the Society
for Neuroscience, 14, 7440–7450.
Kiskinis, E., Sandoe, J., Williams, L. A., Boulting, G. L., Moccia, R., Wainger, B. J., et al.
(2014). Pathways disrupted in human ALS motor neurons identified through genetic
correction of mutant SOD1. Cell Stem Cell, 14, 781–795.
Koshimura, K., Tanaka, J., Murakami, Y., & Kato, Y. (2002). Involvement of nitric oxide in
glucose toxicity on differentiated PC12 cells: Prevention of glucose toxicity by
tetrahydrobiopterin, a cofactor for nitric oxide synthase. Neuroscience Research, 43, 31–38.
Kramer, I., Sigrist, M., de Nooij, J. C., Taniuchi, I., Jessell, T. M., & Arber, S. (2006). A role
for Runx transcription factor signaling in dorsal root ganglion sensory neuron diversifi-
cation. Neuron, 49, 379–393.
Lallemend, F., & Ernfors, P. (2012). Molecular interactions underlying the specification of
sensory neurons. Trends in Neurosciences, 35, 373–381.
Lawson, S. N. (1995). Neuropeptides in morphologically and functionally identified primary
afferent neurons in dorsal root ganglia: Substance P, CGRP and somatostatin. Progress in
Brain Research, 104, 161–173.
Lawson, S. N., & Biscoe, T. J. (1979). Development of mouse dorsal root ganglia: An auto-
radiographic and quantitative study. Journal of Neurocytology, 8, 265–274.
Lawson, S. N., Perry, M. J., Prabhakar, E., & McCarthy, P. W. (1993). Primary sensory neu-
rones: Neurofilament, neuropeptides, and conduction velocity. Brain Research Bulletin,
30, 239–243.
Lelkes, E., Unsworth, B. R., & Lelkes, P. I. (2001). Reactive oxygen species, apoptosis and
altered NGF-induced signaling in PC12 pheochromocytoma cells cultured in elevated
glucose: An in vitro cellular model for diabetic neuropathy. Neurotoxicity Research, 3,
189–203.
Lindsay, R. M. (1988). Nerve growth factors (NGF, BDNF) enhance axonal regeneration
but are not required for survival of adult sensory neurons. The Journal of Neuroscience: The
Official Journal of the Society for Neuroscience, 8, 2394–2405.
Liu, Q., Spusta, S. C., Mi, R., Lassiter, R. N., Stark, M. R., Hoke, A., et al. (2012). Human
neural crest stem cells derived from human ESCs and induced pluripotent stem cells:
Induction, maintenance, and differentiation into functional schwann cells. Stem Cells
Translational Medicine, 1, 266–278.
Luo, Z. J., King, R. H., Lewin, J., & Thomas, P. K. (2002). Effects of nonenzymatic glyco-
sylation of extracellular matrix components on cell survival and sensory neurite extension
in cell culture. Journal of Neurology, 249, 424–431.
84 N.J. Gardiner and O.J. Freeman
Marmigere, F., & Ernfors, P. (2007). Specification and connectivity of neuronal subtypes in
the sensory lineage. Nature Reviews Neuroscience, 8, 114–127.
Marmigere, F., Montelius, A., Wegner, M., Groner, Y., Reichardt, L. F., & Ernfors, P.
(2006). The Runx1/AML1 transcription factor selectively regulates development and
survival of TrkA nociceptive sensory neurons. Nature Neuroscience, 9, 180–187.
McMahon, S. B., Armanini, M. P., Ling, L. H., & Phillips, H. S. (1994). Expression and
coexpression of Trk receptors in subpopulations of adult primary sensory neurons
projecting to identified peripheral targets. Neuron, 12, 1161–1171.
Melli, G., Jack, C., Lambrinos, G. L., Ringkamp, M., & Hoke, A. (2006). Erythropoietin
protects sensory axons against paclitaxel-induced distal degeneration. Neurobiology of Dis-
ease, 24, 525–530.
Melli, G., Keswani, S. C., Fischer, A., Chen, W., & Hoke, A. (2006). Spatially distinct and
functionally independent mechanisms of axonal degeneration in a model of HIV-
associated sensory neuropathy. Brain: A Journal of Neurology, 129, 1330–1338.
Michailov, G. V., Sereda, M. W., Brinkmann, B. G., Fischer, T. M., Haug, B.,
Birchmeier, C., et al. (2004). Axonal neuregulin-1 regulates myelin sheath thickness.
Science, 304, 700–703.
Mirsky, R., Jessen, K. R., Brennan, A., Parkinson, D., Dong, Z., Meier, C., et al. (2002).
Schwann cells as regulators of nerve development. Journal of Physiology, Paris, 96, 17–24.
Molliver, D. C., Radeke, M. J., Feinstein, S. C., & Snider, W. D. (1995). Presence or absence
of TrkA protein distinguishes subsets of small sensory neurons with unique cytochemical
characteristics and dorsal horn projections. The Journal of Comparative Neurology, 361,
404–416.
Molliver, D. C., & Snider, W. D. (1997). Nerve growth factor receptor TrkA is down-
regulated during postnatal development by a subset of dorsal root ganglion neurons.
The Journal of Comparative Neurology, 381, 428–438.
Molliver, D. C., Wright, D. E., Leitner, M. L., Parsadanian, A. S., Doster, K., Wen, D., et al.
(1997). IB4-binding DRG neurons switch from NGF to GDNF dependence in early
postnatal life. Neuron, 19, 849–861.
Murinson, B. B., & Griffin, J. W. (2004). C-fiber structure varies with location in peripheral
nerve. Journal of Neuropathology and Experimental Neurology, 63, 246–254.
Murinson, B. B., Hoffman, P. N., Banihashemi, M. R., Meyer, R. A., & Griffin, J. W.
(2005). C-fiber (Remak) bundles contain both isolectin B4-binding and calcitonin gene-
related peptide-positive axons. The Journal of Comparative Neurology, 484, 392–402.
Nishikawa, T., Edelstein, D., Du, X. L., Yamagishi, S., Matsumura, T., Kaneda, Y., et al.
(2000). Normalizing mitochondrial superoxide production blocks three pathways of
hyperglycaemic damage. Nature, 404, 787–790.
Obrosova, I. G. (2002). How does glucose generate oxidative stress in peripheral nerve? Inter-
national Review of Neurobiology, 50, 3–35.
Obrosova, I. G. (2003). Update on the pathogenesis of diabetic neuropathy. Current Diabetes
Reports, 3, 439–445.
Owens, D. M., & Lumpkin, E. A. (2014). Diversification and specialization of touch
receptors in skin. Cold Spring Harbor Perspectives in Medicine, 4, pii: a013656.
Ozturk, G., Sekeroglu, M. R., Erdogan, E., & Ozturk, M. (2006). The effect of non-
enzymatic glycation of extracellular matrix proteins on axonal regeneration in vitro. Acta
Neuropathologica, 112, 627–632.
Polydefkis, M., Hauer, P., Sheth, S., Sirdofsky, M., Griffin, J. W., & McArthur, J. C. (2004).
The time course of epidermal nerve fibre regeneration: Studies in normal controls and in
people with diabetes, with and without neuropathy. Brain: A Journal of Neurology, 127,
1606–1615.
Purves, T., Middlemas, A., Agthong, S., Jude, E. B., Boulton, A. J., Fernyhough, P., et al.
(2001). A role for mitogen-activated protein kinases in the etiology of diabetic
Can Diabetic Neuropathy Be Modeled In Vitro? 85
neuropathy. FASEB Journal: Official Publication of the Federation of American Societies for
Experimental Biology, 15, 2508–2514.
Radio, N. M., Breier, J. M., Shafer, T. J., & Mundy, W. R. (2008). Assessment of chemical
effects on neurite outgrowth in PC12 cells using high content screening. Toxicological
Sciences: An Official Journal of the Society of Toxicology, 105, 106–118.
Radu, B. M., Dumitrescu, D. I., Mustaciosu, C. C., & Radu, M. (2012). Dual effect of
methylglyoxal on the intracellular Ca2+ signaling and neurite outgrowth in mouse sen-
sory neurons. Cellular and Molecular Neurobiology, 32, 1047–1057.
Ristic, H., Srinivasan, S., Hall, K. E., Sima, A. A., & Wiley, J. W. (1998). Serum from dia-
betic BB/W rats enhances calcium currents in primary sensory neurons. Journal of Neu-
rophysiology, 80, 1236–1244.
Russell, J. W., & Feldman, E. L. (1999). Insulin-like growth factor-I prevents apoptosis in
sympathetic neurons exposed to high glucose. Hormone and Metabolic Research ¼ Hormon-
und Stoffwechselforschung ¼ Hormones et metabolisme, 31, 90–96.
Russell, J. W., Golovoy, D., Vincent, A. M., Mahendru, P., Olzmann, J. A., Mentzer, A.,
et al. (2002). High glucose-induced oxidative stress and mitochondrial dysfunction in
neurons. FASEB Journal: Official Publication of the Federation of American Societies for Exper-
imental Biology, 16, 1738–1748.
Salzer, J. L., Brophy, P. J., & Peles, E. (2008). Molecular domains of myelinated axons in the
peripheral nervous system. Glia, 56, 1532–1540.
Sandoe, J., & Eggan, K. (2013). Opportunities and challenges of pluripotent stem cell neu-
rodegenerative disease models. Nature Neuroscience, 16, 780–789.
Sanes, J. R. (1983). Roles of extracellular matrix in neural development. Annual Review of
Physiology, 45, 581–600.
Sango, K., Horie, H., Okamura, A., Inoue, S., & Takenaka, T. (1995). Diabetes impairs
DRG neuronal attachment to extracellular matrix proteins in vitro. Brain Research Bul-
letin, 37, 533–537.
Sango, K., Yanagisawa, H., Takaku, S., Kawakami, E., & Watabe, K. (2011). Immortalized
adult rodent Schwann cells as in vitro models to study diabetic neuropathy. Experimental
Diabetes Research, 2011, 374943.
Schmidt, R. E. (2001). Neuronal preservation in the sympathetic ganglia of rats with chronic
streptozotocin-induced diabetes. Brain Research, 921, 256–259.
Semra, Y. K., Smith, N. C., & Lincoln, J. (2004). Comparative effects of high glucose on
different adult sympathetic neurons in culture. Neuroreport, 15, 2321–2325.
Sharifi, A. M., Mousavi, S. H., Farhadi, M., & Larijani, B. (2007). Study of high glucose-
induced apoptosis in PC12 cells: Role of bax protein. Journal of Pharmacological Sciences,
104, 258–262.
Silos-Santiago, I., Molliver, D. C., Ozaki, S., Smeyne, R. J., Fagan, A. M., Barbacid, M.,
et al. (1995). Non-TrkA-expressing small DRG neurons are lost in TrkA deficient mice.
The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 15, 5929–5942.
Silva, A., Wang, Q., Wang, M., Ravula, S. K., & Glass, J. D. (2006). Evidence for direct
axonal toxicity in vincristine neuropathy. Journal of the Peripheral Nervous System: JPNS,
11, 211–216.
Smith, D. S., & Skene, J. H. (1997). A transcription-dependent switch controls competence
of adult neurons for distinct modes of axon growth. The Journal of Neuroscience: The Official
Journal of the Society for Neuroscience, 17, 646–658.
Snider, W. D., & Silos-Santiago, I. (1996). Dorsal root ganglion neurons require functional
neurotrophin receptors for survival during development. Philosophical Transactions of the
Royal Society of London. Series B, Biological Sciences, 351, 395–403.
Spallone, V., Ziegler, D., Freeman, R., Bernardi, L., Frontoni, S., Pop-Busui, R., et al.
(2011). Cardiovascular autonomic neuropathy in diabetes: Clinical impact, assessment,
diagnosis, and management. Diabetes/Metabolism Research and Reviews, 27, 639–653.
86 N.J. Gardiner and O.J. Freeman
Srinivasan, S., Stevens, M. J., Sheng, H., Hall, K. E., & Wiley, J. W. (1998). Serum from
patients with type 2 diabetes with neuropathy induces complement-independent,
calcium-dependent apoptosis in cultured neuronal cells. The Journal of Clinical Investiga-
tion, 102, 1454–1462.
Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., et al. (2007).
Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell,
131, 861–872.
Tannenbaum, J., & Bennett, B. T. (2015). Russell and Burch’s 3Rs then and now: The need
for clarity in definition and purpose. Journal of the American Association for Laboratory Animal
Science: JAALAS, 54, 120–132.
Taveggia, C., Zanazzi, G., Petrylak, A., Yano, H., Rosenbluth, J., Einheber, S., et al. (2005).
Neuregulin-1 type III determines the ensheathment fate of axons. Neuron, 47, 681–694.
Thakur, M., Crow, M., Richards, N., Davey, G. I., Levine, E., Kelleher, J. H., et al. (2014).
Defining the nociceptor transcriptome. Frontiers in Molecular Neuroscience, 7, 87.
Tomlinson, D. R., & Gardiner, N. J. (2008). Glucose neurotoxicity. Nature Reviews
Neuroscience, 9, 36–45.
Tsantoulas, C., Farmer, C., Machado, P., Baba, K., McMahon, S. B., & Raouf, R. (2013).
Probing functional properties of nociceptive axons using a microfluidic culture system.
PLoS One, 8, e80722.
Usoskin, D., Furlan, A., Islam, S., Abdo, H., Lonnerberg, P., Lou, D., et al. (2015). Unbiased
classification of sensory neuron types by large-scale single-cell RNA sequencing. Nature
Neuroscience, 18, 145–153.
Viader, A., Golden, J. P., Baloh, R. H., Schmidt, R. E., Hunter, D. A., & Milbrandt, J.
(2011). Schwann cell mitochondrial metabolism supports long-term axonal survival
and peripheral nerve function. The Journal of Neuroscience: The Official Journal of the Society
for Neuroscience, 31, 10128–10140.
Viader, A., Sasaki, Y., Kim, S., Strickland, A., Workman, C. S., Yang, K., et al. (2013). Aber-
rant Schwann cell lipid metabolism linked to mitochondrial deficits leads to axon degen-
eration and neuropathy. Neuron, 77, 886–898.
Vincent, A. M., & Feldman, E. L. (2008). Can drug screening lead to candidate therapies for
testing in diabetic neuropathy? Antioxidants & Redox Signaling, 10, 387–393.
Vincent, A. M., Hayes, J. M., McLean, L. L., Vivekanandan-Giri, A., Pennathur, S., &
Feldman, E. L. (2009). Dyslipidemia-induced neuropathy in mice: The role of
oxLDL/LOX-1. Diabetes, 58, 2376–2385.
Vincent, A. M., Olzmann, J. A., Brownlee, M., Sivitz, W. I., & Russell, J. W. (2004).
Uncoupling proteins prevent glucose-induced neuronal oxidative stress and
programmed cell death. Diabetes, 53, 726–734.
Vincent, A. M., Russell, J. W., Low, P., & Feldman, E. L. (2004). Oxidative stress in the
pathogenesis of diabetic neuropathy. Endocrine Reviews, 25, 612–628.
Vogelbaum, M. A., Tong, J. X., & Rich, K. M. (1998). Developmental regulation of apo-
ptosis in dorsal root ganglion neurons. The Journal of Neuroscience: The Official Journal of the
Society for Neuroscience, 18, 8928–8935.
Wilson, N. M., & Wright, D. E. (2014). Experimental motor neuropathy in diabetes. Hand-
book of Clinical Neurology, 126, 461–467.
Wright, D. E., & Snider, W. D. (1995). Neurotrophin receptor mRNA expression defines
distinct populations of neurons in rat dorsal root ganglia. The Journal of Comparative Neu-
rology, 351, 329–338.
Yang, I. H., Siddique, R., Hosmane, S., Thakor, N., & Hoke, A. (2009). Compartmental-
ized microfluidic culture platform to study mechanism of paclitaxel-induced axonal
degeneration. Experimental Neurology, 218, 124–128.
Yasuda, H., Terada, M., Maeda, K., Kogawa, S., Sanada, M., Haneda, M., et al. (2003). Dia-
betic neuropathy and nerve regeneration. Progress in Neurobiology, 69, 229–285.
Can Diabetic Neuropathy Be Modeled In Vitro? 87
Yu, C., Rouen, S., & Dobrowsky, R. T. (2008). Hyperglycemia and downregulation of
caveolin-1 enhance neuregulin-induced demyelination. Glia, 56, 877–887.
Zhang, Y., Bekku, Y., Dzhashiashvili, Y., Armenti, S., Meng, X., Sasaki, Y., et al. (2012).
Assembly and maintenance of nodes of Ranvier rely on distinct sources of proteins and
targeting mechanisms. Neuron, 73, 92–107.
Zhang, K., Osakada, Y., Vrljic, M., Chen, L., Mudrakola, H. V., & Cui, B. (2010). Single-
molecule imaging of NGF axonal transport in microfluidic devices. Lab on a Chip, 10,
2566–2573.
Zhang, L., Yu, C., Vasquez, F. E., Galeva, N., Onyango, I., Swerdlow, R. H., et al. (2010).
Hyperglycemia alters the schwann cell mitochondrial proteome and decreases coupled
respiration in the absence of superoxide production. Journal of Proteome Research, 9,
458–471.
Zherebitskaya, E., Akude, E., Smith, D. R., & Fernyhough, P. (2009). Development of
selective axonopathy in adult sensory neurons isolated from diabetic rats: Role of
glucose-induced oxidative stress. Diabetes, 58, 1356–1364.
Zherebitskaya, E., Schapansky, J., Akude, E., Smith, D. R., Van der Ploeg, R.,
Solovyova, N., et al. (2012). Sensory neurons derived from diabetic rats have diminished
internal Ca2 + stores linked to impaired re-uptake by the endoplasmic reticulum. ASN
Neuro, 4. pii: e00072.
Zochodne, D. W. (2014). Mechanisms of diabetic neuron damage: Molecular pathways.
Handbook of Clinical Neurology, 126, 379–399.
CHAPTER SIX
Alternatives to the
Streptozotocin-Diabetic Rodent
M.A. Yorek1
Iowa City Health Care System, Iowa City, IA, United States
University of Iowa, Iowa City, IA, United States
Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States
1
Corresponding author: e-mail address: [email protected]
Contents
1. Introduction 90
2. Rodent Models of Obesity 91
2.1 High-Fat Fed Sprague-Dawley Rats and C57Bl6/J Mice 91
2.2 Zucker Rats 92
3. Rodent Models of Type 2 Diabetes 93
3.1 Zucker Diabetic Fatty Rats 93
3.2 Spontaneously Diabetic Torii Rat 94
3.3 Zucker Diabetic Sprague-Dawley Rat 94
3.4 Goto–Kakizaki Rat 95
3.5 BioBreeding Zucker Diabetic Rat 96
3.6 Otsuka Long-Evans Tokushima Fatty Rat 96
3.7 Ob/ob and db/db Mice 97
3.8 Tsumura Suzuki Obese Diabetes Mouse 98
3.9 Combined High-Fat Fed, Low-Dose Streptozotocin Models of
Type 2 Diabetes 98
3.10 Streptozotocin–Nicotinamide Rat 101
4. Rodent Models of Type 1 Diabetes 101
4.1 Spontaneously Hypertensive Rat 101
4.2 BioBreeding/Worcester Rat 102
4.3 Ins2Akita Mouse 102
4.4 Nonobese Diabetic Mouse 103
5. Other Animal Models 103
6. Conclusions 103
References 104
Abstract
The study of diabetic neuropathy has relied primarily on the use of streptozotocin-
treated rat and mouse models of type 1 diabetes. This chapter will review the creation
and use of other rodent models that have been developed in order to investigate the
contribution of factors besides insulin deficiency to the development and progression
#
International Review of Neurobiology, Volume 127 2016 Elsevier Inc. 89
ISSN 0074-7742 All rights reserved.
http://dx.doi.org/10.1016/bs.irn.2016.03.002
90 M.A. Yorek
1. INTRODUCTION
For years the standard animal model for the study of diabetic neurop-
athy has been the streptozotocin-treated rodent. In rats depending on the
species, age, and delivery, a single dose of streptozotocin ranging from
40 to 75 mg/kg is usually sufficient to destroy enough β cells to cause an
insulin-deficient form of diabetes (Rees & Alcolado, 2005; Tesch &
Allen, 2007). Such rats fail to gain weight and often lose weight unless
supported with a low-dose insulin treatment regime, which can allow such
animals to be maintained for extended periods of time in a hyperglycemic
state (Calcutt, 2004). In mice, the dose of streptozotocin required to create
a model of type 1 diabetes is generally higher than that used for rats, with
single doses ranging from 100 to 200 mg/kg (Rees & Alcolado, 2005;
Tesch & Allen, 2007). More recently, multiple low dosing of streptozo-
tocin has become the preferred method to induce an insulin-deficient
form of diabetes in mice (O’Brien, Sakowski, & Feldman, 2014; Rees &
Alcolado, 2005; Tesch & Allen, 2007). Using the latter approach creates a
type 1 diabetic mouse model that is more stable in regard to maintaining
their initial weight and will even gain weight compared to mice treated with
a single high dose of streptozotocin (personal observation). The potential for
high doses of streptozotocin to cause nonspecific effects on nerve and kidney
has for many years been a criticism of this type 1 diabetic model, even though
studies have shown that neurotoxicity is not the cause of slowing of nerve
conduction velocity or changes in thermal nociception in streptozotocin-
treated diabetic rats or mice (Davidson et al., 2009; Wiese, Matsushita,
Lowe, Stokes, & Yorek, 1996). Nonetheless, investigators seeking the
ultimate animal model to mimic the human development and progression
Alternatives to the Streptozotocin-Diabetic Rodent 91
Calcutt, Oltman, & Yorek, 2010; Groover et al., 2013; Nowicki, Kosacka,
Serke, Bluher, & Spanel-Borowski, 2012; O’Brien, Sakowski, et al., 2014;
Obrosova et al., 2007; Yorek et al., 2015). There have been many different
formulations of these diets, but in general they cause excess weight gain, raise
circulating lipid levels, and cause insulin resistance. Our studies have shown
that rats fed a high-fat (45% kcal) diet rapidly gained weight, became hyp-
erinsulinemic, and developed insulin resistance in 4–6 weeks (unpublished
data). However, fasting blood glucose and hemoglobin A1C levels were
not increased (Davidson et al., 2010; Davidson, Coppey, Dake, & Yorek,
2011; Davidson, Coppey, Kardon, & Yorek, 2014). In 12 weeks the rats
developed sensory neuropathy, as indicated by slowing of sensory nerve
conduction velocity and onset of thermal hypoalgesia (Davidson et al.,
2010; Davidson, Coppey, Dake, et al., 2011; Davidson, Coppey, Kardon,
et al., 2014). C57Bl6/J mice fed a high-fat diet also develop a prediabetes
phenotype including insulin resistance, impaired glucose utilization, fasting
hyperglycemia, and sensory neuropathy (Coppey et al., 2011; Yorek et al.,
2015). In our studies, diet-induced obese mice, like their rat counterparts,
had a normal motor nerve conduction velocity after 12 weeks of a high-fat
diet. Others have reported both motor and sensory neuropathy in C57Bl6/J
mice fed a high-fat diet for 16 weeks (Obrosova et al., 2007) or longer
(Anderson, King, Delbruck, & Jolivalt, 2014). Reasons for these differences
could include the type of high-fat diet used or study duration. It has been
reported that exercise can alleviate some of the neuropathic deficits associ-
ated with diet-induced obesity (Groover et al., 2013). C57Bl6/J mice have
been commonly used by investigators. However, it should be noted that
other strains of mice are also used and may respond differently to high-
fat-containing diets. For example, Swiss Webster mice develop insulin resis-
tance and cognitive impairment but do not gain weight or show indices of
peripheral neuropathy (Anderson et al., 2014). Human subjects determined
to have impaired glucose tolerance have also been reported to develop a
sensory neuropathy (Asghar et al., 2014; Kannan et al., 2014; Papanas,
Vinik, & Ziegler, 2011; Papanas & Ziegler, 2012; Smith & Singleton, 2008).
become noticeably obese by 3–5 weeks of age, and by 14 weeks of age their
body composition is over 40% lipid. Obese Zucker rats do not become
hyperglycemic, but are hyperlipemic, hypercholesterolemic, hyperinsuli-
nemic, and develop adipocyte hypertrophy and hyperplasia (Alderson
et al., 2003; Bray, 1977; Kurtz, Morris, & Pershadsingh, 1989). The Zucker
rat has also occasionally been used in investigations of obesity-associated
noninsulin-dependent diabetes mellitus. In our studies with this model,
we found that vascular and neural dysfunction developed at a slower rate
in obese Zucker rats than in the Zucker diabetic fatty (ZDF) rat model of
type 2 diabetes (see later). In both models, vascular impairment preceded
slowing of motor nerve conduction velocity, which occurs at 12–14 weeks
of age in ZDF rats and 32 weeks of age in obese Zucker rats (Oltman et al.,
2005).
the types 1 and 2 diabetic mouse models (Yorek et al., 2015). However,
motor and sensory nerve conduction velocity, thermal and mechanical sen-
sitivity, and intraepidermal nerve fiber density in the skin were all impacted
similarly in diet-induced obesity and types 1 and 2 diabetic mice. Loss of
corneal nerves in the subepithelial layer and penetrating the corneal epithe-
lium occurred more rapidly in the diet-induced obesity mice and type 2 dia-
betic mice compared to type 1 diabetic mouse. This suggests that
hyperglycemia is not the only factor contributing to nerve fiber loss in
the cornea and that loss of intraepidermal and corneal nerve fibers is medi-
ated by different factors and/or occurs at different rates.
Overall, these studies demonstrate that the high-fat fed low-dose
streptozotocin-treated rodents are a good animal model for preclinical
studies for discovery and evaluation of new treatments for diabetic neurop-
athy especially in relation to changes in nerve structure in the skin and
cornea.
(Gregory, Jolivalt, Goor, Mizisin, & Calcutt, 2012; Sanada et al., 2015) to
study the combined effects of diabetes and hypertension on peripheral
neuropathy.
6. CONCLUSIONS
In 1997, Dr. Tomlinson coauthored a review article titled “Does neu-
ropathy develop in animal models?” (Hounsom & Tomlinson, 1997). Inves-
tigators have been addressing this question for the past 18 years. In the
abstract of that paper the authors wrote in reference to diabetic neuropathy,
“currently the cornerstone of treatment lies with the maintenance of
euglycemia and development of effective treatments for diabetic neuropathy
is urgently needed.” The same statement could be made today. The only
accepted treatment for diabetic neuropathy remains good glycemic control,
104 M.A. Yorek
but it is now clear that this is ineffective, especially in patients with type 2
diabetes (Callaghan et al., 2012). Hounsom and Tomlinson further noted
that animal models have been developed to investigate the pathogenesis
of diabetic neuropathy and evaluate potential therapeutic agents. However,
no model is perfect and no one would suggest that diabetic rodents can
replicate the human condition fully. Since this article was written, animal
models, including those discussed earlier, continue to be used to investigate
and identify potential new treatments for diabetic neuropathy. However,
translation of findings to humans with diabetic peripheral neuropathy has
continued to fail. Many articles have been written to rationalize these fail-
ures. We know that diabetic neuropathy is a complex disease with mul-
tiple etiologies. It is my belief that animal models of diabetes and diabetic
neuropathy can still play a role in discovery of a treatment. However, this
treatment will likely be a combination therapy that will delay progression
and induce nerve repair when used in combination with glycemic control.
It should also be accepted that the preclinical efficacy of any treatment be
tested in several animal models utilizing multiple endpoints. A consensus
statement regarding the phenotyping of rodent models for diabetic peri-
pheral neuropathy was published following a joint meeting of the Diabetic
Neuropathy Study Group of EASD (Biessels et al., 2014). Following
these guidelines will help in standardizing studies coming from multiple
laboratories.
REFERENCES
Akash, M. S., Rehman, K., & Chen, S. (2013). Goto-Kakizaki rats: Its suitability as non-
obese diabetic animal model for spontaneous type 2 diabetes mellitus. Current Diabetes
Reviews, 9, 387–396.
Alderson, N. L., Chachich, M. E., Youssef, N. N., Beattie, R. J., Nachtigal, M.,
Thorpe, S. R., et al. (2003). The AGE inhibitor pyridoxamine inhibits lipemia and
development of renal and vascular disease in Zucker obese rats. Kidney International,
63, 2123–2133.
Anderson, N. J., King, M. R., Delbruck, L., & Jolivalt, C. G. (2014). Role of insulin signaling
impairment, adiponectin and dyslipidemia in peripheral and central neuropathy in mice.
Disease Models & Mechanisms, 7, 625–633.
Asghar, O., Petropoulos, I. N., Alam, U., Jones, W., Jeziorska, M., Marshall, A., et al. (2014).
Corneal confocal microscopy detects neuropathy in subjects with impaired glucose
tolerance. Diabetes Care, 37, 2643–2646.
Bansal, P., Paul, P., Mudgal, J., Nayak, P., Pannakal, S., Priyadarsini, K., et al. (2012).
Antidiabetic, antihyperlipidemic and antioxidant effects of the flavonoid rich fraction
of Pilea microphylla (L.) in high fat diet/streptozotocin-induced diabetes in mice. Exper-
imental and Toxicologic Pathology, 64, 651–658.
Alternatives to the Streptozotocin-Diabetic Rodent 105
Beltowski, J. (2012). Leptin and the regulation of endothelial function in physiological and
pathological conditions. Clinical and Experimental Pharmacology and Physiology, 39,
168–178.
Biessels, G. J., Bril, V., Calcutt, N. A., Cameron, N. E., Cotter, M. A., Dobrowsky, R., et al.
(2014). Phenotyping animal models of diabetic neuropathy: A consensus statement of
the diabetic neuropathy study group of the EASD (Neurodiab). Journal of the Peripheral
Nervous System, 19, 77–87.
Bour-Jordan, H., Thompson, H. L., Giampaolo, J. R., Davini, D., Rosenthal, W., &
Bluestone, J. A. (2013). Distinct genetic control of autoimmune neuropathy and diabetes
in the non-obese diabetic background. Journal of Autoimmunity, 45, 58–67.
Bray, G. A. (1977). The Zucker-fatty rat: A review. Federation Proceedings, 36, 148–153.
Calcutt, N. A. (2004). Modeling diabetic sensory neuropathy in rats. Methods in Molecular
Medicine, 99, 55–65.
Callaghan, B. C., Little, A. A., Feldman, E. L., & Hughes, R. A. (2012). Enhanced glucose
control for preventing and treating diabetic neuropathy. Cochrane Database of Systematic
Reviews, 6, CD007543.
Cameron, N. E., Cotter, M. A., Archibald, V., Dines, K. C., & Maxfield, E. K. (1994a).
Anti-oxidant and pro-oxidant effects on nerve conduction velocity, endoneurial blood
flow and oxygen tension in non-diabetic and streptozotocin-diabetic rats. Diabetologia,
37, 449–459.
Cameron, N. E., Cotter, M. A., Dines, K. C., & Love, A. (1992). Effects of aminoguanidine
on peripheral nerve function and polyol pathway metabolites in streptozotocin-diabetic
rats. Diabetologia, 35, 946–950.
Cameron, N. E., Cotter, M. A., Dines, K. C., Maxfield, E. K., Carey, F., & Mirrlees, D. J.
(1994b). Aldose reductase inhibition, nerve perfusion, oxygenation and function in
streptozotocin-diabetic rats: Dose-response considerations and independence from a
myo-inositol mechanism. Diabetologia, 37, 651–663.
Cameron, N. E., Cotter, M. A., & Low, P. A. (1991). Nerve blood flow in early experi-
mental diabetes in rats: Relation to conduction deficits. American Journal of Physiology.
Endocrinology and Metabolism, 261, E1–E8.
Cho, Y. R., Lim, J. H., Kim, M. Y., Kim, T. W., Hong, B. Y., Kim, Y. S., et al. (2014).
Therapeutic effects of fenofibrate on diabetic peripheral neuropathy by improving
endothelial and neural survival in db/db mice. PLoS One, 9, e83204.
Cohen, R. A., Tesfamariam, B., Weisbrod, R. M., & Zitnay, K. M. (1990). Adrenergic
denervation in rabbits with diabetes mellitus. American Journal of Physiology. Heart and
Circulatory Physiology, 259, H55–H61.
Coppey, L. J., Davidson, E. P., Dunlap, J. A., Lund, D. D., & Yorek, M. A. (2000). Slowing
of motor nerve conduction velocity in streptozotocin-induced diabetic rats is preceded
by impaired vasodilation in arterioles that overlie the sciatic nerve. International Journal of
Experimental Diabetes Research, 1, 131–143.
Coppey, L., Davidson, E., Lu, B., Gerard, C., & Yorek, M. (2011). Vasopeptidase inhibitor
ilepatril (AVE7688) prevents obesity- and diabetes-induced neuropathy in C57Bl/6J
mice. Neuropharmacology, 60, 259–266.
Coppey, L. J., Gellett, J. S., Davidson, E. P., Dunlap, J. A., & Yorek, M. A. (2002). Changes
in endoneurial blood flow, motor nerve conduction velocity and vascular relaxation of
epineurial arterioles of the sciatic nerve in ZDF-obese diabetic rats. Diabetes/Metabolism
Research and Reviews, 18, 49–56.
Cotter, M. A., Dines, K. C., & Cameron, N. E. (1993). Prevention and reversal of motor and
sensory peripheral nerve conduction abnormalities in streptozotocin-diabetic rats by the
prostacyclin analogue iloprost. Naunyn Schmiedeberg’s Archives of Pharmacology, 347,
534–540.
106 M.A. Yorek
Davidson, E. P., Coppey, L. J., Calcutt, N. A., Oltman, C. L., & Yorek, M. A. (2010).
Diet-induced obesity in Sprague-Dawley rats causes microvascular and neural dysfunc-
tion. Diabetes/Metabolism Research and Reviews, 26, 306–318.
Davidson, E. P., Coppey, L. J., Dake, B., & Yorek, M. A. (2011a). Effect of treatment of
Sprague Dawley rats with AVE7688, enalapril, or candoxatril on diet-induced obesity.
Journal of Obesity, 2011. 686952.
Davidson, E. P., Coppey, L. J., Holmes, A., Dake, B., & Yorek, M. A. (2011b). Effect of
treatment of high fat fed/low dose streptozotocin-diabetic rats with Ilepatril on vascular
and neural complications. European Journal of Pharmacology, 668, 497–506.
Davidson, E. P., Coppey, L. J., Holmes, A., Lupachyk, S., Dake, B. L., Oltman, C. L., et al.
(2014a). Characterization of diabetic neuropathy in the Zucker diabetic Sprague-Dawley
rat: A new animal model for type 2 diabetes. Journal of Diabetes Research, 2014. 714273.
Davidson, E. P., Coppey, L. J., Kardon, R. H., & Yorek, M. A. (2014b). Differences and
similarities in development of corneal nerve damage and peripheral neuropathy and
in diet-induced obesity and type 2 diabetic rats. Investigative Ophthalmology and Visual
Science, 55, 1222–1230.
Davidson, E., Coppey, L., Lu, B., Arballo, V., Calcutt, N. A., Gerard, C., et al. (2009). The
roles of streptozotocin neurotoxicity and neural endopeptidase in murine experimental
diabetic neuropathy. Experimental Diabetes Research, 2009. 431980.
Davidson, E. P., Kleinschmidt, T. L., Oltman, C. L., Lund, D. D., & Yorek, M. A. (2007).
Treatment of streptozotocin-induced diabetic rats with AVE7688, a vasopeptidase
inhibitor: Effect on vascular and neural disease. Diabetes, 56, 355–362.
Davis, J. E., Cain, J., Banz, W. J., & Peterson, R. G. (2013). Age-related differences in
response to high-fat feeding on adipose tissue and metabolic profile in ZDSD rats. ISRN
Obesity, 2013. 584547.
de Preux Charles, A. S., Verdier, V., Zenker, J., Peter, B., Medard, J. J., Kuntzer, T., et al.
(2010). Global transcriptional programs in peripheral nerve endoneurium and DRG are
resistant to the onset of type 1 diabetic neuropathy in Ins2 mice. PLoS One, 5, e10832.
Ding, S. Y., Shen, Z. F., Chen, Y. T., Sun, S. J., Liu, Q., & Xie, M. Z. (2005). Pioglitazone
can ameliorate insulin resistance in low-dose streptozotocin and high sucrose-fat diet
induced obese rats. Acta Pharmacologica Sinica, 26, 575–580.
Drel, V. R., Mashtalir, N., Ilnytska, O., Shin, J., Li, F., Lyzogubov, V. V., et al. (2006).
The leptin-deficient (ob/ob) mouse: A new animal model of peripheral neuropathy
of type 2 diabetes and obesity. Diabetes, 55, 3335–3343.
Drel, V. R., Pacher, P., Stavniichuk, R., Xu, W., Zhang, J., Kuchmerovska, T. M., et al.
(2011). Poly(ADP-ribose)polymerase inhibition counteracts renal hypertrophy and mul-
tiple manifestations of peripheral neuropathy in diabetic Akita mice. International Journal
of Molecular Medicine, 28, 629–635.
Estrella, J. S., Nelson, R. N., Sturges, B. K., Vernau, K. M., Williams, D. C.,
LeCouteur, R. A., et al. (2008). Endoneurial microvascular pathology in feline diabetic
neuropathy. Microvascular Research, 75, 403–410.
Figueroa-Romero, C., Sadidi, M., & Feldman, E. L. (2008). Mechanisms of disease: The
oxidative stress theory of diabetic neuropathy. Review in Endocrine and Metabolic Disorders,
9, 301–314.
Gade, W., Schmit, J., Collins, M., & Gade, J. (2010). Beyond obesity: The diagnosis and
pathophysiology of metabolic syndrome. Clinical Laboratory Science, 23, 51–61.
Gaikwad, A. B., Viswanad, B., & Ramarao, P. (2007). PPAR gamma agonists partially
restores hyperglycemia induced aggravation of vascular dysfunction to angiotensin II
in thoracic aorta isolated from rats with insulin resistance. Pharmacological Research, 55,
400–407.
Gao, F., & Zheng, Z. M. (2014). Animal models of diabetic neuropathic pain. Experimental
and Clinical Endocrinology and Diabetes, 122, 100–106.
Alternatives to the Streptozotocin-Diabetic Rodent 107
Gregory, J. A., Jolivalt, C. G., Goor, J., Mizisin, A. P., & Calcutt, N. A. (2012).
Hypertension-induced peripheral neuropathy and the combined effects of hypertension
and diabetes on nerve structure and function in rats. Acta Neuropathologica, 124, 561–573.
Groover, A. L., Ryals, J. M., Guilford, B. L., Wilson, N. M., Christianson, J. A., &
Wright, D. E. (2013). Exercise-mediated improvements in painful neuropathy associated
with prediabetes in mice. Pain, 154, 2658–2667.
Gross, V., Tank, J., Partke, H. J., Plehm, R., Diedrich, A., da Costa Goncalves, A. C., et al.
(2008). Cardiovascular autonomic regulation in non-obese diabetic (NOD) mice.
Autonomic Neuroscience: Basic and Clinical, 138, 108–113.
Grote, C. W., Groover, A. L., Ryals, J. M., Geiger, P. C., Feldman, E. L., & Wright, D. E.
(2013). Peripheral nervous system insulin resistance in ob/ob mice. Acta Neuropathologica
Communications, 1, 15. http://dx.doi.org/10.1186/2051-5960-1-15.
Hounsom, L., & Tomlinson, D. R. (1997). Does neuropathy develop in animal models?
Clinical Neuroscience, 4, 380–389.
Islam, M. S. (2013). Animal models of diabetic neuropathy: Progress since 1960s. Journal of
Diabetes Research, 2013. 149452.
Jun, J. Y., Ma, Z., & Segar, L. (2011). Spontaneously diabetic Ins2(+/Akita): ApoE-deficient
mice exhibit exaggerated hypercholesterolemia and atherosclerosis. American Journal of
Physiology. Endocrinology and Metabolism, 301, E145–E154.
Kamenov, Z., Higashino, H., Todorova, M., Kajimoto, N., & Suzuki, A. (2006). Physio-
logical characteristics of diabetic neuropathy in sucrose-fed Otsuka Long-Evans
Tokushima fatty rats. Methods and Findings in Experimental and Clinical Pharmacology,
28, 13–18.
Kamiya, H., Murakawa, Y., Zhang, W., & Sima, A. A. (2005). Unmyelinated fiber sensory
neuropathy differs in type 1 and type 2 diabetes. Diabetes/Metabolism Research and Reviews,
21, 448–458.
Kamiya, H., Zhang, W., & Sima, A. A. (2004). C-peptide prevents nociceptive sensory
neuropathy in type 1 diabetes. Annals of Neurology, 56, 827–835.
Kamiya, H., Zhang, W., & Sima, A. A. (2009). Dynamic changes of neuroskeletal proteins in
DRGs underlie impaired axonal maturation and progressive axonal degeneration in type
1 diabetes. Experimental Diabetes Research, 2009. 793281.
Kannan, M. A., Sarva, S., Kandadai, R. M., Paturi, V. R., Jabeen, S. A., & Borgohain, R.
(2014). Prevalence of neuropathy in patients with impaired glucose tolerance using
various electrophysiological tests. Neurology India, 62, 656–661.
Kaur, J. (2014). A comprehensive review on metabolic syndrome. Cardiology Research and
Practice, 2014. 943162.
Kava, R., Greenwood, M. R. C., & Johnson, P. R. (1990). Zucker (fa/fa) rat. ILAR Journal,
32, 4–8.
Kawada, T., Miyata, S., Shimada, T., Sanzen, Y., Ito, M., Hemmi, C., et al. (2010). A study
of cardiovascular function in Tsumura Suzuki obese diabetes, a new model mouse of type
2 diabetes. Biological and Pharmaceutical Bulletin, 33, 998–1003.
Kemmochi, Y., Fukui, K., Maki, M., Kimura, S., Ishii, Y., Sasase, T., et al. (2013). Metabolic
disorders and diabetic complications in spontaneously diabetic Torii Lepr (fa) rat: A new
obese type 2 diabetic model. Journal of Diabetes Research, 2013. 948257.
Kim, H., Kim, J. J., & Yoon, Y. S. (2012). Emerging therapy for diabetic neuropathy: Cell
therapy targeting vessels and nerves. Endocrine, Metabolic and Immune Disorders Drug
Targets, 12, 168–178.
Kurtz, T. W., Morris, R. C., & Pershadsingh, H. A. (1989). The Zucker fatty rat as a genetic
model of obesity and hypertension. Hypertension, 13, 896–901.
Li, F., Abatan, O. I., Kim, H., Burnett, D., Larkin, D., Obrosova, I. G., et al. (2006). Taurine
reverses neurological and neurovascular deficits in Zucker diabetic fatty rats. Neurobiology
of Disease, 22, 669–676.
108 M.A. Yorek
Lin, Y., Hu, G., Li, K., Zhao, Y., Wei, J., & Zhen, Y. (2015). The protection of Rhein
lysinate to liver in diabetic mice induced by high-fat diet and streptozotocin. Archives
of Pharmacal Research, 38, 885–892.
Lirk, P., Flatz, M., Haller, I., Hausott, B., Blumenthal, S., Stevens, M. F., et al. (2012).
In Zucker diabetic fatty rats, subclinical diabetic neuropathy increases in vivo lidocaine
block duration but not in vitro neurotoxicity. Regional Anesthesia and Pain Medicine, 37,
601–606.
Lirk, P., Verhamme, C., Boeckh, R., Stevens, M. F., ten Hoope, W., Gerner, P., et al. (2015).
Effects of early and late diabetic neuropathy on sciatic nerve block duration and neuro-
toxicity in Zucker diabetic fatty rats. British Journal of Anesthesia, 114, 319–326.
Lizuka, S., Suzuki, W., Tabuchi, M., Nagata, M., Imamura, S., Kobayashi, Y., et al. (2005).
Diabetic complications in a new animal model (TSOD mouse) of spontaneous NIDDM
with obesity. Experimental Animals, 54, 71–83.
Lupachyk, S., Watcho, P., Hasanova, N., Julius, U., & Obrosova, I. G. (2012). Triglyceride,
nonesterified fatty acids, and prediabetic neuropathy: Role for oxidative-nitrosative
stress. Free Radical Biology and Medicine, 52, 1255–1263.
Lupachyk, S., Watcho, P., Obrosov, A. A., Stavniichuk, R., & Obrosova, I. G. (2013).
Endoplasmic reticulum stress contributes to prediabetic peripheral neuropathy. Experi-
mental Neurology, 247, 342–348.
Lv, L., Wu, S., Wang, G., Zhang, J., Pang, J., Liu, Z., et al. (2010). Effect of astragaloside IV
on hepatic glucose-regulating enzymes in diabetic mice induced by a high-fat diet and
streptozotocin. Phytotherapy Research, 24, 219–224.
Mali, V., Ning, R., Chen, J., Yang, X., Xu, J., & Palaniyandi, S. (2014). Impairment of
aldehyde dehydrogenase-2 by 4-hydroxy-2-nonenal adduct formation and cardiomyo-
cyte hypertrophy in mice fed a high-fat diet and injected with low-dose streptozotocin.
Experimental Biology and Medicine, 239, 610–618.
Mesangeau, D., Laude, D., & Elghozi, J. L. (2000). Early detection of cardiovascular
autonomic neuropathy in diabetic pigs using blood pressure and heart rate variability.
Cardiovascular Research, 45, 889–899.
Mizisin, A. P., Nelson, R. W., Sturges, B. K., Vernau, K. M., Lecouteur, R. A.,
Williams, D. C., et al. (2007). Comparable myelinated nerve pathology in feline and
human diabetes mellitus. Acta Neuropathologica, 113, 431–442.
Morgan, M. J., Vite, C. H., Radhakrishnan, A., & Hess, R. S. (2008). Clinical peripheral
neuropathy associated with diabetes mellitus in 3 dogs. The Canadian Veterinary Journal,
49, 583–586.
Murakawa, Y., Zhang, W., Pierson, C. R., Brismar, T., Ostenson, C. G., Efendic, S., et al.
(2002). Impaired glucose tolerance and insulinopenia in the GK-rat causes peripheral
neuropathy. Diabetes/Metabolism Research and Reviews, 18, 473–483.
Nakamura, J., Hamada, Y., Sakakibara, F., Hara, T., Wakao, T., Mori, K., et al. (2001).
Physiological and morphometric analyses of neuropathy in sucrose-fed OLETF rats.
Diabetes Research and Clinical Practice, 51, 9–20.
Negi, G., Kumar, A., Kaundal, R. K., Gulati, A., & Sharma, S. S. (2010). Functional and
biochemical evidence indicating beneficial effect of melatonin and nicotinamide alone
and in combination in experimental diabetic neuropathy. Neuropharmacology, 58,
585–592.
Norido, F., Canella, R., Zanoni, R., & Gorio, A. (1984). Development of diabetic neurop-
athy in the C57BL/Ks (db/db) mouse and its treatment with gangliosides. Experimental
Neurology, 83, 221–232.
Nowicki, M., Kosacka, J., Serke, H., Bluher, M., & Spanel-Borowski, K. (2012). Altered
sciatic nerve fiber morphology and endoneural microvessels in mouse models relevant
for obesity, peripheral diabetic polyneuropathy, and the metabolic syndrome. Journal
of Neuroscience Research, 90, 122–131.
Alternatives to the Streptozotocin-Diabetic Rodent 109
Nukada, H., & Dyck, P. J. (1984). Microsphere embolization of nerve capillaries and fiber
degeneration. The American Journal of Pathology, 115, 275–287.
O’Brien, P. D., Hur, J., Hayes, J. M., Backus, C., Sakowski, S. A., & Feldman, E. L. (2014).
BTBR ob/ob mice as a novel diabetic neuropathy model: Neurological characterization
and gene expression analyses. Neurobiology of Disease, 73C, 348–355.
O’Brien, P. D., Sakowski, S. A., & Feldman, E. L. (2014). Mouse models of diabetic
neuropathy. ILAR Journal, 54, 259–272.
Obrosova, I. G., Ilnytska, O., Lyzogubov, V. V., Pavlov, I. A., Mashtalir, N., Nadler, J. L.,
et al. (2007). High-fat diet induced neuropathy of pre-diabetes and obesity: Effects of
“healthy” diet and aldose reductase inhibition. Diabetes, 56, 2598–2608.
Obrosova, I. G., Mabley, J. G., Zsengeller, Z., Charniauskaya, T., Abatan, O. I.,
Groves, J. T., et al. (2005). Role for nitrosative stress in diabetic neuropathy: Evidence
from studies with a peroxynitrite decomposition catalyst. FASEB Journal, 19, 401–403.
Oltman, C. L., Coppey, L. J., Gellett, J. S., Davidson, E. P., Lund, D. D., & Yorek, M. A.
(2005). Progression of vascular and neural dysfunction in sciatic nerves of Zucker diabetic
fatty and Zucker rats. American Journal of Physiology. Endocrinology and Metabolism, 289,
E113–E122.
Oltman, C. L., Davidson, E. P., Coppey, L. J., Kleinschmidt, T. L., Lund, D. D.,
Adebara, E. T., et al. (2008). Vascular and neural dysfunction in Zucker diabetic fatty
rats: A difficult condition to reverse. Diabetes, Obesity and Metabolism, 10, 64–74.
Oltman, C. L., Davidson, E. P., Coppey, L. J., Kleinschmidt, T. L., & Yorek, M. A. (2009).
Treatment of Zucker diabetic fatty rats with AVE7688 improves vascular and neural
dysfunction. Diabetes, Obesity and Metabolism, 11, 223–233.
Otto, K. J., Wyse, B. D., Cabot, P. J., & Smith, M. T. (2011). Longitudinal study of painful
diabetic neuropathy in the Zucker diabetic fatty rat model of type 2 diabetes: Impaired
basal G-protein activity appears to underpin marked morphine hyposensitivity at
6 months. Pain Medicine, 12, 437–450.
Pande, M., Hur, J., Hong, Y., Backus, C., Hayes, J. M., Oh, S. S., et al. (2011). Transcrip-
tional profiling of diabetic neuropathy in the BKS db/db mouse: A model of type 2
diabetes. Diabetes, 60, 1981–1989.
Papanas, N., Vinik, A. I., & Ziegler, D. (2011). Neuropathy in prediabetes: Does the clock
start ticking early? Nature Reviews. Endocrinology, 7, 682–690.
Papanas, N., & Ziegler, D. (2012). Prediabetic neuropathy: Does it exist? Current Diabetes
Reports, 12, 376–383.
Peterson, R. G., Jackson, C. V., Zimmeran, K., de Winter, W., Huebert, N., &
Hansen, M. K. (2015). Characterization of the ZDSD rat: A translational model for
the study of metabolic syndrome and type 2 diabetes. Journal of Diabetes Research,
2015. 487816.
Peterson, R. G., Shaw, W. N., Neel, M. A., Little, L. A., & Eichberg, J. (1990).
Zuckerdiabetic fatty rat as a model for non-insulin-dependent diabetes mellitus. ILAR
Journal, 32, 16–19.
Pop-Busui, R., Sima, A., & Stevens, M. (2006). Diabetic neuropathy and oxidative stress.
Diabetes/Metabolism Research and Reviews, 22, 257–273.
Portha, B., Giroix, M. H., Tourrel-Cuzin, C., Le-Stunff, H., & Movassat, J. (2012). The GK
rat: A prototype for the study of non-overweight type 2 diabetes. Methods in Molecular
Biology, 933, 125–159.
Quattrini, C., Tavakoli, M., Jeziorska, M., Kallinikos, P., Tesfaye, S., Finnigan, J., et al.
(2007). Surrogate markers of small fiber damage in human diabetic neuropathy.
Diabetes, 56, 2148–2154.
Reed, M. J., Meszaros, K., Entes, L. J., Claypool, M. D., Pinkett, J. G., Gadbois, T. M., et al.
(2000). A new rat model of type 2 diabetes: The fat-fed, streptozotocin-treated rat.
Metabolism, 49, 1390–1394.
110 M.A. Yorek
Rees, D. A., & Alcolado, J. C. (2005). Animal models of diabetes mellitus. Diabetic Medicine,
22, 359–370.
Reinwald, S., Peterson, R. G., Allen, M. R., & Burr, D. B. (2009). Skeletal changes
associated with the onset of type 2 diabetes in the ZDF and ZDSD rodent models.
American Journal of Physiology. Endocrinology and Metabolism, 296, E765–E774.
Ricci, R., & Bevilacqua, F. (2012). The potential role of leptin and adiponectin in obesity:
A comparative review. Veterinary Journal, 191, 292–298.
Robertson, D. M., & Sima, A. A. (1980). Diabetic neuropathy in the mutant mouse
[C57BL/ks(db/db)]: A morphometric study. Diabetes, 29, 60–67.
Sanada, L. S., Tavares, M. R., Sato, K. L., Ferreira Rda, S., Neubern, M. C., Castania, J. A.,
et al. (2015). Association of chronic diabetes and hypertension in sural nerve morphom-
etry: An experimental study. Diabetology and Metabolic Syndrome, 7, 9.
Sasase, T., Ohta, T., Masuyama, T., Yokoi, N., Kakehashi, A., & Shinohara, M. (2013). The
spontaneously diabetic Torii rat: An animal model of nonobese type 2 diabetes with
severe diabetic complications. Journal of Diabetes Research, 2013. 976209.
Sasase, T., Yokoi, N., Pezzolesi, M. G., & Shinohara, M. (2015). Animal models of diabetes
and metabolic disease 2014. Journal of Diabetes Research, 2015. 571809.
Sharma, A. K., Sharma, A., Kumari, R., Kishore, K., Sharma, D., Srinivasan, B. P., et al.
(2012). Sitagliptin, sitagliptin and metformin, or sitagliptin and amitriptyline attenuate
streptozotocin-nicotinamide induced diabetic neuropathy in rats. Journal of Biomedical
Research, 26, 200–210.
Shi, T. J., Zhang, M. D., Zeberg, H., Nilsson, J., Grunler, J., Liu, S. X., et al. (2013).
Coenzyme Q10 prevents peripheral neuropathy and attenuates neuron loss in the
db-/db- mouse, a type 2 diabetes model. Proceedings of the National Academy of Sciences
of the United States of America, 110, 690–695.
Shinohara, M., Masuyama, T., Shoda, T., Takahashi, T., Katsuda, Y., Komeda, K., et al.
(2000). A new spontaneously diabetic non-obese Torii rat strain with severe ocular
complications. International Journal of Experimental Diabetes Research, 1, 89–100.
Sima, A. A. (2006). Pathological mechanisms involved in diabetic neuropathy: Can we slow
the process? Current Opinion in Investigational Drugs, 7, 324–337.
Sima, A. A., & Robertson, D. M. (1978). Peripheral neuropathy in mutant diabetic mouse
[C57BL/Ks (db/db)]. Acta Neuropathologica, 41, 85–89.
Sima, A. A., Zhang, W., Sugimoto, K., Henry, D., Li, Z., Wahren, J., et al. (2001).
C-peptide prevents and improves chronic type 1 diabetic polyneuropathy in the BB/
Wor rat. Diabetologia, 44, 889–897.
Sima, A. A., Zhang, W., Xu, G., Sugimoto, K., Guberski, D., & Yorek, M. A. (2000).
A comparison of diabetic polyneuropathy in type II diabetic BBZDR/Wor rats and
in type I diabetic BB/Wor rats. Diabetologia, 43, 786–793.
Skovso, S. (2014). Modeling type 2 diabetes in rats using high fat diet and streptozotocin.
Journal of Diabetes Investigation, 5, 349–358.
Smith, A. G., & Singleton, J. R. (2008). Impaired glucose tolerance and neuropathy.
The Neurologist, 14, 23–29.
Srinivasan, K., Viswanad, B., Asrat, L., Kaul, C. L., & Ramarao, P. (2005). Combination of
high-fat diet-fed and low-dose streptozotocin-treated rat: A model for type 2 diabetes
and pharmacological screening. Pharmacological Research, 52, 313–320.
Stevens, M. J., Li, F., Drel, V. R., Abatan, O. I., Kim, H., Burnett, D., et al. (2007).
Nicotinamide reverses neurological and neurovascular deficits in streptozotocin diabetic
rats. The Journal of Pharmacology and Experimental Therapeutics, 320, 458–464.
Stevens, M. J., Zhang, W., Li, F., & Sima, A. A. (2004). C-peptide corrects endoneurial
blood flow but not oxidative stress in type 1 BB/Wor rats. American Journal of Physiology.
Endocrinology and Metabolism, 287, E497–E505.
Alternatives to the Streptozotocin-Diabetic Rodent 111
Sullivan, K. A., Hayes, J. M., Wiggin, T. D., Backus, C., Su Oh, S., Lentz, S. I., et al. (2007).
Mouse models of diabetic neuropathy. Neurobiology of Disease, 28, 276–285.
Szkudelski, T. (2012). Streptozotocin-nicotinamide-induced diabetes in the rat. Character-
istics of the experimental model. Experimental Biology and Medicine, 237, 481–490.
Taborsky, G. J., Jr., Mei, Q., Hackney, D. J., Figlewicz, D. P., LeBoeuf, R., & Mundinger, T. O.
(2009). Loss of islet sympathetic nerves and impairment of glucagon secretion in the
NOD mouse: Relationship to invasive insulitis. Diabetologia, 52, 2602–2611.
Terata, K., Coppey, L. J., Davidson, E. P., Dunlap, J. A., Gutterman, D., & Yorek, M. A.
(1999). Acetylcholine-induced arteriolar dilation is reduced in streptozotocin-
induced diabetic rats with motor nerve dysfunction. British Journal of Pharmacology,
128, 837–843.
Tesch, G. H., & Allen, T. J. (2007). Rodent models of streptozotocin-induced diabetic
nephropathy. Nephrology, 12, 261–266.
Tirabassi, R. S., Flanagan, J. F., Wu, T., Kislauskis, E. H., Birckbichler, P. J., &
Guberski, D. L. (2004). The BBZDR/Wor rat model for investigating the complications
of type 2 diabetes mellitus. ILAR Journal, 45, 292–302.
Ueta, K., Ishihara, T., Matsumoto, Y., Oku, A., Nawano, M., Fujita, T., et al. (2005).
Long-term treatment with the Na+-glucose cotransporter inhibitor T-1095 causes
sustained improvement in hyperglycemia and prevents diabetic neuropathy in Goto-
Kakizaki rats. Life Sciences, 76, 2655–2668.
Ullevig, S., Zhao, Q., Zamora, D., & Asmis, R. (2011). Ursolic acid protects diabetic mice
against monocyte dysfunction and accelerated atherosclerosis. Atherosclerosis, 219,
409–416.
Vareniuk, I., Pavlov, I. A., Drel, V. R., Lyzogubov, V. V., Ilnystska, O., Bell, S. R., et al.
(2007). Nitrosative stress and peripheral diabetic neuropathy in leptin-deficient (ob/ob)
mice. Experimental Neurology, 205, 425–436.
Wada, R., Koyama, M., Mizukami, H., Odaka, H., Ikeda, H., & Yagihashi, S. (1999). Effects
of long-term treatment with alpha-glucosidase inhibitor on the peripheral nerve function
and structure in Goto-Kakizaki rats: A genetic model for type 2 diabetes. Diabetes/
Metabolism Research and Reviews, 15, 332–337.
Wang, F., Gao, N., Yin, J., & Yu, F. S. (2012). Reduced innervation and delayed re-in-
nervation after epithelial wounding in type 2 diabetic Goto-Kakizaki rats. The American
Journal of Pathology, 181, 2058–2066.
Wang, Z., Hsu, C., Lin, H., & Chen, J. (2014). Antidiabetic effects of Carassius auratus
complex formula in high fat diet combined streptozotocin-induced diabetic mice.
Evidence Based Complementary and Alternative Medicine, 2014. 628473.
Wang, B., Li, Y., Liu, X., Liu, S., & Sun, C. (2011). Effect of the duration of high-fat diet
and the dosage of streptozotocin on establishing experimental animal model of type 2
diabetes mellitus. Wei Sheng Yan Jiu, 40, 99–102.
Wang, C., Li, J., Lv, X., Zhang, M., Song, Y., Chen, L., et al. (2009). Ameliorative effect of
berberine on endothelial dysfunction in diabetic rats induced by high-fat diet and
streptozotocin. European Journal of Pharmacology, 620, 131–137.
Whalen, B. J., Mordes, J. P., & Rossini, A. A. (2001). The BB rat as a model of human
insulin-dependent diabetes mellitus. Current Protocols in Immunology (Suppl. 19).
Chapter 15, 15.3.1–15.3.15.
Wiese, T. J., Matsushita, K., Lowe, W. L., Jr., Stokes, J. B., & Yorek, M. A. (1996).
Localization and regulation of renal Na+/myo-inositol cotransporter in diabetic rats.
Kidney International, 50, 1202–1211.
Xue, J., Ding, W., & Liu, Y. (2010). Anti-diabetic effects of emodin involved in the
activation of PPAR gamma on high-fat diet-fed and low dose of streptozotocin-induced
diabetic mice. Fitoterapia, 81, 173–177.
112 M.A. Yorek
Yaguchi, M., Nagashima, K., Izumi, T., & Okamoto, K. (2003). Neuropathological study of
C57BL/6Akita mouse, type 2 diabetic model: Enhanced expression of alphaB-crystallin
in oligodendrocytes. Neuropathology, 23, 44–50.
Yamaguchi, T., Sasase, T., Mera, Y., Tomimoto, D., Tadaki, H., Kemmochi, Y., et al.
(2012). Diabetic peripheral neuropathy in spontaneously diabetic Torii-Lepr (fa)
(SDT fatty) rats. The Journal of Veterinary Medical Science, 74, 1669–1673.
Yang, B., & Chon, K. H. (2011). Assessment of diabetic cardiac autonomic neuropathy
in type 1 diabetic mice. In Annual international conference IEEE engineering medicine biology
society (pp. 6560–6563).
Yorek, M. S., Obrosov, A., Shevalye, H., Holmes, A., Harper, M. M., Kardon, R. H., et al.
(2015). Effect of diet-induced obesity or type 1 or type 2 diabetes on corneal nerves and
peripheral neuropathy in C57Bl/6J mice. Journal of the Peripheral Nervous System, 20,
24–31.
Zhang, M., Lv, X. Y., Li, J., Xu, Z. G., & Chen, L. (2008). The characterization of high-fat
diet and multiple low-dose streptozotocin induced type 2 diabetes rat model. Experimen-
tal Diabetes Research, 2008. 704045.
Zhang, W., Yorek, M., Pierson, C. R., Murakawa, Y., Breidenbach, A., & Sima, A. A.
(2001). Human C-peptide dose dependently prevents early neuropathy in the BB/
Wor rat. International Journal of Experimental Diabetes Research, 2, 187–193.
Zhu, N., Liu, B., Luo, W., Zhang, Y., Li, H., Li, S., et al. (2014). Vasoconstrictor role of
cyclooxygenase-1-mediated prostacyclin synthesis in non-insulin-dependent diabetic
mice induced by high-fat diet and streptozotocin. American Journal of Physiology. Heart
and Circulatory Physiology, 307, H319–H327.
Zhu, C. F., Peng, H. B., Liu, G. Q., Zhang, F., & Li, Y. (2010). Beneficial effects of
oligopeptides from marine salmon skin in a rat model of type 2 diabetes. Nutrition,
26, 1014–1020.
CHAPTER SEVEN
#
International Review of Neurobiology, Volume 127 2016 Elsevier Inc. 115
ISSN 0074-7742 All rights reserved.
http://dx.doi.org/10.1016/bs.irn.2016.03.012
116 P. Fernyhough and N.A. Calcutt
is the true site of action. AR has been localized to Schwann cells of myelin-
ated fibers, epineurial endothelial cells, and axons of sympathetic neurons,
but a clear explanation as to how excessive polyol pathway activity drives
distal axonal damage in small sensory fibers remains to be fully developed
(Jiang, Calcutt, Ramos, & Mizisin, 2006). Yagihashi revisits some of these
issues in his contribution entitled “Glucotoxic Mechanisms and Related
Therapeutic Approaches.”
The clinical failure of ARIs has not, in our opinion, denigrated the
central idea that excessive glucose flux through the polyol pathway is a key
pathogenic process in diabetic neuropathy. As ARI-related research has
stalled, various research groups have explored putative downstream conse-
quences of hyperglycemia, primarily in endothelial cell cultures, to study
the impact of excessive glucose metabolism and polyol pathway flux in
augmenting ischemia and oxidative stress (Figueroa-Romero, Sadidi, &
Feldman, 2008; Nishikawa, Edelstein, & Brownlee, 2000). The role of ische-
mia due to hyperglycemia-induced impaired nerve blood flow remains
a major source of controversy. Diabetic neuropathy is classified by many as
a microvascular disease. However, the pathology of nerve damage in diabetes
does not reflect an ischemic process. For example, in ischemic nerve damage
large fibers are preferentially targeted, which does not appear to be the case in
diabetic neuropathy (Fujimura, Lacroix, & Said, 1991). In the contribution
“Sensory Neurodegeneration in Diabetes: Beyond Glucotoxicity” by
Zochodne, the inconsistencies in the vascular hypothesis as a cogent explana-
tion of nerve damage in diabetes are briefly discussed.
Oxidative stress clearly occurs in the nerve of diabetic rodents and
diverse antioxidants are protective in various animal models of the disease
(Figueroa-Romero et al., 2008). Unfortunately, clinical trials of antioxidants
in diabetic neuropathy have been underwhelming, although some support
has been gained from use of agents such as alpha lipoic acid that includes
antioxidant activity among its properties (Ziegler et al., 2011). The caveats
separating successful preclinical studies from demonstrable clinical efficacy,
as discussed above for ARI, may equally apply to antioxidants with the addi-
tional problem that the financial incentive for drug development in this area
is limited. A much more cohesive understanding of the multiple sources
of ROS in nerve tissue is also required. For example, endothelial cells
may indeed produce excessive ROS via high glucose flux-driven aberrant
overactivity of the proximal aspect of the mitochondrial electron transport
chain (Nishikawa, Edelstein, Du, et al., 2000). However, it is becoming rec-
ognized that neurons and glia have distinct energy demands and produce
History and Controversies of the Pathogenesis of Diabetic Neuropathy 117
REFERENCES
Akude, E., Zherebitskaya, E., Chowdhury, S. K., Smith, D. R., Dobrowsky, R. T., &
Fernyhough, P. (2011). Diminished superoxide generation is associated with respiratory
chain dysfunction and changes in the mitochondrial proteome of sensory neurons from
diabetic rats. Diabetes, 60, 288–297.
Apfel, S. C. (2002). Nerve growth factor for the treatment of diabetic neuropathy: What
went wrong, what went right, and what does the future hold? International Review of
Neurobiology, 50, 393–413.
Bierhaus, A., Fleming, T., Stoyanov, S., Leffler, A., Babes, A., Neacsu, C., et al. (2012).
Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes
hyperalgesia in diabetic neuropathy. Nature Medicine, 18, 926–933.
Brewster, W. J., Fernyhough, P., Diemel, L. T., Mohiuddin, L., & Tomlinson, D. R. (1994).
Diabetic neuropathy, nerve growth factor and other neurotrophic factors. Trends in
Neurosciences, 17, 321–325.
Calcutt, N. A., Jolivalt, C. G., & Fernyhough, P. (2008). Growth factors as therapeutics for
diabetic neuropathy. Current Drug Targets, 9, 47–59.
Chowdhury, S. K., Dobrowsky, R. T., & Fernyhough, P. (2011). Nutrient excess and altered
mitochondrial proteome and function contribute to neurodegeneration in diabetes.
Mitochondrion, 11, 845–854.
Chowdhury, S. K., Smith, D. R., & Fernyhough, P. (2013). The role of aberrant mitochon-
drial bioenergetics in diabetic neuropathy. Neurobiology of Disease, 51, 56–65.
Chowdhury, S. K., Zherebitskaya, E., Smith, D. R., Akude, E., Chattopadhyay, S.,
Jolivalt, C. G., et al. (2010). Mitochondrial respiratory chain dysfunction in dorsal root
ganglia of streptozotocin-induced diabetic rats and its correction by insulin treatment.
Diabetes, 59, 1082–1091.
Figueroa-Romero, C., Sadidi, M., & Feldman, E. L. (2008). Mechanisms of disease: The
oxidative stress theory of diabetic neuropathy. Reviews in Endocrine & Metabolic Disorders,
9, 301–314.
Fujimura, H., Lacroix, C., & Said, G. (1991). Vulnerability of nerve fibres to ischaemia.
A quantitative light and electron microscope study. Brain, 114(Pt. 4), 1929–1942.
Halim, N. D., McFate, T., Mohyeldin, A., Okagaki, P., Korotchkina, L. G., Patel, M. S.,
et al. (2010). Phosphorylation status of pyruvate dehydrogenase distinguishes metabolic
phenotypes of cultured rat brain astrocytes and neurons. Glia, 58, 1168–1176.
Ishii, D. N. (1995). Implication of insulin-like growth factors in the pathogenesis of diabetic
neuropathy. Brain Research. Brain Research Reviews, 20, 47–67.
Jiang, Y., Calcutt, N. A., Ramos, K. M., & Mizisin, A. P. (2006). Novel sites of aldose reduc-
tase immunolocalization in normal and streptozotocin-diabetic rats. Journal of the Periph-
eral Nervous System, 11, 274–285.
Mizisin, A. P., Calcutt, N. A., DiStefano, P. S., Acheson, A., & Longo, F. M. (1997). Aldose
reductase inhibition increases CNTF-like bioactivity and protein in sciatic nerves from
galactose-fed and normal rats. Diabetes, 46, 647–652.
Mizisin, A. P., Vu, Y., Shuff, M., & Calcutt, N. A. (2004). Ciliary neurotrophic factor
improves nerve conduction and ameliorates regeneration deficits in diabetic rats.
Diabetes, 53, 1807–1812.
Nishikawa, T., Edelstein, D., & Brownlee, M. (2000). The missing link: A single unifying
mechanism for diabetic complications. Kidney International. Supplement, 77, S26–S30.
Nishikawa, T., Edelstein, D., Du, X. L., Yamagishi, S., Matsumura, T., Kaneda, Y., et al.
(2000b). Normalizing mitochondrial superoxide production blocks three pathways of
hyperglycaemic damage. Nature, 404, 787–790.
Oates, P. J. (2008). Aldose reductase, still a compelling target for diabetic neuropathy. Current
Drug Targets, 9, 14–36.
120 P. Fernyhough and N.A. Calcutt
Orestes, P., Osuru, H. P., McIntire, W. E., Jacus, M. O., Salajegheh, R., Jagodic, M. M.,
et al. (2013). Reversal of neuropathic pain in diabetes by targeting glycosylation of Ca(V)
3.2T-type calcium channels. Diabetes, 62, 3828–3838.
Thornalley, P. J. (2002). Glycation in diabetic neuropathy: Characteristics, consequences,
causes, and therapeutic options. International Review of Neurobiology, 50, 37–57.
Tomlinson, D. R., Fernyhough, P., & Diemel, L. T. (1997). Role of neurotrophins in
diabetic neuropathy and treatment with nerve growth factors. Diabetes, 46(Suppl. 2),
S43–S49.
Tomlinson, D. R., & Gardiner, N. J. (2008). Glucose neurotoxicity. Nature Reviews.
Neuroscience, 9, 36–45.
Vincent, A. M., Hayes, J. M., McLean, L. L., Vivekanandan-Giri, A., Pennathur, S., &
Feldman, E. L. (2009). Dyslipidemia-induced neuropathy in mice: The role of
oxLDL/LOX-1. Diabetes, 58, 2376–2385.
Vincent, A. M., Hinder, L. M., Pop-Busui, R., & Feldman, E. L. (2009). Hyperlipidemia:
A new therapeutic target for diabetic neuropathy. Journal of the Peripheral Nervous System,
14, 257–267.
Vlassara, H., Brownlee, M., & Cerami, A. (1981). Nonenzymatic glycosylation of peripheral
nerve protein in diabetes mellitus. Proceedings of the National Academy of Sciences of the
United States of America, 78, 5190–5192.
Zhang, L., Zhao, H., Blagg, B. S., & Dobrowsky, R. T. (2012). C-terminal heat shock
protein 90 inhibitor decreases hyperglycemia-induced oxidative stress and improves
mitochondrial bioenergetics in sensory neurons. Journal of Proteome Research, 11,
2581–2593.
Ziegler, D., Low, P. A., Litchy, W. J., Boulton, A. J., Vinik, A. I., Freeman, R., et al. (2011).
Efficacy and safety of antioxidant treatment with alpha-lipoic acid over 4 years in diabetic
polyneuropathy: The NATHAN 1 trial. Diabetes Care, 34, 2054–2060.
CHAPTER EIGHT
Contents
1. Introduction 122
2. Glucose-Metabolizing Pathways Relevant to DPN 124
3. Polyol (AR or Sorbitol–Fructose) Pathway 125
3.1 Metabolic Sequelae After Increased Flux Through the Polyol Pathway 125
3.2 Studies in Transgenic and Knockout Mice 128
3.3 Effects of AR Inhibition 129
3.4 Clinical Application of ARIs 130
3.5 Polyol Pathway in Ischemia/Reperfusion Injury 131
4. Nonenzymatic Glycation and AGEs 131
4.1 Glycation of Proteins in Diabetes 131
4.2 Glycation and AGE in DPN 132
4.3 Transgenic and Knockout Mice Studies 134
4.4 Clinical Application of Antiglycation Agents 134
5. Oxidative Stress 135
5.1 Production of Oxidative Stress by Hyperglycemia 135
5.2 Oxidative Stress and Diabetic Neuropathy 136
5.3 Effects of Antioxidants on Diabetic Neuropathy 137
6. PKC Activity 138
7. Glycosylation 139
7.1 The Hexosamine Pathway 139
7.2 Other Glycosylation Events 140
8. Conclusion 141
Acknowledgments 141
References 141
Abstract
Neuropathy is the earliest and commonest complication of diabetes. With increasing
duration of diabetes, frequency and severity of neuropathy are worsened. Long-term
1
Present address: The Nukada Institute of Medical and Biological Research, 4-16 Inage-machi,
Inage-ku, Chiba, Japan. E-mail: [email protected].
1. INTRODUCTION
There is a long history of research to clarify the pathogenetic mech-
anism of diabetic polyneuropathy (DPN), but it remains unsettled. Since the
prevalence of DPN is primarily dependent on the duration of diabetes and
the degree of blood glucose control, long-term metabolic aberration is con-
sidered to be a major cause of DPN (Pirart, 1978). A large prospective study
disclosed that hyperglycemia, duration of diabetes, hypertension, hyperlip-
idemia, and smoking are significant risk factors for the development of DPN
in patients with type 1 diabetes (Tesfaye et al., 2005). Indeed, the 10-year
follow-up of patients with type 1 diabetes by the Diabetic Complication
and Control Trial (DCCT) demonstrated that meticulous blood glucose
control by intensive insulin therapy suppressed the incidence of DPN
(DCCT Research Group, 1993). The effects of tight glycemic control per-
sisted for a further 8 years after the termination of the trial as legacy effects
termed glucose or metabolic memory (Martin et al., 2006). Although the
role of hyperglycemia in driving the progression of DPN in type 2 diabetes
is less clear (UKPDS Group, 1998), continuous lowering of glycated hemo-
globin below 7% for 6 years was found to suppress the deterioration of DPN
in patients with type 2 diabetes (Ohkubo et al., 1995). Despite the ample
epidemiological data, there remains no clear-cut explanation why long-term
hyperglycemia leads to DPN.
It is known that the most distal portions of somatic nerves are preferen-
tially affected in diabetes. Both anatomical and biochemical characteristics of
Glucotoxicity and Diabetic Neuropathy 123
the peripheral nervous system seem to contribute to the distal and sensory
predominant nerve lesions in diabetes (Dyck et al., 1984; Dyck, Lais,
Karnes, O’Brien, & Rizza, 1986). Structurally, the sensory neurons extend
extremely long axons from their cell bodies in the dorsal root ganglia
(DRG). The relative imbalance in this architecture between the soma
and axon can result in insufficient supply of nerve nutrients and energy from
the cell body to the distal processes. The anatomic organization of the vas-
cular supply to the peripheral nerve may also impose negative impacts on
the peripheral nerve because of the relative paucity of endoneurial blood
vessels and their lack of autoregulation (Kihara, Schmelzer, et al., 1991;
Smith, Kobrine, & Rizzoli, 1977). Such a vascular system likely renders
the peripheral nerve ischemic, resulting in severe pathology when ischemia
is further exaggerated by vascular occlusion (Nukada, McMorran, Baba,
Ogasawara, & Yagihashi, 2011), though ischemic diabetic nerve is tempo-
rarily resistant to nerve conduction failure (Low, Schmelzer, & Ward,
1986). Endoneurial microangiopathic changes characteristic of diabetes
may further augment nerve damage due to increased permeability and
ischemia (Dyck & Giannini, 1996; Malik et al., 2005; Thrainsdottir
et al., 2003). Features such as the strong expression of aldose reductase
(AR) in peripheral nerve and the abundance of interstitial collagen impli-
cate the polyol pathway and glycation as etiological factors in DPN.
Glucose metabolism in the peripheral nerve is not completely under-
stood. Peripheral nerves express the GLUT-1 glucose transporter at the
perineurial and endothelial blood:nerve barriers (Stark, Carlstedt,
Cullheim, & Risling, 2000), and glucose uptake into nerve is therefore insu-
lin independent so that endoneurial glucose concentrations will follow
plasma glucose concentrations. Under normoglycemic conditions, the
majority of glucose entering the cytoplasm is used by mitochondria to pro-
duce adenosine triphosphate (ATP) through the tricarboxylic acid (TCA)
cycle, downstream from the glycolytic pathway. In neurons, synthesized
proteins, cytoplasmic organelles, cargoes of microtubules and neuro-
filaments, as well as neurotransmitters, are all transported in an energy-
requiring process to the distal nerve endings. It has been proposed that if
the energy production in the axon is deficient then the distal portion is first
affected by the insufficient supply of these materials (Scott, Clark, &
Zochodne, 1999; Yagihashi, Kamijo, & Watanabe, 1990). Additionally,
hyperglycemia makes glucose available for other uses and perturbed glyco-
lytic pathways may lead to impaired nerve function and disruption of
124 S. Yagihashi
nerve structure. In this review, I will list the collateral glycolytic pathways
operating downstream of hyperglycemia and discuss their possible implica-
tion in the onset and development of DPN.
Fig. 1 Glycolytic pathways in hyperglycemia proposed for the cause of diabetic com-
plications. In normoglycemia, intracellular glucose undergoes glycolysis and enters the
TCA cycle in mitochondria to produce ATP. Hyperglycemia induces an increased flux
through the polyol pathway in endothelial and Schwann cells, nonenzymatic glycation
with AGE formation, superoxide production in endothelial cell mitochondria, increased
activity of protein kinase C, and enhancement of the hexosamine pathway. These com-
plicated glycolytic pathways are proposed to jointly lead to functional and structural
abnormalities encountered in diabetic complications. It should be of note, however,
that this theory is mainly built from data on vascular complications in diabetes, and
not uniformly applied to neuropathy. For example, reactive oxygen species production
in mitochondria is reduced in diabetic neurons (see text).
Glucotoxicity and Diabetic Neuropathy 125
(Fig. 2). The original osmotic theory proposed that increased polyol path-
way flux during hyperglycemia caused intracellular hyperosmolarity by
accumulation of sorbitol, resulting in the expansion of cells and ultimately
cell lysis (Gabbay, 1975; Kinoshita, 1990). However, although increased
sorbitol concentrations were demonstrated in nerves of persons with diabe-
tes, there was no evidence of nerve edema, swollen cells, or correlation with
nerve fiber pathology (Dyck et al., 1980, 1988).
Greene and his associates proposed the poor energy utilization theory as a
surrogate of the osmotic theory (Greene, Lattimer, & Sima, 1987; Greene,
Sima, Stevens, Feldman, & Lattimer, 1992). With accumulation of sorbitol,
other osmolytes including myo-inositol, taurine, and adenosine are depleted
Glucotoxicity and Diabetic Neuropathy 127
Fig. 3 Topographic difference in the localization of the enzymes that catalyze polyol
pathway. Aldose reductase locates in the endoneurium, mainly Schwann cells and
the wall of epineurial artery. On the other hand, sorbitol dehydrogenase (SDH)
expresses mainly in the wall of epineurial artery, but not much in the endoneurium.
Such difference may contribute to the distinct downstream of metabolic signals such
as protein kinase C and redox changes. Cited from Kasajima, H., Yamagishi, S., Sugai,
S., Yagihashi, N., & Yagihashi, S. (2001). Enhanced in situ expression of aldose reductase
in peripheral nerve and renal glomeruli in diabetic patients. Virchows Arch, 439, 46–54.
Mizisin, 2006), whereas SDH was more highly expressed in vascular tissues
(Fig. 3). Such anatomical differences in the localization of polyol pathway
enzymes may also be involved in the differential effects of diabetes on
PKC activity between nerve and vascular tissues, where it is increased in vas-
cular tissues and decreased in neural tissues (Yamagishi et al., 2003).
Fig. 4 Biological reactions elicited by the binding of AGE with RAGE in endothelial cells
and neural tissues. AGE exerts activation of NADPH oxidase to release oxygen radicals
after binding with RAGE. Concurrently, nuclear factor-κB (NF-κB) is transferred into the
nuclei to promote gene activation. This results in cellular dysfunction and activation of
cell death signals.
Federlin, 1996). However, since these studies used a rather small number of
subjects, more definitive trials are required.
Other agents that intervene against AGE accumulation or the biological
reactions downstream of AGE/RAGE interaction have also been devel-
oped, including pyridoxamine/vitamin B6 (Metz, Alderson, Thorpe, &
Baynes, 2003), AGE breakers (Vasan, Foiles, & Founds, 2003), and soluble
RAGE (Nagai, Shirakawa, Ohno, Moroishi, & Nagai, 2014), although at
present there is no information regarding the effects of these agents against
diabetic neuropathy in clinical trials.
5. OXIDATIVE STRESS
5.1 Production of Oxidative Stress by Hyperglycemia
The generation of free radicals due to increased flux of glucose through
glycolysis has been proposed as a major contributor to diabetic complications,
including DPN. This hypothesis was originally raised by the short-term
experiments mostly on in vitro endothelial cells and is still not confirmed
in neural tissues. During hyperglycemia, increased mitochondrial oxidation
of NADH and FADH2 leads to excessive formation of superoxide ions,
a form of ROS (Brownlee, 2005) (Fig. 5). Reduced expression of
manganese–superoxide dismutase also contributes to production of ROS
(Boyle, Newsom, Janssen, Lappas, & Friedman, 2013; Santos, Tewari,
Goldberg, & Kowluru, 2011). Thus, in endothelial cells superoxide produc-
tion, possibly via enhanced mitochondrial electron transport, could induce
DNA injury, which in turn drives increased expression of poly(ADP)-ribose
polymerase (PARP) to repair DNA damage. Activated PARP inhibits the
action of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (Obrosova,
Drel, et al., 2005). Consequently, upstream glycolytic metabolites before
GAPDH will be increased in the cytoplasm, which in turn induces
enhancement of four glycolytic pathways during hyperglycemia (Baynes &
Thorpe, 1999; Brownlee, 1992). However, recent studies do not support
the oxidative stress having a mitochondrial origin in sensory neurons in
experimental diabetes (Akude et al., 2011; Zherebitskaya, Akude, Smith, &
Fernyhough, 2009). These authors found reduced electron transport in mito-
chondria which was associated with reduced superoxide production, which
makes sense since less electron flux equates with less electron leakage. The
Dobrowsky group also detected reductions in the rates of electron transport
in adult sensory neurons exposed to hyperglycemia or derived from diabetic
mice (Urban et al., 2012; Zhang, Zhao, Blagg, & Dobrowsky, 2012). Studies
136 S. Yagihashi
Fig. 5 Mitochondrial electron transport and superoxide production. For the production
of ATP NADH is first oxidized to NAD with release of electrons at the site of complex I of
inner mitochondrial membrane. At the site of complex II, conversion of succinate to
fumarate is mediated with FADH2 conversion to FAD. Then, electrons are directly trans-
ferred at the complex III from coenzyme Q (Co-Q) to cytochrome C (Cyt-C). If this step is
augmented by hyperglycemia in endothelial cells or blocked by drugs such as actino-
mycin D, superoxide radical formation is enhanced. Recent data suggest that this
theory is only applicable to vascular tissues, but not neurons (see text). Modified from
Brownlee, M. (2005). The pathobiology of diabetic complications: A unifying mechanism.
Diabetes, 54, 1615–1625.
with diabetes (Ametov et al., 2003), and this finding was replicated in the
NATHAN study of 4 years duration (Ziegler et al., 2011). However, the
results concerning efficacy against nerve conduction slowing and other objec-
tive endpoints were inconclusive due to minimal progression of neuropathy
over the study period.
6. PKC ACTIVITY
PKC plays a role in maintaining normal nerve function, and its aber-
rant regulation may contribute to the pathogenesis of DPN (Way, Katai, &
King, 2001). PKC has several isoforms from α, β, γ, δ, and so on. Under
ambient hyperglycemia, glucose is converted to glyceraldehydes-3-
phosphate and phosphatidic acid, which in turn changes into diacylglycerol
that serves as a substrate of PKC-β. Vascular tissues in diabetes thus show
increased PKC activity, leading to increased permeability and dysfunction
(Geraldes & King, 2010). An inhibitor of PKC-β was found to be effective
for correction of vascular abnormalities in diabetic animals (King &
Brownlee, 1996). Based on the premise that microangiopathy in nerve
may contribute to development of neuropathy, preclinical and clinical trials
of PKC-β inhibitor were conducted. Experimental studies demonstrated
beneficial effects of a PKC-β inhibitor on peripheral nerve dysfunction
and nerve blood flow in diabetic rats (Cameron & Cotter, 2002). However,
the alterations of PKC activity are complex in nerves and their supportive
vascular systems, in part due to the fact that the major enzymes of collateral
glycolytic pathways are different between these two tissues (Fig. 3) (Kasajima
et al., 2001). Such nonuniform tissue composition might explain inconsis-
tent findings regarding PKC activity in diabetic nerves. Thus, Nakamura
et al. (1999) did not find any significant change of PKC activity in homog-
enates of whole peripheral nerve from STZ-diabetic rats, although a PKC-
β-specific inhibitor improved nerve conduction slowing and nerve blood
flow. In contrast, in our studies on STZ-induced diabetic mice, we separated
the peripheral nerve tissues into endoneurium and epineurium for the mea-
surement of PKC activity, the latter of which was rich in microvessels
(Yamagishi et al., 2003). We found that the former showed decreased
PKC activity with significantly decreased expression of the membranous
PKC-α isoform, whereas the latter showed increased PKC activity with
enhanced expression of the PKC-β isoform. This increase of PKC activity
in the epineurial compartment is consistent with reports in other systemic
vascular tissues, while the decrease in the endoneurial compartment is
Glucotoxicity and Diabetic Neuropathy 139
consistent with a report that Schwann cells exposed to high glucose in vitro
show reduced expression of PKC-α and reduced PKC activity (Kamiya
et al., 2003). Given these findings, the application of PKC-β-specific inhib-
itor would be expected to be a useful for the treatment of diabetic vascular
complications, but not any disorders related to reduced endoneurial expres-
sion of PKC-α. A 6-month clinical trial of the PKC-β-specific inhibitor
Ruboxistaurin in subjects with DPN showed that it increased skin blood
flow and mitigated some sensory symptoms but was without effect on objec-
tive measures of neuropathy such as large-fiber nerve conduction velocity
slowing, reduced small sensory fiber nerve density in the epidermis, or
impaired autonomic function (Casellini et al., 2007).
7. GLYCOSYLATION
7.1 The Hexosamine Pathway
Under conditions of elevated intracellular glucose and flux into glycolysis,
excess fructose 6-phosphate is converted to glucosamine 6-phosphate
(GlcN-6-phosphate) by glutamine-fructose-6-phosphate amidotransferase
(GFAT). GlcN-6-phosphate is then modified to UDP-N-acetylglucosamine
(UDP-GlcNAc) (Du et al., 2000; Issad, Masson, & Pagesy, 2010) (Fig. 6).
UDP-GlcNAc has an affinity for cell membrane or nucleic acid or transcrip-
tion factors and excessive modification of these substrates by posttranslational
modification of serine and threonine residues (termed O-GlcNAcylation) can
perturb cellular functions and may contribute to both the pathogenesis of
type 2 diabetes by promoting insulin resistance and also to the complications
of diabetes (Peterson & Hart, 2016). Evidence is strongest for a role of the
hexosamine pathway in predominantly vascular complications such as reti-
nopathy and nephropathy. For example, inhibition of GFAT suppresses
expression of genes for transforming growth factor (TGF)-α and TGF-β1
gene and also the activation of plasminogen activator inhibitor-I (PAI-I).
As increased expression and activity of these factors are known to be induced
by hyperglycemia, the hexosamine pathway is therefore implicated in vascular
complications of diabetes. UDP-GlcNAc modification of Akt activation sites
in vascular endothelial cells also reduced eNOS activity. Further, GFAT in
vascular smooth muscle cells is activated during hyperglycemia and muscle
proteins are excessively modified by UDP-GlcNAc (Heath et al., 2014). In
contrast to the plentiful data in vascular tissues, it is not yet entirely clear what
kinds of peripheral nerve proteins may be modified by the hexosamine path-
way during diabetes, although there is emerging evidence that imbalances in
140 S. Yagihashi
8. CONCLUSION
It is well established that long-term hyperglycemia contributes to the
genesis and development of DPN. However, the process is complicated and
the precise mechanisms of how high glucose affects the assorted cell types
within a nerve are still mysterious. Epidemiological studies suggest that many
factors are responsible for DPN, and the targeting of a single collateral path-
way may not be sufficient for the protection and suppression of DPN.
Experimental studies have identified multiple pathways that appear to be
involved in the functional and structural alterations in the peripheral nerves
in animal models of diabetes and there may yet be other undiscovered path-
ways. As the clinical signs and symptoms of DPN differ from case to case,
the dominant pathogenic mechanisms may also vary based on individual
genotype and phenotype. If this is the case, in addition to meticulous blood
glucose control, it will be important to identify which collateral pathway is
most relevant to specific clinical manifestations. Future investigations should
perhaps explore in more detail the relationships between each collateral
pathway and specific clinical manifestations of DPN.
ACKNOWLEDGMENTS
The author appreciates the previous collaborators who contributed to the published works
from the author’s laboratory. The author also is grateful for the research funding from the
Japanese government of the Ministry of Science, Culture, Education, and Sports, the
Ministry of Health and Welfare, and the Juvenile Diabetes Foundation International,
New York.
Duality of Interest: There is no conflict of interest for the author regarding the content of
this review.
REFERENCES
Akude, E., Zherebitskaya, E., Chowdhury, S. K., Smith, D. R., Dobrowsky, R. T., &
Fernyhough, P. (2011). Diminished superoxide generation is associated with respiratory
chain dysfunction and changes in the mitochondrial proteome of sensory neurons from
diabetic rats. Diabetes, 60, 288–297.
Ametov, A. S., Barinov, A., Dyck, P. J., Hermann, R., Kozlova, N., Litchy, W. J., et al.
(2003). The sensory symptoms of diabetic polyneuropathy are improved with alpha-
lipoic acid: The SYDNEY trial. Diabetes Care, 26, 770–776.
Baba, M., Kimura, K., Suda, T., Yagihashi, S., & Aomori Diabetic Study (ADNS) Group.
(2006). Three-year inhibition of aldose reductase on development of symptomatic neu-
ropathy in diabetic patients. Journal of the Peripheral Nervous System, 11, 176–178.
Baynes, J. W., & Thorpe, S. R. (1999). Role of oxidative stress in diabetic complications:
A new perspective on an old paradigm. Diabetes, 48, 1–9.
142 S. Yagihashi
Bierhaus, A., Fleming, T., Stoyanov, S., Leffler, A., Babes, A., Neacsu, C., et al. (2012).
Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes
hyperalgesia in diabetic neuropathy. Nature Medicine, 18, 926–933.
Bierhaus, A., Haslbeck, K. M., Humpert, P. M., Liliensiek, B., Dehmer, T., Morcos, M.,
et al. (2004). Loss of pain perception in diabetes is dependent on a receptor of the immu-
noglobulin superfamily. The Journal of Clinical Investigation, 114, 1741–1751.
Bolton, W. K., Cattran, D. C., Williams, M. E., Adler, S. G., Appel, G. B., Cartwright, K.,
et al. (2004). Randomized trial of an inhibitor of formation of advanced glycation end
products in diabetic nephropathy. American Journal of Nephrology, 24, 32–40.
Boyle, K. E., Newsom, S. A., Janssen, R. C., Lappas, M., & Friedman, J. E. (2013). Skeletal
muscle MnSOD, mitochondrial complex II, and SIRT3 enzyme activities are decreased
in maternal obesity during human pregnancy and gestational diabetes mellitus. The Jour-
nal of Clinical Endocrinology and Metabolism, 98, E1601–E1609.
Bril, V., & Buchanan, R. A. (2006). Long-term effects of ranirestat (AS-3201) on peripheral
nerve function in patients with diabetic sensorimotor polyneuropathy. Diabetes Care, 29,
68–72.
Brownlee, M. (1992). Nonenzymatic glycosylation of macromolecules. Prospects of pharma-
cologic modulation. Diabetes, 41(Suppl. 2), 57–60.
Brownlee, M. (2005). The pathobiology of diabetic complications: A unifying mechanism.
Diabetes, 54, 1615–1625.
Calcutt, N. A., Tomlinson, D. R., & Biswas, S. (1990). Coexistence of nerve conduction
deficit with increased Na(+)-K(+)-ATPase activity in galactose-fed mice. Implications
for polyol pathway and diabetic neuropathy. Diabetes, 39, 663–666.
Cameron, N. E., & Cotter, M. A. (2002). Effects of protein kinase C-β inhibition on neu-
rovascular dysfunction in diabetic rats: Interaction with oxidative stress and essential fatty
acid dysmetabolism. Diabetes/Metabolism Research and Reviews, 18, 315–323.
Cameron, N. E., Cotter, M. A., Archibald, V., Dines, K. C., & Maxfield, E. K. (1994). Anti-
oxidant and pro-oxidant effects on nerve conduction velocity, endoneurial blood flow
and oxygen tension in non-diabetic and streptozotocin-diabetic rats. Diabetologia, 37,
449–459.
Cameron, N. E., Cotter, M. A., Horrobin, D. H., & Tritschler, H. J. (1998). Effects of alpha-
lipoic acid on neurovascular function in diabetic rats: Interaction with essential fatty
acids. Diabetologia, 41, 390–399.
Cameron, N. E., Cotter, M. A., & Maxfield, E. K. (1993). Anti-oxidant treatment prevents
the development of peripheral nerve dysfunction in streptozotocin-diabetic rats.
Diabetologia, 36, 299–304.
Casellini, C. M., Barlow, P. M., Rice, A. L., Casey, M., Simmons, K., Pittenger, G., et al.
(2007). A 6-month, randomized, double-masked, placebo-controlled study evaluating
the effects of the protein kinase C-β inhibitor ruboxistaurin on skin microvascular blood
flow and other measures of diabetic peripheral neuropathy. Diabetes Care, 30, 896–902.
Cheung, A. K., Lo, A. C., So, K. F., Chung, S. S., & Chung, S. K. (2007). Gene deletion and
pharmacological inhibition of aldose reductase protect against retinal ischemic injury.
Experimental Eye Research, 85, 608–616.
Chowdhury, S. K., Zherebitskaya, E., Smith, D. R., Akude, E., Chattopadhyay, S.,
Jolivalt, C. G., et al. (2010). Mitochondrial respiratory chain dysfunction in dorsal root
ganglia of streptozotocin-induced diabetic rats and its correction by insulin treatment.
Diabetes, 59, 1082–1091.
Corbett, J. A., & McDaniel, M. L. (1996). Selective inhibition of inducible nitric oxide
synthase by aminoguanidine. Methods in Enzymology, 268, 398–408.
Diabetes Control and Complications Trial Research Group. (1993). The effect of
intensive treatment of diabetes on the development and progression of long-term com-
plications in insulin-dependent diabetes mellitus. The New England Journal of Medicine,
329, 977–986.
Glucotoxicity and Diabetic Neuropathy 143
Du, X. L., Edelstein, D., Rossetti, L., Fantus, I. G., Goldberg, H., Ziyadeh, F., et al. (2000).
Hyperglycemia-induced mitochondrial superoxide overproduction activates the
hexosamine pathway and induces plasminogen activator inhibitor-1 expression by
increasing Sp1 glycosylation. Proceedings of the National Academy of Sciences of the United
States of America, 97, 12222–12226.
Duran-Jimenez, B., Dobler, D., Moffatt, S., Rabbani, N., Streuli, C. H., Thornalley, P. J.,
et al. (2009). Advanced glycation end products in extracellular matrix proteins contribute
to the failure of sensory nerve regeneration in diabetes. Diabetes, 58, 2893–2903.
Dyck, P. J., & Giannini, C. (1996). Pathologic alterations in the diabetic neuropathies of
humans: A review. Journal of Neuropathology and Experimental Neurology, 55, 1181–1193.
Dyck, P. J., Karnes, J., O’Brien, P., Nukada, H., Lais, A., & Low, P. (1984). Spatial pattern of
nerve fiber abnormality indicative of pathologic mechanism. The American Journal of
Pathology, 117, 225–238.
Dyck, P. J., Lais, A., Karnes, J. L., O’Brien, P., & Rizza, R. (1986). The spatial distribution of
fiber loss in diabetic polyneuropathy suggests ischemia. Annals of Neurology, 19, 440–449.
Dyck, P. J., Sherman, W. R., Hallcher, L. M., Service, F. J., O’Brien, P. C., Grina, L. A.,
et al. (1980). Human diabetic endoneurial sorbitol, fructose, and myo-inositol related to
sural nerve morphometry. Annals of Neurology, 8, 590–596.
Dyck, P. J., Zimmerman, B. R., Vilen, T. H., Minnerath, S. R., Karnes, J. L., Yao, J. K.,
et al. (1988). Nerve glucose, fructose, sorbitol, myo-inositol, and fiber degeneration and
regeneration in diabetic neuropathy. The New England Journal of Medicine, 319, 542–548.
Gabbay, K. H. (1975). Hyperglycemia, polyol metabolism, and complications of diabetes
mellitus. Annual Review of Medicine, 26, 521–536.
Geraldes, P., & King, G. L. (2010). Activation of protein kinase C isoforms and its impact on
diabetic complications. Circulation Research, 106, 1319–1331.
Goto, Y., Hotta, N., Shigeta, Y., Sakamoto, N., & Kikkawa, R. (1995). Effects of an aldose
reductase inhibitor, epalrestat, on diabetic neuropathy. Clinical benefit and indication for
the drug assessed from the results of a placebo-controlled double-blind study. Biomedi-
cine & Pharmacotherapy, 49, 269–277.
Greene, D. A., Arezzo, J. C., & Brown, M. B. (1999). Effect of aldose reductase inhibition on
nerve conduction and morphometry in diabetic neuropathy. Zenarestat Study Group.
Neurology, 53, 580–591.
Greene, D. A., Lattimer, S. A., & Sima, A. A. (1987). Sorbitol, phosphoinositides, and
sodium-potassium-ATPase in the pathogenesis of diabetic complications. The
New England Journal of Medicine, 316, 599–606.
Greene, D. A., Sima, A. A., Stevens, M. J., Feldman, E. L., & Lattimer, S. A. (1992).
Complications: Neuropathy, pathogenetic considerations. Diabetes Care, 15, 1902–1925.
Greene, D. A., Stevens, M. J., Obrosova, I., & Feldman, E. L. (1999). Glucose-induced
oxidative stress and programmed cell death in diabetic neuropathy. European Journal of
Pharmacology, 375, 217–223.
Hardiville, S., & Hart, G. W. (2014). Nutrient regulation of signaling, transcription, and cell
physiology by O-GlcNAcylation. Cell Metabolism, 20, 208–213.
Hart, G. W. (2014). Minireview series on the thirtieth anniversary of research on
O-GlcNAcylation of nuclear and cytoplasmic proteins: Nutrient regulation of cellular
metabolism and physiology by O-GlcNAcylation. The Journal of Biological Chemistry,
289, 34422–34433.
Haslbeck, K. M., Schleicher, E., Bierhaus, A., Nawroth, P., Haslbeck, M., Neund€ orfer, B.,
et al. (2005). The AGE/RAGE/NF-(kappa)B pathway may contribute to the pathogen-
esis of polyneuropathy in impaired glucose tolerance (IGT). Experimental and Clinical
Endocrinology & Diabetes, 113, 288–291.
Haupt, E., Ledermann, H., & K€ opcke, W. (2005). Benfotiamine in the treatment of diabetic
polyneuropathy—A three-week randomized, controlled pilot study (BEDIP study).
International Journal of Clinical Pharmacology & Therapeutics, 43, 71–77.
144 S. Yagihashi
Heath, J. M., Sun, Y., Yuan, K., Bradley, W. E., Litovsky, S., Dell’Italia, L. J., et al. (2014).
Activation of AKT by O-linked N-acetylglucosamine induces vascular calcification in
diabetes mellitus. Circulation Research, 114, 1094–1102.
Ho, E. C., Lam, K. S., Chen, Y. S., Yip, J. C., Arvindakshan, M., Yamagishi, S., et al. (2006).
Aldose reductase-deficient mice are protected from delayed motor nerve conduction
velocity, increased c-Jun NH2-terminal kinase activation, depletion of reduced glutathi-
one, increased superoxide accumulation, and DNA damage. Diabetes, 55, 1946–1953.
Hotta, N., Akanuma, Y., Kawamori, R., Matsuoka, K., Oka, Y., Shichiri, M., et al. (2006).
Long-term clinical effects of epalrestat, an aldose reductase inhibitor, on diabetic periph-
eral neuropathy: The 3-year, multicenter, comparative Aldose Reductase Inhibitor-
Diabetes Complications Trial. Diabetes Care, 29, 1538–1544.
Hotta, N., Kawamori, R., Atsumi, Y., Baba, M., Kishikawa, H., Nakamura, J., et al. (2008).
Stratified analyses for selecting appropriate target patients with diabetic peripheral neu-
ropathy for long-term treatment with an aldose reductase inhibitor, epalrestat. Diabetic
Medicine, 25, 818–825.
Huang, T. J., Price, S. A., Chilton, L., Calcutt, N. A., Tomlinson, D. R., Verkhratsky, A.,
et al. (2003). Insulin prevents depolarization of the mitochondrial inner membrane in
sensory neurons of type 1 diabetic rats in the presence of sustained hyperglycemia.
Diabetes, 52, 2129–2136.
Ido, Y., Nyengaard, J. R., Chang, K., Tilton, R. G., Kilo, C., Mylari, B. L., et al. (2010).
Early neural and vascular dysfunctions in diabetic rats are largely sequelae of increased
sorbitol oxidation. Antioxidants & Redox Signaling, 12, 39–51.
Ilnytska, O., Lyzogubov, V. V., Stevens, M. J., Drel, V. R., Mashtalir, N., Pacher, P., et al.
(2006). Poly(ADP-ribose) polymerase inhibition alleviates experimental diabetic sensory
neuropathy. Diabetes, 55, 1686–1694.
Inagi, R., Yamamoto, Y., Nangaku, M., Usuda, N., Okamato, H., Kurokawa, K., et al.
(2006). A severe diabetic nephropathy model with early development of nodule-like
lesions induced by megsin overexpression in RAGE/iNOS transgenic mice. Diabetes,
55, 356–366.
Issad, T., Masson, E., & Pagesy, P. (2010). O-GlcNAc modification, insulin signaling and
diabetic complications. Diabetes & Metabolism, 36, 423–435.
Iwata, K., Matsuno, K., Nishinaka, T., Persson, C., & Yabe-Nishimura, C. (2006). Aldose
reductase inhibitors improve myocardial reperfusion injury in mice by a dual mechanism.
Journal of Pharmacological Sciences, 102, 37–46.
Jack, M. M., Ryals, J. M., & Wright, D. E. (2011). Characterisation of glyoxalase I in a
streptozocin-induced mouse model of diabetes with painful and insensate neuropathy.
Diabetologia, 54, 2174–2182.
James, L. R., Tang, D., Ingram, A., Ly, H., Thai, K., Cai, L., et al. (2002). Flux through the
hexosamine pathway is a determinant of nuclear factor κB-dependent promoter activa-
tion. Diabetes, 51, 1146–1156.
Jiang, Y., Calcutt, N. A., Ramos, K. M., & Mizisin, A. P. (2006). Novel sites of aldose reduc-
tase immunolocalization in normal and streptozotocin-diabetic rats. Journal of the Periph-
eral Nervous System, 11, 274–285.
Juranek, J. K., Geddis, M. S., Song, F., Zhang, J., Garcia, J., Rosario, R., et al. (2013). RAGE
deficiency improves postinjury sciatic nerve regeneration in type 1 diabetic mice.
Diabetes, 62, 931–943.
Kamiya, H., Nakamura, J., Hamada, Y., Nakashima, E., Naruse, K., Kato, K., et al. (2003).
Polyol pathway and protein kinase C activity of rat Schwannoma cells. Diabetes/Metab-
olism Research and Reviews, 19, 131–139.
Kasajima, H., Yamagishi, S., Sugai, S., Yagihashi, N., & Yagihashi, S. (2001). Enhanced
in situ expression of aldose reductase in peripheral nerve and renal glomeruli in diabetic
patients. Virchows Archiv, 439, 46–54.
Glucotoxicity and Diabetic Neuropathy 145
Kihara, M., Schmelzer, J. D., Poduslo, J. F., Curran, G. L., Nickander, K. K., & Low, P. A.
(1991). Aminoguanidine effects on nerve blood flow, vascular permeability, electrophys-
iology, and oxygen free radicals. Proceedings of the National Academy of Sciences of the United
States of America, 88, 6107–6111.
Kihara, M., Weerasuriya, A., & Low, P. A. (1991). Endoneurial blood flow in rat sciatic
nerve during development. The Journal of Physiology, 439, 351–360.
Kikuchi, S., Shinpo, K., Moriwaka, F., Makita, Z., Miyata, T., & Tashiro, K. (1999).
Neurotoxicity of methylglyoxal and 3-deoxyglucosone on cultured cortical neurons:
Synergism between glycation and oxidative stress, possibly involved in neurodegenera-
tive diseases. Journal of Neuroscience Research, 57, 280–289.
King, G. L., & Brownlee, M. (1996). The cellular and molecular mechanisms of diabetic
complications. Endocrinology and Metabolism Clinics of North America, 25, 255–270.
Kinoshita, J. H. (1990). A thirty year journey in the polyol pathway. Experimental Eye
Research, 50, 567–573.
Kishi, Y., Schmelzer, J. D., Yao, J. K., Zollman, P. J., Nickander, K. K., Tritschler, H. J.,
et al. (1999). Alpha-lipoic acid: Effect on glucose uptake, sorbitol pathway, and energy
metabolism in experimental diabetic neuropathy. Diabetes, 48, 2045–2051.
Lambourne, J. E., Tomlinson, D. R., Brown, A. M., & Willars, G. B. (1987). Opposite
effects of diabetes and galactosaemia on adenosine triphosphatase activity in rat nervous
tissue. Diabetologia, 30, 360–362.
Leinninger, G. M., Edwards, J. L., Lipshaw, M. J., & Feldman, E. (2006). Mechanisms of
disease: Mitochondria as new therapeutic targets in diabetic neuropathy. Nature Clinical
Practice. Neurology, 2, 620–628.
Lim, J. G., Lee, J. J., Park, S. H., Park, J. H., Kim, S. J., Cho, H. C., et al. (2010). Glucagon-
like peptide-1 protects NSC-34 motor neurons against glucosamine through
Epac-mediated glucose uptake enhancement. Neuroscience Letters, 479, 13–17.
Low, P. A., Nickander, K. K., & Tritschler, H. J. (1997). The roles of oxidative stress and anti-
oxidant treatment in experimental diabetic neuropathy. Diabetes, 46(Suppl. 2), S38–S42.
Low, P. A., Schmelzer, J. D., & Ward, K. K. (1986). The effect of age on energy metabolism
and resistance to ischaemic conduction failure in rat peripheral nerve. The Journal of Phys-
iology, 374, 263–271.
Lupachyk, S., Shevalye, H., Maksimchyk, Y., Drel, V. R., & Obrosova, I. G. (2011). PARP
inhibition alleviates diabetes-induced systemic oxidative stress and neural tissue
4-hydroxynonenal adduct accumulation: Correlation with peripheral nerve function.
Free Radical Biology & Medicine, 50, 1400–1409.
Malik, R. A., Tesfaye, S., Newrick, P. G., Walker, D., Rajbhandari, S. M., Siddique, I., et al.
(2005). Sural nerve pathology in diabetic patients with minimal but progressive neurop-
athy. Diabetologia, 48, 578–585.
Martin, C. L., Albers, J., Herman, W. H., Cleary, P., Waberski, B., Greene, D. A., et al.
(2006). Neuropathy among the diabetes control and complications trial cohort 8 years
after trial completion. Diabetes Care, 29, 340–344.
McLean, W. G., Pekiner, C., Cullum, N. A., & Casson, I. F. (1992). Posttranslational mod-
ifications of nerve cytoskeletal proteins in experimental diabetes. Molecular Neurobiology,
6, 225–237.
Metz, T. O., Alderson, N. L., Thorpe, S. R., & Baynes, J. W. (2003). Pyridoxamine, an
inhibitor of advanced glycation and lipoxidation reactions: A novel therapy for treatment
of diabetic complications. Archives of Biochemistry and Biophysics, 419, 41–49.
Nagai, R., Shirakawa, J., Ohno, R., Moroishi, N., & Nagai, M. (2014). Inhibition of AGEs
formation by natural products. Amino Acids, 46, 261–266.
Nakamura, J., Kato, K., Hamada, Y., Nakayama, M., Chaya, S., Nakashima, E., et al. (1999).
A protein kinase C-beta-selective inhibitor ameliorates neural dysfunction in
streptozotocin-induced diabetic rats. Diabetes, 48, 2090–2095.
146 S. Yagihashi
Ng, T. F., Lee, F. K., Song, Z. T., Calcutt, N. A., Lee, A. Y., Chung, S. S., et al. (1998).
Effects of sorbitol dehydrogenase deficiency on nerve conduction in experimental dia-
betic mice. Diabetes, 47, 961–966.
Nishizawa, Y., Wada, R., Baba, M., Takeuchi, M., Hanyu-Itabashi, C., & Yagihashi, S.
(2010). Neuropathy induced by exogenously administered advanced glycation end-
products. Journal of Diabetes Investigation, 1, 40–49.
Nukada, H., Dyck, P. J., Low, P. A., Lais, A. C., & Sparks, M. F. (1986). Axonal caliber and
neurofilaments are proportionately decreased in galactose neuropathy. Journal of Neuro-
pathology and Experimental Neurology, 45, 140–150.
Nukada, H., McMorran, P. D., Baba, M., Ogasawara, S., & Yagihashi, S. (2011). Increased
susceptibility to ischemia and macrophage activation in STZ-diabetic rat nerve. Brain
Research, 1373, 172–182.
Obrosova, I. G. (2009). Diabetes and the peripheral nerve. Biochimica et Biophysica Acta, 1792,
931–940.
Obrosova, I. G., Drel, V. R., Pacher, P., Ilnytska, O., Wang, Z. Q., Stevens, M. J.,
et al. (2005). Oxidative-nitrosative stress and poly(ADP-ribose) polymerase (PARP)
activation in experimental diabetic neuropathy: The relation is revisited. Diabetes,
54, 3435–3441.
Obrosova, I. G., Ilnytska, O., Lyzogubov, V. V., Pavlov, I. A., Mashtalir, N., Nadler, J. L.,
et al. (2007). High-fat diet induced neuropathy of pre-diabetes and obesity: Effects of
“healthy” diet and aldose reductase inhibition. Diabetes, 56, 2598–2608.
Obrosova, I. G., Li, F., Abatan, O. I., Forsell, M. A., Komjáti, K., Pacher, P., et al. (2004).
Role of poly(ADP-ribose) polymerase activation in diabetic neuropathy. Diabetes, 53,
711–720.
Obrosova, I. G., Pacher, P., Szabó, C., Zsengeller, Z., Hirooka, H., Stevens, M. J., et al.
(2005). Aldose reductase inhibition counteracts oxidative-nitrosative stress and
poly(ADP-ribose) polymerase activation in tissue sites for diabetes complications.
Diabetes, 54, 234–242.
Ohkubo, Y., Kishikawa, H., Araki, E., Miyata, T., Isami, S., Motoyoshi, S., et al. (1995).
Intensive insulin therapy prevents the progression of diabetic microvascular complications
in Japanese patients with non-insulin-dependent diabetes mellitus: A randomized prospec-
tive 6-year study. Diabetes Research and Clinical Practice, 28, 103–117.
Peterson, S. B., & Hart, G. W. (2016). New insights: A role for O-GlcNAcylation in diabetic
complications. Critical Reviews in Biochemistry and Molecular Biology, 51, in press. http://dx.
doi.org/10.3109/10409238.2015.1135102.
Pirart, J. (1978). Diabetes mellitus and its degenerative complications: A prospective study of
4400 patients observed between 1947 and 1973. Diabetes Care, 1, 168–188.
Polydefkis, M., Arezzo, J., Nash, M., Bril, V., Shaibani, A., Gordon, R. J., et al. (2015).
Safety and efficacy of ranirestat in patients with mild-to-moderate diabetic sensorimotor
polyneuropathy. Journal of the Peripheral Nervous System, 20, 363–371.
Pop-Busui, R., Sima, A., & Stevens, M. (2006). Diabetic neuropathy and oxidative stress.
Diabetes/Metabolism Research and Reviews, 22, 257–273.
Rabbani, N., & Thornalley, P. J. (2015). Dicarbonyl stress in cell and tissue dysfunction con-
tributing to ageing and disease. Biochemical and Biophysical Research Communications, 458,
221–226.
Ramasamy, R., & Goldberg, I. J. (2010). Aldose reductase and cardiovascular diseases, cre-
ating human-like diabetic complications in an experimental model. Circulation Research,
106, 1449–1458.
Roy Chowdhury, S. K., Smith, D. R., Saleh, A., Schapansky, J., Marquez, A., Gomes, S.,
et al. (2012). Impaired adenosine monophosphate-activated protein kinase signalling
in dorsal root ganglia neurons is linked to mitochondrial dysfunction and peripheral
neuropathy in diabetes. Brain, 135, 1751–1766.
Glucotoxicity and Diabetic Neuropathy 147
Ryle, C., Leow, C. K., & Donaghy, M. (1997). Nonenzymatic glycation of peripheral and
central nervous system proteins in experimental diabetes mellitus. Muscle & Nerve, 20,
577–584.
Santos, J. M., Tewari, S., Goldberg, A. F., & Kowluru, R. A. (2011). Mitochondrial biogen-
esis and the development of diabetic retinopathy. Free Radical Biology & Medicine, 51,
1849–1860.
Schartner, E., Chowdhury, S. R., Smith, D., & Fernyhough, P. (2015). High glucose con-
centration down-regulates SIRT2 expression in sensory neurons via a sorbitol
dehydrogenase-dependent pathway. In (Abstract) Neurodiab 2015, 25th annual meeting
of the diabetic neuropathy study group of the EASD, September 11–13, Elsinore, Denmark
(Abstract O29) (p. 48).
Schmidt, R. E. (2002). Neuropathology and pathogenesis of diabetic autonomic neuropathy.
International Review of Neurobiology, 50, 257–292.
Schmidt, R. E., Dorsey, D. A., Beaudet, L. N., Plurad, S. B., Parvin, C. A., Yarasheski, K. E.,
et al. (2001). Inhibition of sorbitol dehydrogenase exacerbates autonomic neuropathy in
rats with streptozotocin-induced diabetes. Journal of Neuropathology and Experimental
Neurology, 60, 1153–1169.
Schmidt, R. E., Plurad, S. B., Sherman, W. R., Williamson, J. R., & Tilton, R. G. (1989).
Effects of aldose reductase inhibitor sorbinil on neuroaxonal dystrophy and levels of
myo-inositol and sorbitol in sympathetic autonomic ganglia of streptozocin-induced dia-
betic rats. Diabetes, 38, 569–579.
Scott, J. N., Clark, A. W., & Zochodne, D. W. (1999). Neurofilament and tubulin gene
expression in progressive experimental diabetes: Failure of synthesis and export by sen-
sory neurons. Brain, 122, 2109–2118.
Sekido, H., Suzuki, T., Jomori, T., Takeuchi, M., Yabe-Nishimura, C., & Yagihashi, S.
(2004). Reduced cell replication and induction of apoptosis by advanced glycation
end products in rat Schwann cells. Biochemical and Biophysical Research Communications,
320, 241–248.
Sima, A. A., Bril, V., Nathaniel, V., McEwen, T. A., Brown, M. B., Lattimer, S. A., et al.
(1988). Regeneration and repair of myelinated fibers in sural-nerve biopsy specimens
from patients with diabetic neuropathy treated with sorbinil. The New England Journal
of Medicine, 319, 548–555.
Simmons, D. A., Kern, E. F., Winegrad, A. I., & Martin, D. B. (1986). Basal pho-
sphatidylinositol turnover controls aortic Na +/K + ATPase activity. The Journal of
Clinical Investigation, 77, 503–513.
Smith, D. R., Kobrine, A. I., & Rizzoli, H. V. (1977). Absence of autoregulation in periph-
eral nerve blood flow. Journal of the Neurological Sciences, 33, 347–352.
Stark, B., Carlstedt, T., Cullheim, S., & Risling, M. (2000). Developmental and lesion-
induced changes in the distribution of the glucose transporter Glut-1 in the central
and peripheral nervous system. Experimental Brain Research, 131, 74–84.
Stevens, M. J., Dananberg, J., Feldman, E. L., Lattimer, S. A., Kamijo, M., Thomas, T. P.,
et al. (1994). The linked roles of nitric oxide, aldose reductase, and (Na+, K +)-ATPase
in the slowing of nerve conduction in the streptozotocin diabetic rat. The Journal of
Clinical Investigation, 94, 853–859.
Stevens, M. J., Obrosova, I., Cao, X., Van Huysen, C., & Greene, D. A. (2000).
Effects of DL-alpha-lipoic acid on peripheral nerve conduction, blood flow, energy
metabolism, and oxidative stress in experimental diabetic neuropathy. Diabetes, 49,
1006–1015.
Stracke, H., Gaus, W., Achenbach, U., Federlin, K., & Bretzel, R. G. (2008). Benfotiamine
in diabetic polyneuropathy (BENDIP): Results of a randomised, double blind, placebo-
controlled clinical study. Experimental and Clinical Endocrinology & Diabetes, 116,
600–605.
148 S. Yagihashi
Winegrad, A. I. (1987). Banting lecture 1986. Does a common mechanism induce the diverse
complications of diabetes? Diabetes, 36, 396–406.
Yagihashi, S., Kamijo, M., Baba, M., Yagihashi, N., & Nagai, K. (1992). Effect of
aminoguanidine on functional and structural abnormalities in peripheral nerve of
STZ-induced diabetic rats. Diabetes, 41, 47–52.
Yagihashi, S., Kamijo, M., Ido, Y., & Mirrlees, D. J. (1990a). Effects of long-term aldose
reductase inhibition on development of experimental diabetic neuropathy. Ultrastruc-
tural and morphometric studies of sural nerve in streptozocin-induced diabetic rats.
Diabetes, 39, 690–696.
Yagihashi, S., Kamijo, M., & Watanabe, K. (1990b). Reduced myelinated fiber size correlates
with loss of axonal neurofilaments in peripheral nerve of chronically streptozotocin
diabetic rats. The American Journal of Pathology, 136, 1365–1373.
Yagihashi, S., Mizukami, H., Ogasawara, S., Yamagishi, S., Nukada, H., Kato, N., et al.
(2010). The role of the polyol pathway in acute kidney injury caused by hindlimb ischae-
mia in mice. The Journal of Pathology, 220, 530–541.
Yagihashi, S., Mizukami, H., & Sugimoto, K. (2011). Mechanism of diabetic neuropathy:
Where are we now and where to go? Journal of Diabetes Investigation, 2, 18–32.
Yagihashi, S., Yamagishi, S. I., Wada, R., Baba, M., Hohman, T. C., Yabe-Nishimura, C.,
et al. (2001). Neuropathy in diabetic mice overexpressing human aldose reductase and
effects of aldose reductase inhibitor. Brain, 124, 2448–2458.
Yagihashi, S., Yamagishi, S., Wada, R., Sugimoto, K., Baba, M., Wong, H. G., et al. (1996).
Galactosemic neuropathy in transgenic mice for human aldose reductase. Diabetes, 45,
56–59.
Yamagishi, S., Nakamura, K., & Imaizumi, T. (2005). Advanced glycation end products
(AGEs) and diabetic vascular complications. Current Diabetes Reviews, 1, 93–106.
Yamagishi, S., Uehara, K., Otsuki, S., & Yagihashi, S. (2003). Differential influence of
increased polyol pathway on protein kinase C expressions between endoneurial and
epineurial tissues in diabetic mice. Journal of Neurochemistry, 87, 497–507.
Yamamoto, Y., Kato, I., Doi, T., Yonekura, H., Ohashi, S., Takeuchi, M., et al. (2001).
Development and prevention of advanced diabetic nephropathy in RAGE-
overexpressing mice. The Journal of Clinical Investigation, 108, 261–268.
Yeung, C. M., Lo, A. C., Cheung, A. K., Chung, S. S., Wong, D., & Chung, S. K. (2010).
More severe type 2 diabetes-associated ischemic stroke injury is alleviated in aldose
reductase-deficient mice. Journal of Neuroscience Research, 88, 2026–2034.
Zhang, L., Yu, C., Vasquez, F. E., Galeva, N., Onyango, I., Swerdlow, R. H., et al. (2010).
Hyperglycemia alters the Schwann cell mitochondrial proteome and decreased coupled
respiration in the absence of superoxide production. Journal of Proteome Research, 9,
458–471.
Zhang, L., Zhao, H., Blagg, B. S., & Dobrowsky, R. T. (2012). C-terminal heat shock pro-
tein 90 inhibitor decreases hyperglycemia-induced oxidative stress and improves mito-
chondrial bioenergetics in sensory neurons. Journal of Proteome Research, 11, 2581–2593.
Zherebitskaya, E., Akude, E., Smith, D. R., & Fernyhough, P. (2009). Development of
selective axonopathy in adult sensory neurons isolated from diabetic rats: Role of
glucose-induced oxidative stress. Diabetes, 58, 1356–1364.
Ziegler, D., & Gries, F. A. (1997). Alpha-lipoic acid in the treatment of diabetic peripheral
and cardiac autonomic neuropathy. Diabetes, 46(Suppl. 2), S62–S66.
Ziegler, D., Low, P. A., Litchy, W. J., Boulton, A. J., Vinik, A. I., Freeman, R., et al. (2011).
Efficacy and safety of antioxidant treatment with α-lipoic acid over 4 years in diabetic
polyneuropathy: The NATHAN 1 trial. Diabetes Care, 34, 2054–2060.
Ziegler, D., Reljanovic, M., Mehnert, H., & Gries, F. A. (1999). Alpha-lipoic acid in the
treatment of diabetic polyneuropathy in Germany: Current evidence from clinical trials.
Experimental and Clinical Endocrinology & Diabetes, 107, 421–430.
CHAPTER NINE
Sensory Neurodegeneration in
Diabetes: Beyond Glucotoxicity
D.W. Zochodne1
Neuroscience and Mental Health Institute and Alberta Diabetes Institute, University of Alberta,
Edmonton, AB, Canada
1
Corresponding author: e-mail address: [email protected]
Contents
1. Terminals at Risk: Sensory Neurodegeneration in Diabetes 152
2. Not Necessarily a Microvascular Disease 154
3. Altered Insulin Signaling 156
4. Ongoing Growth: Other Forms of Support 160
4.1 C-Peptide 160
4.2 Glucagon-Like Peptide-1 161
4.3 Heat-Shock Proteins 162
5. Neurons “on Edge” 162
6. Diabetes, Neurons, and Epigenetics 165
7. A Regeneration Strategy 169
8. Conclusions 174
Acknowledgments 174
References 174
Abstract
Diabetic polyneuropathy in humans is of gradual, sometimes insidious onset, and is
more likely to occur if glucose control is poor. Arguments that the disorder arises chiefly
from glucose toxicity however ignore the greater complexity of a unique neurodegen-
erative disorder. For example, sensory neurons regularly thrive in media with levels of
glucose at or exceeding those of poorly controlled diabetic persons. Also, all of the link-
ages between hyperglycemia and neuropathy develop in the setting of altered insulin
availability or sensitivity. Insulin itself is recognized as a potent growth, or trophic factor
for adult sensory neurons. Low doses of insulin, insufficient to alter blood glucose levels,
reverse features of diabetic neurodegeneration in animal models. Insulin resistance, as
occurs in diabetic adipose tissue, liver, and muscle, also develops in sensory neurons,
offering a mechanism for neurodegeneration in the setting of normal or elevated insulin
levels. Other interventions that “shore up” sensory neurons prevent features of diabetic
polyneuropathy from developing despite persistent hyperglycemia. More recently evi-
dence has emerged that a series of subtle molecular changes in sensory neurons can be
sensory loss and often accompanies it. Collectively, these findings have indi-
cated that neurons do not experience classical apoptosis early in diabetes and
that substantial dropout is not a feature of the disease. Thus, acute hypergly-
cemia or “glucotoxicity” in humans does not appear to lead to early and
massive neuronopathy. Overall, this information is important news for
DPN since it indicates that once DM is reversed by therapy or islet trans-
plantation, preserved parent neurons are available to reestablish connections
with terminals. Improvement in the sensory deficits of DPN may be
possible.
Why chronic DM exhibits this topographical distribution of change has
generated considerable thought. That it is a generalized feature of neu-
rodegeneration is supported by similar, albeit more rapid changes in ALS,
where motor terminals are retracted before the loss of anterior horn cells.
Several ideas are relevant to understanding this form of neuronal alteration.
Loss of distal terminals in models accompanies early key, but nonlethal alter-
ation of the perikaryal (cell body) gene output and protein synthesis (Cheng,
Kobayashi, Martinez, et al., 2015). For example, declines in the gene expres-
sion of neurofilament polymer subunits in the cell body accompany parallel
declines of similar magnitude in neurofilament investment of distal axons
(Scott, Clark, & Zochodne, 1999). A decline in neurofilaments, part of
the key internal structural lattice of the axon, within distal nerves accom-
panies distal axon atrophy. However, declines in neurofilament investment
probably do not completely account for a number of other features of DPN,
such as conduction velocity slowing. In mice lacking axonal neurofilaments,
diabetes caused accelerated conduction abnormalities, indicating a separate
alteration in membrane excitability (Zochodne, Sun, Cheng, & Eyer, 2004).
Since axons themselves are now recognized to translate proteins, declines in
perikaryal mRNAs targeted toward distal axonal ribosomes may similarly
connect subtle changes in cell body function with the behavior and later
retraction of terminals (Willis & Twiss, 2006). Three additional mechanisms
might account for distal axon targeting by diabetes. First, the intimate relation-
ship between Schwann cells (SCs) and axons, including the exchange of ribo-
somes, illustrates an important codependence (Court, Hendriks, MacGillavry,
Alvarez, & van Minnen, 2008). Axons support SCs with mitogenic molecules
such as neuregulin and CGRP, whereas SCs supply extracellular basement
membrane molecules that support axon growth and trophic factors that are
taken up and signal axons and their parent neurons. Second, considerable
attention has been devoted toward explaining DPN as a mitochondrio-
pathy with neuronal energy failure arising from oxidative stress and other
154 D.W. Zochodne
Fig. 1 Insulin directly ligates receptors on sensory neurons. In (A) sensory neurons and
axons in the lumbar dorsal root ganglia (DRG) of rats are labeled with an antibody
directed against IRβ, a subunit of the insulin receptor (IR) (bar ¼ 200 μm). In (B) DRG neu-
rons are labeled by FITC-labeled insulin that attached to the cell surface. The panel to
the right is the identical section viewed under light microscopy (bar ¼ 20 μm). Labeled
insulin was delivered by intrathecal injection and improved phenotypic features of
Sensory Neurodegeneration in Diabetes 159
experimental diabetic neuropathy. In (C) dissociated adult rat sensory neurons are
triple labeled with neurofilament antibody (red; gray in the print version), FITC-labeled
insulin (green; gray in the print version) added to the media, and DAPI (blue; gray in the
print version) illustrating that insulin is taken up by neurons and labels both the cyto-
plasm and nucleus of sensory neurons. Reproduced with permission from the American
Diabetes Association and Brussee, V., Cunningham, F. A., & Zochodne, D.W., 2004. Direct
insulin signaling of neurons reverses diabetic neuropathy. Diabetes, 53, 1824–1830.
160 D.W. Zochodne
The earlier work has established that insulin ligation of receptors at the
level of the perikaryon, or cell body influences the behavior of the entire
neuronal tree, repairing damage in distal axon segments. There is additional
evidence that distal axons also express the insulin receptor. During injury
and regeneration, receptors were identified on regrowing axons just beyond
the injury site (Xu et al., 2004). Singhal, Cheng, Sun, and Zochodne (1997)
injected low subhypoglycemic doses of insulin around sciatic nerves of rats
with experimental diabetes and compared their function with contralateral
limb sciatic nerves exposed to carrier. Despite ongoing evidence of hyper-
glycemia, and progressive DPN in the opposite limb, nerves exposed to
local insulin had improvements in electrophysiology and had a rise in the
number of small myelinated axons. Dermal and epidermal axons also express
the insulin receptor and mRNAs for downstream insulin transduction
pathways, IRS-1 and IRS-2, are found in skin. As discussed earlier, this
supports the idea that this “normal” population of axons is in a growth state,
constantly remodeling to keep apace of keratinocyte turnover. Guo, Kan,
Martinez, and Zochodne (2011) injected small dermal doses of insulin,
insufficient to alter systemic glucose levels into the hind paw of mice with
chronic DM of 5 months duration. The contralateral paws in the same
mice were injected with carrier. Local insulin, but not carrier, unilaterally
improved epidermal innervation of the hind paw over a surprisingly
short period of within 1 week. Improvements occurred in both type 1
and 2 diabetic mice. These findings support the concept that ongoing
plasticity of skin innervation offers opportunities for short-term manipula-
tion of regrowth to improve neurological function. Chen, Calcutt, and col-
leagues (Chen et al., 2013) have also demonstrated similar short-term
plasticity, over 4 weeks, of nerve fibers in the subbasal plexus of cornea
in rats. DM in this model was associated with a progressive decline in
corneal innervation, whereas local administration of insulin over the cornea
prevented loss of corneal axons.
for chronic therapy in diabetic patients. These patients may have already
been exposed to daily systemic injection or they may exhibit systemic insulin
resistance. In patients with established DPN, often first presenting to
clinicians, reversing DPN has usually not been possible and features of
DPN persist despite improvements in glucose control. Experimentally, it
also appears difficult to reverse features of DPN in models with established
disease (Kan, Guo, Singh, Singh, & Zochodne, 2012). For all of these
reasons, moving downstream of insulin, or identifying approaches to sup-
plement its actions may be important. For example, extensive work on
C-peptide, the normally cleaved bridging peptide of proinsulin, suggests that
it benefits DPN through an insulin-sensitizing action. C-peptide has evi-
dence of benefit in models and early human trials (Ekberg et al., 2003;
Kamiya, Zhang, Ekberg, Wahren, & Sima, 2006; Zhang et al., 2001).
STZ mice untreated for 5 months. These mice exhibit key phenotypic
features of DPN including motor and sensory conduction slowing, loss of
sensation to thermal and mechanical stimuli, and loss of epidermal innerva-
tion. GW/P bodies are ultrastructural cytoplasmic complexes that contain
key elements of RISC. Their expression may identify neurons under
“stress.” We noted that GW/P expression was heightened in the sensory
neurons of mice with DM, indicating overt structural evidence of altered
mRNA processing as a feature of this chronic disease (Cheng et al., 2015).
In the same cohort of diabetic mice and littermates, we examined 28,869
DRG mRNAs for differential expression of at least a 1.5-fold change in
DM samples and within these identified 261 mRNAs that included
91 upregulated and 170 downregulated. Of these 24 achieved a statistical
difference between diabetics and nondiabetics of p < 0.05 (5 down and
19 up). Almost all coded for proteins of unknown function in sensory
neurons or diabetes. For example, one upregulated molecule, CWC22, pro-
vided a valuable lead to analyze spliceosome function in diabetic sensory
neurons, work in progress that offers new ideas about specific molecular
deficits in diabetic sensory neurons (Kobayashi, Cheng, de la Hoz, &
Zochodne, 2015). We further explored concurrent changes in miRNAs,
their supraregulatory companions. As in the case of mRNAs, we identified
a number of differentially expressed miRNAs in DRGs from mice with
chronic DM and DPN. Of 1042 examined, there were 19 altered that
included 12 downregulated and 7 upregulated high-abundance miRNAs.
Additionally, there were 123 low-abundance miRNAs altered including
56 downregulated and 67 upregulated. Focusing on miRNAs in the
high-abundance group, we studied expression of mmu-let-7i, which was
downregulated by 39% in diabetic DRGs, a change also confirmed by
qRT-PCR. mmu-let-7i was an interesting miRNA to consider first, given
widely divergent impacts on over 900 targets. To begin with, we assessed
whether this miRNA was expressed in neurons, rather than DRG satellite
cells or vessels. By in situ hybridization, we noted striking expression in most
sensory neurons of the DRG (Fig. 2). To determine whether alterations in
mmu-let-7i might be associated with an overall phenotype, given its diver-
gent actions, we studied the impact of sensory neuron transfection with
an exogenous mmu-let-7i mimic in vitro. The approach was associated
with robust trophic actions including an increase in neurite outgrowth
and branching. Collectively these findings suggested that the downregu-
lation of mmu-let-7i in chronic diabetic DRG neurons might impair overall
supportive or trophic mechanisms.
Fig. 2 Epigenetic miRNAs are expressed in sensory neurons and influence their growth
properties. In (A), in situ hybridization images of normal mouse DRGs identify mmu-
let-7i expressed in sensory neurons at lower (left) and higher power (right) (bar ¼ 100
and 50 μm, respectively). A control image without label is below. In (B) are illustrated
confocal images of preinjured (axotomized) adult rat sensory neurons harvested from
lumbar DRG and grown in the presence of control carrier solution (top images) or a
mimic molecule of mmu-let-7i for 20 h and stained with antineurofilament antibody
(bar ¼ 100 μm). Reproduced with permission from Cheng, C., Kobayashi, M., Martinez, J. A.,
et al. (2015). Evidence for epigenetic regulation of gene expression and function in chronic
experimental diabetic neuropathy. Journal of Neuropathology and Experimental Neurology,
74, 804–817.
168 D.W. Zochodne
Identifying the specific and relevant target genes responsible for a growth
phenotype in sensory neurons is challenging. A comparison of parallel micro-
arrays, in the same diabetic and nondiabetic mouse cohorts, examining statis-
tically significant changes in mRNAs due to diabetes with four of our altered
miRNAs identified some predicted changes. For example, these included
upregulation of connective tissue growth factor, upregulation of dual-
specifity phosphatase 1, upregulation of F3 and TXNIP (a thioredoxin-
interacting protein that may mediate oxidative stress), a small rise in insulin
receptor, and downregulation of CACNG4, a subunit of the calcium L-type
channel. Most have unclarified relationships to sensory neuron function
to date. Two previous reports have linked abnormalities in calcium
channels to experimental diabetes (Hall, Sima, & Wiley, 1995; Voitenko,
Kruglikov, Kostyuk, & Kostyuk, 2000). These predicted changes were only
based on four high-abundance miRNA changes, suggesting that much more
extensive interactions occur than we explored in this work. However, none
of the apparent alterations mediated by these mRNAs offered clear or
established candidates for growth enhancement.
The next step in addressing the relevance of miRNA changes in chronic
DM is understanding whether these changes represent an epiphenomenon
of the disease or whether they effect critical changes in neuron function.
Given confirmation that mmu-let-7i was downregulated in DRGs with
clear impacts on sensory neuron biology, we tested whether supplementing
neurons with exogenous mmu-let-7i, in the form of a mimic molecule,
might alter the DPN phenotype. The approach to this was not trivial given
that access of the nucleotide to sensory perikarya would be key to its poten-
tial actions. A viral vector approach to transfection may be a potent option
for stable replenishment of mmu-let-7i. However, with a thought toward
downstream human translation, we tested a nonviral approach that involved
intranasal administration of an mmu-let-7i mimic molecule. To test whether
miRNAs might access the CSF and DRGs through their root sleeves by this
approach, we first administered an unrelated miRNA by intranasal
delivery—a plant species miRNA–Arabidopsis thaliana miR171, not present
in mammals. After administration for 6 days, miR171 was detected in both
the olfactory bulb and the lumbar DRG of mice indicating access to the CSF
through this route. Next we administered a mimic mmu-let-7i by the
intranasal route and confirmed that in nondiabetic mice, daily dosing for
1 week could raise mmu-let-7i levels as tested in DRG by qRT-PCR.
Given the evidence that a nonconventional route of miRNA delivery
had the potential to access the CNS and spinal spaces including DRG root
Sensory Neurodegeneration in Diabetes 169
sleeves, we tested its impact on diabetic mice. Using mice with 5-month
duration DM and well-established indices of DPN, we administered intra-
nasal mmu-let-7i mimic over 6 days and retested after a further 3 weeks. The
rationale was that alterations in gene expression effected by mmu-let-7i
might require at least 2–3 weeks to alter epidermal innervation and other
features of DPN. Intranasal mmu-let-7i, but not control treatment,
improved impaired diabetic mechanical sensitivity to the levels of mice
without DM and improved loss of thermal sensation, both accompanied
by an improvement in epidermal innervation. Motor and sensory conduc-
tion velocities were also improved by the mmu-let-7i mimic. Additional
work examined whether knockdown of a second separate, but in this case,
elevated miRNA mmu-341, in diabetic mice might also impact DPN. In
contrast to mmu-let7i, mmu-341 was the most robustly upregulated
miRNA detected in our array survey. In this case contrary knockdown
with an miRNA anti-miR also improved features of DPN, although the
results were less robust, influencing only sensory conduction and thermal
sensation. Taken together, however, these findings identified a striking
impact of nucleotide delivery on structure, electrophysiology, and behavior
in diabetic mice.
Epigenetic manipulation of a range of mRNA targets altered in DM is an
attractive “single bullet” that might be available to repair neuropathic dam-
age. Albeit promising, particularly in the use of nonviral delivery, the
approach needs independent confirmation. The overall possibilities linked
to manipulating gene alterations in neurological disease using intranasal
access to the CSF are also enormous, but require verification. Despite these
caveats, the observations that relatively short-term interventions, presum-
ably targeting altered but not lost parent neurons, might allow recovery from
deficits that have developed chronically over several months are exciting.
There are other facets of epigenetic regulation in DM that may be equally
important, including targeting of SCs, also key targets of DM, that are not
discussed in this review.
7. A REGENERATION STRATEGY
The axon terminals of adult sensory neurons might be considered as
being in a permanent and ongoing state of growth. Within the epidermis,
the constant movement, replenishment, and shedding of keratinocytes
require that axons adapt and remodel to retain their roles. Indeed, the tra-
jectories of epidermal axons, while well spaced, are highly irregular, with
170 D.W. Zochodne
twists and turns that suggest ongoing growth. More direct evidence arises
from their prominent expression of growth proteins such as GAP43 and
the finding that interventions, including simple and noninvasive steps such
as hair clipping, can dramatically influence the density of innervation
(Cheng et al., 2010).
In chronic DM with DPN, epidermal terminals are retracted, requiring
regrowth to restore their innervation. Collateral sprouting, distinct from
regenerative sprouting, allows reinnervation of denervated territories by
invasion from intact neighbor axons. Work by Diamond and colleagues
(Diamond, Coughlin, Macintyre, Holmes, & Visheau, 1987; Diamond,
Holmes, & Coughlin, 1992) has shown that skin collateral sprouting, but
not regenerative sprouting, is NGF dependent. Collateral sprouting does
occur in DM (Theriault, Dort, Sutherland, & Zochodne, 1998), but this
form of recovery generally may not be sufficiently robust to completely
restore sensory fidelity. With this caveat in mind, retracted axons in DM
may be called upon to regrow into their previous territories, forestalling
ingrowth by neighbors. Taken together, strategies that support both forms
of neurological recovery may be required.
In DM, there are other considerations that make regenerative strategies
important. Patients with DM are disposed to develop single nerve lesions, or
focal neuropathies such as carpal tunnel syndrome or ulnar neuropathy at the
elbow (Zochodne, 2007). A more exacting clinical terminology thus refers
to diabetic neuropathies, recognizing a range of additional complications
involving the peripheral nervous system. Focal neuropathies are common
and disabling. Considering the range of neurological complications identi-
fied, DM imposes a “double hit,” a degenerative polyneuropathy, or DPN,
but also a deficit in regenerative capacity. For example, in carpal tunnel syn-
drome, surgical decompression of the entrapped nerve at the wrist and sub-
sequent recovery has a less satisfactory outcome in DM.
Mechanisms for impaired diabetic nerve regeneration have been consid-
ered extensively and have included impaired neurotrophic support, micro-
angiopathy that renders the regenerative microenvironment unsupportive,
local oxidative stress, impaired macrophage clearance, accelerated retrograde
loss of neurons, polyol flux, mitochondrial dysfunction, nonenzymatic gly-
cosylation of basement membrane regeneration scaffolds, SC dysfunction,
and others (Kennedy & Zochodne, 2005). New molecular approaches to
coax greater plasticity and regrowth out of reluctant adult neurons have
highlighted the potential roles of targeting their intrinsic growth mecha-
nisms. These operate downstream of growth factor signals and may be final
Sensory Neurodegeneration in Diabetes 171
animals displayed regeneration deficits with significantly fewer and smaller caliber
axons regenerating 3 weeks after injury. PTEN inhibition partially rescued the deficit
(bar ¼ 50 μm). In (D) are representative immunohistochemically labeled (PGP 9.5)
images of footpads (*external surface of the skin) from wild type and diabetics with
and without PTEN siRNA indicating newly regenerating sensory afferents in the epider-
mis. Reinnervation was reduced in diabetics with few reinnervating axons crossing
into the epidermis and improved with PTEN siRNA (bar ¼ 100 μm). Reproduced with
permission from Singh, B., Singh, V., Krishnan, A., et al. Regeneration of diabetic axons is
enhanced by selective knockdown of the PTEN gene. Brain, 137, 1051–1067.
174 D.W. Zochodne
8. CONCLUSIONS
Exploiting novel molecular approaches to support sensory neurons in
chronic DM may offer a palette of therapeutic targets. These are urgently
required in a field that has suffered from a series of failed clinical trials. Mov-
ing beyond “microvascular” ideas might include approaches to offer direct
neuronal insulin signaling through new delivery routes, adding GLP-1
agonism to the mix and supporting neurons with HSP27 or inhibitors of
caspase-3, PARP, or AGE–RAGE signaling. New epigenetic understand-
ing of the pathogenesis of this complex disorder is warranted. Finally, new
strategies that support the regenerative potential of peripheral neurons dam-
aged by DM may be essential to restore neurological function in patients.
ACKNOWLEDGMENTS
The work highlighted in this review was supported by operating grants from the Canadian
Institutes of Health Research (FRN184584), the Canadian Diabetes Association (OG-3-12-
3669), the Juvenile Diabetes Research Foundation, and the National Institutes of Health
(NIDDK), USA. The author has been supported by the Alberta Heritage Foundation for
Medical Research and the University Hospital Foundation, Edmonton. The Zochodne
laboratory is supported by the Neuroscience and Mental Health Institute, Alberta
Diabetes Institute, Division of Neurology and Department of Medicine, University of
Alberta.
REFERENCES
Ahn, K. S., & Aggarwal, B. B. (2005). Transcription factor NF-kappaB: A sensor for smoke
and stress signals. The Annals of the New York Academy of Sciences, 1056, 218–233.
Baggio, L. L., & Drucker, D. J. (2007). Biology of incretins: GLP-1 and GIP. Gastroenterology,
132, 2131–2157.
Bennett, G. J., Doyle, T., & Salvemini, D. (2014). Mitotoxicity in distal symmetrical sensory
peripheral neuropathies. Nature Reviews. Neurology, 10, 326–336.
Brownlee, M., Cerami, A., & Vlassara, H. (1988). Advanced glycosylation end products in
tissue and the biochemical basis of diabetic complications. The New England Journal of
Medicine, 318, 1315–1321.
Brussee, V., Cunningham, F. A., & Zochodne, D. W. (2004). Direct insulin signaling of
neurons reverses diabetic neuropathy. Diabetes, 53, 1824–1830.
Cameron, N. E., Cotter, M. A., & Low, P. A. (1991). Nerve blood flow in early experimen-
tal diabetes in rats: Relation to conduction deficits. The American Journal of Physiology,
261, E1–E8.
Cameron, N. E., Gibson, T. M., Nangle, M. R., & Cotter, M. A. (2005). Inhibitors of
advanced glycation end product formation and neurovascular dysfunction in experimen-
tal diabetes. The Annals of the New York Academy of Sciences, 1043, 784–792.
Chen, D. K., Frizzi, K. E., Guernsey, L. S., Ladt, K., Mizisin, A. P., & Calcutt, N. A. (2013).
Repeated monitoring of corneal nerves by confocal microscopy as an index of peripheral
Sensory Neurodegeneration in Diabetes 175
neuropathy in type-1 diabetic rodents and the effects of topical insulin. Journal of the
Peripheral Nervous System, 18, 306–315.
Cheng, C., Guo, G. F., Martinez, J. A., Singh, V., & Zochodne, D. W. (2010). Dynamic
plasticity of axons within a cutaneous milieu. The Journal of Neuroscience, 30,
14735–14744.
Cheng, C., Kobayashi, M., Martinez, J. A., et al. (2015). Evidence for epigenetic regulation
of gene expression and function in chronic experimental diabetic neuropathy. Journal of
Neuropathology and Experimental Neurology, 74, 804–817.
Cheng, C., Webber, C. A., Wang, J., et al. (2008). Activated RHOA and peripheral axon
regeneration. Experimental Neurology, 212, 358–369.
Cheng, C., & Zochodne, D. W. (2003). Sensory neurons with activated caspase-3 survive
long-term experimental diabetes. Diabetes, 52, 2363–2371.
Christie, K. J., Krishnan, A., Martinez, J. A., et al. (2014). Enhancing adult nerve regener-
ation through the knockdown of retinoblastoma protein. Nature Communications, 5,
3670.
Christie, K. J., Webber, C. A., Martinez, J. A., Singh, B., & Zochodne, D. W. (2010). PTEN
inhibition to facilitate intrinsic regenerative outgrowth of adult peripheral axons. The
Journal of Neuroscience, 30, 9306–9315.
Court, F. A., Hendriks, W. T., MacGillavry, H. D., Alvarez, J., & van Minnen, J.
(2008). Schwann cell to axon transfer of ribosomes: Toward a novel understanding
of the role of glia in the nervous system. The Journal of Neuroscience, 28(43),
11024–11029.
Diamond, J., Coughlin, M., Macintyre, L., Holmes, M., & Visheau, B. (1987). Evidence that
endogenous beta nerve growth factor is responsible for the collateral sprouting, but not
the regeneration, of nociceptive axons in adult rats. Proceedings of the National Academy of
Sciences of the United States of America, 84, 6596–6600.
Diamond, J., Holmes, M., & Coughlin, M. (1992). Endogenous NGF and nerve impulses
regulate the collateral sprouting of sensory axons in the skin of the adult rat. The Journal
of Neuroscience, 12, 1454–1466.
Drel, V. R., Pacher, P., Vareniuk, I., et al. (2007). A peroxynitrite decomposition catalyst
counteracts sensory neuropathy in streptozotocin-diabetic mice. European Journal of Phar-
macology, 569, 48–58.
Ekberg, K., Brismar, T., Johansson, B. L., Jonsson, B., Lindstrom, P., & Wahren, J. (2003).
Amelioration of sensory nerve dysfunction by C-peptide in patients with type 1 diabetes.
Diabetes, 52, 536–541.
Fernyhough, P., & McGavock, J. (2014). Mechanisms of disease: Mitochondrial dysfunction
in sensory neuropathy and other complications in diabetes. Handbook of Clinical Neurol-
ogy, 126, 353–377.
Fernyhough, P., Smith, D. R., Schapansky, J., et al. (2005). Activation of nuclear factor-
kappaB via endogenous tumor necrosis factor alpha regulates survival of axotomized
adult sensory neurons. The Journal of Neuroscience, 25, 1682–1690.
Fernyhough, P., Willars, G. B., Lindsay, R. M., & Tomlinson, D. R. (1993). Insulin and
insulin-like growth factor I enhance regeneration in cultured adult rat sensory neurones.
Brain Research, 607, 117–124.
Folli, F., Bonfanti, L., Renard, E., Kahn, C. R., & Merighi, A. (1994). Insulin receptor sub-
strate-1 (IRS-1) distribution in the rat central nervous system. The Journal of Neuroscience,
14, 6412–6422.
Francis, G. J., Martinez, J. A., Liu, W. Q., et al. (2011). Motor end plate innervation loss in
diabetes and the role of insulin. Journal of Neuropathology and Experimental Neurology, 70,
323–339.
Frazier, W. A., Angeletti, R. H., & Bradshaw, R. A. (1972). Nerve growth factor and insulin.
Science, 176, 482–488.
176 D.W. Zochodne
Giniger, E. (2002). How do Rho family GTPases direct axon growth and guidance?
A proposal relating signaling pathways to growth cone mechanics. Differentiation, 70,
385–396.
Greenbaum, D., Richardson, P. C., Salmon, M. V., & Urich, H. (1964). Pathological obser-
vations on six cases of diabetic neuropathy. Brain, 87, 201–214.
Grote, C. W., Groover, A. L., Ryals, J. M., Geiger, P. C., Feldman, E. L., & Wright, D. E.
(2013). Peripheral nervous system insulin resistance in ob/ob mice. Acta Neuropathologica
Communications, 1, 15.
Grote, C. W., Morris, J. K., Ryals, J. M., Geiger, P. C., & Wright, D. E. (2011). Insulin
receptor substrate 2 expression and involvement in neuronal insulin resistance in diabetic
neuropathy. Experimental Diabetes Research, 2011, 212571.
Guo, G., Kan, M., Martinez, J. A., & Zochodne, D. W. (2011). Local insulin and the rapid
regrowth of diabetic epidermal axons. Neurobiology of Disease, 43, 414–421.
Guo, G., Singh, V., & Zochodne, D. W. (2014). Growth and turning properties of adult glial
cell-derived neurotrophic factor coreceptor alpha 1 nonpeptidergic sensory neurons.
Journal of Neuropathology and Experimental Neurology, 73(9), 820–836.
Hall, K. E., Sima, A. A., & Wiley, J. W. (1995). Voltage-dependent calcium currents are
enhanced in dorsal root ganglion neurones from the Bio Bred/Worchester diabetic
rat. Journal of Physiology, 486, 313–322.
Himeno, T., Kamiya, H., Naruse, K., et al. (2011). Beneficial effects of exendin-4 on exper-
imental polyneuropathy in diabetic mice. Diabetes, 60, 2397–2406.
Huang, T. J., Price, S. A., Chilton, L., et al. (2003). Insulin prevents depolarization of the
mitochondrial inner membrane in sensory neurons of type 1 diabetic rats in the presence
of sustained hyperglycemia. Diabetes, 52, 2129–2136.
Jin, M., Guan, C. B., Jiang, Y. A., et al. (2005). Ca2+-dependent regulation of rho GTPases
triggers turning of nerve growth cones. The Journal of Neuroscience, 25, 2338–2347.
Jolivalt, C. G., Fineman, M., Deacon, C. F., Carr, R. D., & Calcutt, N. A. (2011). GLP-1
signals via ERK in peripheral nerve and prevents nerve dysfunction in diabetic mice.
Diabetes, Obesity & Metabolism, 13, 990–1000.
Kamiya, H., Zhang, W., Ekberg, K., Wahren, J., & Sima, A. A. (2006a). C-peptide reverses
nociceptive neuropathy in type 1 diabetes. Diabetes, 55, 3581–3587.
Kamiya, H., Zhang, W., & Sima, A. A. (2006b). Degeneration of the Golgi and neuronal loss
in dorsal root ganglia in diabetic BioBreeding/Worcester rats. Diabetologia, 49,
2763–2774.
Kamiya, H., Zhangm, W., & Sima, A. A. (2005). Apoptotic stress is counterbalanced by sur-
vival elements preventing programmed cell death of dorsal root ganglions in subacute
type 1 diabetic BB/Wor rats. Diabetes, 54, 3288–3295.
Kan, M., Guo, G., Singh, B., Singh, V., & Zochodne, D. W. (2012). Glucagon-like peptide
1, insulin, sensory neurons, and diabetic neuropathy. Journal of Neuropathology and Exper-
imental Neurology, 71, 494–510.
Kennedy, J. M., & Zochodne, D. W. (2002). Influence of experimental diabetes on the
microcirculation of injured peripheral nerve. Functional and morphological aspects.
Diabetes, 51, 2233–2240.
Kennedy, J. M., & Zochodne, D. W. (2005). Impaired peripheral nerve regeneration in
diabetes mellitus. Journal of the Peripheral Nervous System, 10, 144–157.
Kim, B., McLean, L. L., Philip, S. S., & Feldman, E. L. (2011). Hyperinsulinemia induces
insulin resistance in dorsal root ganglion neurons. Endocrinology, 152, 3638–3647.
Kobayashi, M., Cheng, C., de la Hoz, C., & Zochodne, D. (2015). Diabetic neuropathy
and the sensory neuron: New molecular targets. American Academy of Neurology, P2.010.
Korngut, L., Ma, C. H., Martinez, J. A., et al. (2012). Overexpression of human HSP27 pro-
tects sensory neurons from diabetes. Neurobiology of Disease, 47, 436–443.
Sensory Neurodegeneration in Diabetes 177
Korngut, L., Mawani, S., Francis, G., et al. (2013). A pilot study of intranasal insulin for
the treatment of diabetic peripheral neuropathy. Journal of the Peripheral Nervous System,
18, S59.
Korthals, J. K., Gieron, M. A., & Dyck, P. J. (1988). Intima of epineurial arterioles is
increased in diabetic polyneuropathy. Neurology, 38, 1582–1586.
Levy, D., Hoke, A., & Zochodne, D. W. (1999). Local expression of inducible nitric oxide
synthase in an animal model of neuropathic pain. Neuroscience Letters, 260, 207–209.
Levy, D., Kubes, P., & Zochodne, D. W. (2001). Delayed peripheral nerve degeneration,
regeneration, and pain in mice lacking inducible nitric oxide synthase. Journal of Neuro-
pathology and Experimental Neurology, 60, 411–421.
Levy, D., Tal, M., Hoke, A., & Zochodne, D. W. (2000). Transient action of the endothelial
constitutive nitric oxide synthase (ecNOS) mediates the development of thermal hyper-
sensitivity following peripheral nerve injury. The European Journal of Neuroscience, 12,
2323–2332.
Luo, L., Jan, L. Y., & Jan, Y. N. (1997). Rho family GTP-binding proteins in growth cone
signalling. Current Opinion in Neurobiology, 7, 81–86.
Ma, J., Farmer, K. L., Pan, P., et al. (2014). Heat shock protein 70 is necessary to
improve mitochondrial bioenergetics and reverse diabetic sensory neuropathy
following KU-32 therapy. The Journal of Pharmacology and Experimental Therapeutics,
348, 281–292.
Ma, J., Pan, P., Anyika, M., Blagg, B. S., & Dobrowsky, R. T. (2015). Modulating
molecular chaperones improves mitochondrial bioenergetics and decreases the inflam-
matory transcriptome in diabetic sensory neurons. ACS Chemical Neuroscience, 6,
1637–1648.
Malik, R. A., Kallinikos, P., Abbott, C. A., et al. (2003). Corneal confocal microscopy:
A non-invasive surrogate of nerve fibre damage and repair in diabetic patients.
Diabetologia, 46, 683–688.
Malik, R. A., Newrick, P. G., Sharma, A. K., et al. (1989). Microangiopathy in human
diabetic neuropathy: Relationship between capillary abnormalities and the severity of
neuropathy. Diabetologia, 32, 92–102.
Malik, R. A., Veves, A., Masson, E. A., et al. (1992). Endoneurial capillary abnormalities in
mild human diabetic neuropathy. Journal of Neurology, Neurosurgery, and Psychiatry, 55,
557–561.
Massa, S. M., Xie, Y., Yang, T., et al. (2006). Small, nonpeptide p75NTR ligands induce
survival signaling and inhibit proNGF-induced death. The Journal of Neuroscience, 26,
5288–5300.
Newrick, P. G., Wilson, A. J., Jakubowski, J., Boulton, A. J., & Ward, J. D. (1986). Sural
nerve oxygen tension in diabetes. British Medical Journal, 293, 1053–1054.
Ng, J., & Luo, L. (2004). Rho GTPases regulate axon growth through convergent and diver-
gent signaling pathways. Neuron, 44, 779–793.
Obrosova, I. G., Drel, V. R., Pacher, P., et al. (2005). Oxidative-nitrosative stress and
poly(ADP-ribose) polymerase (PARP) activation in experimental diabetic neuropathy:
The relation is revisited. Diabetes, 54, 3435–3441.
Obrosova, I. G., Li, F., Abatan, O. I., et al. (2004). Role of poly(ADP-ribose) polymerase
activation in diabetic neuropathy. Diabetes, 53, 711–720.
Perry, T., Holloway, H. W., Weerasuriya, A., et al. (2007). Evidence of GLP-1-mediated
neuroprotection in an animal model of pyridoxine-induced peripheral sensory neurop-
athy. Experimental Neurology, 203, 293–301.
Polydefkis, M., Hauer, P., Griffin, J. W., & McArthur, J. C. (2001). Skin biopsy as a tool to
assess distal small fiber innervation in diabetic neuropathy. Diabetes Technology & Thera-
peutics, 3, 23–28.
178 D.W. Zochodne
Purves, T. D., & Tomlinson, D. R. (2002). Diminished transcription factor survival signals in
dorsal root ganglia in rats with streptozotocin-induced diabetes. Annals of the New York
Academy of Sciences, 973, 472–476.
Reddy, V. P., & Beyaz, A. (2006). Inhibitors of the Maillard reaction and AGE breakers as
therapeutics for multiple diseases. Drug Discovery Today, 11, 646–654.
Reger, M. A., Watson, G. S., Green, P. S., et al. (2008). Intranasal insulin improves cognition
and modulates B-amyloid in early AD. Neurology, 70, 440–448.
Rundles, R. W. (1945). Diabetic neuropathy—General review with report of 125 cases.
Medicine, 24, 111–160.
Russell, J. W., Sullivan, K. A., Windebank, A. J., Herrmann, D. N., & Feldman, E. L. (1999).
Neurons undergo apoptosis in animal and cell culture models of diabetes. Neurobiology of
Disease, 6, 347–363.
Saleh, A., Smith, D. R., Tessler, L., et al. (2013). Receptor for advanced glycation end-
products (RAGE) activates divergent signaling pathways to augment neurite outgrowth
of adult sensory neurons. Experimental Neurology, 249, 149–159.
Schmelzer, J. D., Zochodne, D. W., & Low, P. A. (1989). Ischemic and reperfusion injury of
rat peripheral nerve. Proceedings of the National Academy of Sciences of the United States of
America, 86, 1639–1642.
Schmidt, R. E., Beaudet, L. N., Plurad, S. B., & Dorsey, D. A. (1997). Axonal cytoskeletal
pathology in aged and diabetic human sympathetic autonomic ganglia. Brain Research,
769, 375–383.
Schmidt, R. E., Dorsey, D. A., Beaudet, L. N., Plurad, S. B., Parvin, C. A., & Bruch, L. A.
(1998). Vacuolar neuritic dystrophy in aged mouse superior cervical sympathetic ganglia
is strain-specific. Brain Research, 806, 141–151.
Schmidt, R. E., Dorsey, D., Parvin, C. A., Beaudet, L. N., Plurad, S. B., & Roth, K. A.
(1997). Dystrophic axonal swellings develop as a function of age and diabetes in human
dorsal root ganglia. Journal of Neuropathology and Experimental Neurology, 56, 1028–1043.
Schmidt, R. E., & Scharp, D. W. (1982). Axonal dystrophy in experimental diabetic auto-
nomic neuropathy. Diabetes, 31, 761–770.
Scott, J. N., Clark, A. W., & Zochodne, D. W. (1999). Neurofilament and tubulin gene
expression in progressive experimental diabetes: Failure of synthesis and export by
sensory neurons. Brain, 122, 2109–2118.
Singh, B., Singh, V., Krishnan, A., et al. (2014). Regeneration of diabetic axons is enhanced
by selective knockdown of the PTEN gene. Brain, 137, 1051–1067.
Singh, B., Xu, Y., Guo, G. F., Cunningham, S., Martinez, J. A., & Zochodne, D. W. (2009).
Insulin signaling and insulin resistance in sensory neurons. Journal of the Peripheral Nervous
System, 13, 254.
Singh, B., Xu, Y., McLaughlin, T., et al. (2012). Resistance to trophic neurite outgrowth of
sensory neurons exposed to insulin. Journal of Neurochemistry, 121, 263–276.
Singhal, A., Cheng, C., Sun, H., & Zochodne, D. W. (1997). Near nerve local insulin
prevents conduction slowing in experimental diabetes. Brain Research, 763, 209–214.
Sivitz, W. I., & Yorek, M. A. (2010). Mitochondrial dysfunction in diabetes: From molecular
mechanisms to functional significance and therapeutic opportunities. Antioxidants &
Redox Signaling, 12, 537–577.
Sugimoto, K., Murakawa, Y., & Sima, A. A. (2002). Expression and localization of insulin
receptor in rat dorsal root ganglion and spinal cord. Journal of the Peripheral Nervous System,
7, 44–53.
Sugimoto, K., Murakawa, Y., Zhang, W., Xu, G., & Sima, A. A. (2000). Insulin receptor in
rat peripheral nerve: Its localization and alternatively spliced isoforms. Diabetes/Metabo-
lism Research and Reviews, 16, 354–363.
Tavakoli, M., Quattrini, C., Abbott, C., et al. (2010). Corneal confocal microscopy: A novel
noninvasive test to diagnose and stratify the severity of human diabetic neuropathy.
Diabetes Care, 33, 1792–1797.
Sensory Neurodegeneration in Diabetes 179
Tesfaye, S., Chaturvedi, N., Eaton, S. E., et al. (2005). Vascular risk factors and diabetic neu-
ropathy. The New England Journal of Medicine, 352, 341–350.
Tesfaye, S., Harris, N., Jakubowski, J. J., et al. (1993). Impaired blood flow and arteriovenous
shunting in human diabetic neuropathy: A novel technique of nerve photography and
fluorescein angiography. Diabetologia, 36, 1266–1274.
The Diabetes Control and Complications Trial Research Group. (1993). The effect of inten-
sive treatment of diabetes on the development and progression of long-term complica-
tions in insulin-dependent diabetes mellitus. The Diabetes Control and Complications
Trial Research Group. The New England Journal of Medicine, 329, 977–986.
Theriault, M., Dort, J., Sutherland, G., & Zochodne, D. W. (1997). Local human sural nerve
blood flow in diabetic and other polyneuropathies. Brain, 120, 1131–1138.
Theriault, M., Dort, J., Sutherland, G., & Zochodne, D. W. (1998). A prospective
quantitative study of sensory deficits after whole sural nerve biopsies in diabetic and
nondiabetic patients. Surgical approach and the role of collateral sprouting. Neurology,
50, 480–484.
Thomsen, K., Rubin, I., & Lauritzen, M. (2002). NO- and non-NO-, non-prostanoid-
dependent vasodilatation in rat sciatic nerve during maturation and developing
experimental diabetic neuropathy. The Journal of Physiology, 543, 977–993.
Thornalley, P. J. (2003). Use of aminoguanidine (Pimagedine) to prevent the formation
of advanced glycation endproducts. Archives of Biochemistry and Biophysics, 419,
31–40.
Toth, C., Brussee, V., Martinez, J. A., McDonald, D., Cunningham, F. A., &
Zochodne, D. W. (2006). Rescue and regeneration of injured peripheral nerve axons
by intrathecal insulin. Neuroscience, 139, 429–449.
Toth, C., Francis, G., Martinez, J. A., Hanson, L., Frey, W., III, & Zochodne, D. W. (2007).
Intranasal insulin delivery reduces morphological, electrophysiological, behavioral, and
molecular changes of diabetic neuropathy without change in glycemia. Journal of the
Peripheral Nervous System, 12(Suppl.), 86.
Toth, C., Schmidt, A. M., Tuor, U., Brussee, V., Kaur, J., & Zochodne, D. W. (2004).
RAGE in the nervous system: Insights into a new pathophysiological relationship to dia-
betic complications within the central and peripheral nervous system. Canadian Journal of
Neurological Sciences, 31(Suppl. 1), S13.
Tucker, B. A., Rahimtula, M., & Mearow, K. M. (2006). Laminin and growth factor recep-
tor activation stimulates differential growth responses in subpopulations of adult DRG
neurons. The European Journal of Neuroscience, 24, 676–690.
Vareniuk, I., Pacher, P., Pavlov, I. A., Drel, V. R., & Obrosova, I. G. (2009). Peripheral
neuropathy in mice with neuronal nitric oxide synthase gene deficiency. International
Journal of Molecular Medicine, 23, 571–580.
Vareniuk, I., Pavlov, I. A., & Obrosova, I. G. (2008). Inducible nitric oxide synthase gene
deficiency counteracts multiple manifestations of peripheral neuropathy in a
streptozotocin-induced mouse model of diabetes. Diabetologia, 51, 2126–2133.
Voitenko, N. V., Kruglikov, I. A., Kostyuk, E. P., & Kostyuk, P. G. (2000). Effect of
streptozotocin-induced diabetes on the activity of calcium channels in rat dorsal horn
neurons. Neuroscience, 95, 519–524.
Willis, D. E., & Twiss, J. L. (2006). The evolving roles of axonally synthesized proteins in
regeneration. Current Opinion in Neurobiology, 16, 111–118.
Xu, Q. G., Li, X.-Q., Kotecha, S. A., Cheng, C., Sun, H. S., & Zochodne, D. W. (2004).
Insulin as an in vivo growth factor. Experimental Neurology, 188, 43–51.
Yasuda, H., & Dyck, P. J. (1987). Abnormalities of endoneurial microvessels and sural nerve
pathology in diabetic neuropathy. Neurology, 37, 20–28.
Zhang, W., Yorek, M., Pierson, C. R., Murakawa, Y., Breidenbach, A., & Sima, A. A.
(2001). Human C-peptide dose dependently prevents early neuropathy in the BB/
Wor-rat. International Journal of Experimental Diabetes Research, 2, 187–193.
180 D.W. Zochodne
Zherebitskaya, E., Akude, E., Smith, D. R., & Fernyhough, P. (2009). Development of
selective axonopathy in adult sensory neurons isolated from diabetic rats: Role of
glucose-induced oxidative stress. Diabetes, 58, 1356–1364.
Zochodne, D. W. (2002). Nerve and ganglion blood flow in diabetes: An appraisal.
In D. Tomlinson (Ed.), Neurobiology of diabetic neuropathy (pp. 161–202). San Diego:
Academic Press.
Zochodne, D. W. (2007). Diabetes mellitus and the peripheral nervous system: Manifesta-
tions and mechanisms. Muscle & Nerve, 36, 144–166.
Zochodne, D. W. (2014). Mechanisms of diabetic neuron damage: Molecular pathways.
Handbook of Clinical Neurology, 126, 379–399.
Zochodne, D. W., & Ho, L. T. (1992). Normal blood flow but lower oxygen tension in
diabetes of young rats: Microenvironment and the influence of sympathectomy.
Canadian Journal of Physiology and Pharmacology, 70, 651–659.
Zochodne, D. W., Levy, D., Zwiers, H., et al. (1999). Evidence for nitric oxide and nitric
oxide synthase activity in proximal stumps of transected peripheral nerves. Neuroscience,
91, 1515–1527.
Zochodne, D. W., & Nguyen, C. (1999). Increased peripheral nerve microvessels in early
experimental diabetic neuropathy: Quantitative studies of nerve and dorsal root ganglia.
Journal of the Neurological Sciences, 166, 40–46.
Zochodne, D. W., Ramji, N., & Toth, C. (2008). Neuronal targeting in diabetes mellitus:
A story of sensory neurons and motor neurons. The Neuroscientist, 14, 311–318.
Zochodne, D. W., Sun, H.-S., Cheng, C., & Eyer, J. (2004). Accelerated diabetic neurop-
athy in axons without neurofilaments. Brain, 127, 2193–2200.
Zochodne, D. W., Verge, V. M., Cheng, C., Sun, H., & Johnston, J. (2001). Does diabetes
target ganglion neurons? Progressive sensory neuron involvement in long term
experimental diabetes. Brain, 124, 2319–2334.
CHAPTER TEN
Contents
1. Introduction 182
2. Molecular Chaperones 183
3. Extracellular Hsp70 185
3.1 Import, Export, and Neuronal Support 185
3.2 Immunomodulation, Inflammation, and Oxidative Stress 186
4. Intracellular Hsp70 189
4.1 Chaperone Functions 189
4.2 Oxidative Stress 190
4.3 Inflammation 194
4.4 Insulin Sensitivity, JNK, and c-Jun 195
5. DPN and Modulating Hsp70 197
6. Concluding Remarks 201
Acknowledgments 201
References 201
Abstract
The etiology of diabetic peripheral neuropathy (DPN) involves an interrelated series of
metabolic and vascular insults that ultimately contribute to sensory neuron degenera-
tion. In the quest to pharmacologically manage DPN, small-molecule inhibitors have
targeted proteins and pathways regarded as “diabetes specific” as well as others whose
activity are altered in numerous disease states. These efforts have not yielded any
significant therapies, due in part to the complicating issue that the biochemical
contribution of these targets/pathways to the progression of DPN does not occur with
temporal and/or biochemical uniformity between individuals. In a complex, chronic
neurodegenerative disease such as DPN, it is increasingly appreciated that effective
disease management may not necessarily require targeting a pathway or protein
considered to contribute to disease progression. Alternatively, it may prove sufficiently
beneficial to pharmacologically enhance the activity of endogenous cytoprotective
pathways to aid neuronal tolerance to and recovery from glucotoxic stress. In pursuing
this paradigm shift, we have shown that modulating the activity and expression of
molecular chaperones such as heat shock protein 70 (Hsp70) may provide translational
potential for the effective medical management of insensate DPN. Considerable evi-
dence supports that modulating Hsp70 has beneficial effects in improving inflamma-
tion, oxidative stress, and glucose sensitivity. Given the emerging potential of
modulating Hsp70 to manage DPN, the current review discusses efforts to characterize
the cytoprotective effects of this protein and the benefits and limitations that may arise
in drug development efforts that exploit its cytoprotective activity.
1. INTRODUCTION
Over 300 million individuals worldwide are estimated to be affected
by diabetes, with projections that exceed half a billion people by 2030
(Whiting, Guariguata, Weil, & Shaw, 2011). About half of these individuals
will develop diabetic peripheral neuropathy (DPN). DPN symptoms can be
quite varied and manifest in the extremities as weakness/numbness (insensate
DPN) or painful tingling, burning, or lancinating sensations (painful DPN).
Though the painful aspects of DPN can be quite debilitating, the loss of sen-
sation can lead to life-altering amputations by exacerbating the seriousness of
ulcerations that occur in 15% of patients. In fact, diabetes is the leading cause
of nonaccidental lower extremity amputations. Current therapies manage
painful symptoms with anticonvulsants, antidepressants, or opiates. How-
ever, pharmacologic approaches to manage insensate DPN have not realized
any significant clinical success. To this point, management of glycemic con-
trol is our most effective option in ameliorating the onset and severity of
DPN symptoms, but this approach is more effective for Type 1 than Type
2 diabetics (Callaghan, Cheng, Stables, Smith, & Feldman, 2012).
The recurrent hyperglycemia central to the development of DPN results
in a host of biochemical and metabolic changes. Elevated blood glucose
levels can increase its flux through the polyol and hexosamine pathways,
activate protein kinase C (PKC), increase poly(ADP-ribose) polymerase
activity, and enhance formation of advanced glycation endproducts
(AGE), which activate AGE receptors (RAGE) to heighten oxidative stress
(Brownlee, 2001; Edwards, Vincent, Cheng, & Feldman, 2008; Farmer,
Li, & Dobrowsky, 2012). Though each pathway is likely to independently
contribute to the metabolic alterations that underlie DPN, current theories
suggest that the progression of DPN is strongly related to a convergence of
these metabolic disturbances on promoting oxidative stress (Obrosova et al.,
Modulating Hsp70 to Treat DPN 183
2005; Stavniichuk et al., 2011; Vareniuk et al., 2007) and mitochondrial dys-
function (Chowdhury, Smith, & Fernyhough, 2013; Farmer et al., 2012).
Additionally, inflammatory signaling, a well-accepted component of Type
2 diabetes, is also fed by oxidative stress and can play a role in exacerbating
DPN through cytokine production, immune cell infiltration, and further
elevation of oxidative stress (Wei et al., 2009; Wellen, 2005; Wright,
Scism-Bacon, & Glass, 2006).
Classic approaches to pharmacologically manage DPN have assumed
that targeting a single biochemical insult is sufficient to alter the interwoven
molecular cascade that culminates in the onset of symptomatic sensory
neuropathy and the eventual dying back of distal axons. If DPN arose from
damage to a single cell population from a single biochemical insult, this
approach may be highly fruitful. However, the natural history of the
disorder develops over decades due to damage to vascular, glial, and neu-
ronal cell populations that are not necessarily affected synchronously by
hyperglycemic stress. Indeed, despite the current focus on minimizing
oxidative stress, antioxidant therapies have also met with limited success
in treating DPN, though the use of α-lipoic acid may have some benefit
(Garcia-Alcala et al., 2015; Ziegler et al., 2006, 2011). Thus, realigning our
thinking toward pharmacologic approaches to address this problem may
prove beneficial.
A rather unexplored and clinically unappreciated alternative approach
for managing DPN is to deemphasize that it is necessary to topple a bio-
chemical kingpin to destroy the downstream network of molecular minions
whose slavings contribute to disease progression. On the other hand, cells
have evolved endogenous mechanisms to tolerate stress. Pharmacologic
upregulation of these pathways may facilitate recovery from glucotoxicity
and/or enhance the efficacy of agents that target direct pathogenetic mech-
anisms of DPN. Growing evidence suggests that modulating endogenous
cytoprotective responses by targeting the activity and expression of molec-
ular chaperones such as heat shock protein 90 (Hsp90) and Hsp70 may
achieve this purpose.
2. MOLECULAR CHAPERONES
Heat shock proteins are divided into families defined by their molec-
ular size: Hsp110, Hsp100, Hsp90, Hsp70, Hsp60, Hsp40, and small Hsps
(Kim, Hipp, Bracher, Hayer-Hartl, & Hartl, 2013; Saibil, 2013). This
184 S.M. Emery and R.T. Dobrowsky
review will focus primarily on the Hsp90 and Hsp70 proteins that can be
found in the endoplasmic reticulum (ER), mitochondria, cytosol, and extra-
cellularly (Becker & Craig, 1994; Haas & Wabl, 1983; Hightower &
Guidon, 1989; Leustek, Dalie, Amir-Shapira, Brot, & Weissbach, 1989).
Both Hsp90 and Hsp70 have isoforms that are constitutively expressed or
induced by thermal, ischemic, oxidative, and metabolic stressors, as well as
by exercise (Beckmann, Lovett, & Welch, 1992; Krause et al., 2007;
Richard, Kaeffer, & Thuillez, 1996; Yang et al., 1996). Under physiologic
conditions Hsp90 binds and inhibits the transcription factor heat shock fac-
tor 1 (HSF1). In response to the accumulation of damaged and/or unfolded
proteins, HSF1 is released from this complex, undergoes trimerization, and
is phosphorylated before being translocated to the nucleus (Neef, Jaeger, &
Thiele, 2011; Vihervaara & Sistonen, 2014). Upon binding to heat shock
elements, HSF1 upregulates an array of inducible chaperones including
forms of Hsp70 (Hsp70.1 and Hsp70.3) and Hsp90 (Hsp90α), as well as
antioxidant proteins (Benarroch, 2011). Hsp70 can help refold unfolded
proteins, prevent inappropriate protein interactions, manage protein aggre-
gate formation, and direct the degradation of damaged or dysfunctional pro-
teins. Importantly, the efficacy of modulating chaperones is not limited to
neurodegenerative diseases driven by the formation of protein aggregates.
Hsp70 has also been shown to downregulate inflammatory signaling,
decrease oxidative stress, and improve mitochondrial function in DPN
and other conditions whose etiology is not linked to the formation of pro-
tein aggregates (Ianaro et al., 2003; Jones, Voegeli, Li, Chen, & William
Currie, 2011; Li, Ma, Zhao, Blagg, & Dobrowsky, 2012; Ma, Pan,
Anyika, Blagg, & Dobrowsky, 2015; Madden, Sandstrom, Lovell, &
McNaughton, 2008; Saibil, 2013; Zhang, Zhao, Blagg, & Dobrowsky,
2012). However, an important aspect of Hsp70 biology that must be con-
sidered in moving forward in therapy development is how modulating the
intra- vs extracellular pools of Hsp70 may affect disease progression.
Opposing effects have been demonstrated for intracellular Hsp70
(iHsp70) vs extracellular Hsp70 (eHsp70) in multiple studies (Krause
et al., 2015; Rodrigues-Krause et al., 2012). Extracellular Hsps were first
described as glia–axon transfer proteins when giant squid axons were sub-
jected to heat stress (Tytell, Greenberg, & Lasek, 1986). Later studies spe-
cifically identified Hsp70 as one of the glia–axon transfer proteins
(Hightower & Guidon, 1989). More recently, eHsp70 has been shown in
blood where it acts as a paracrine factor released from dendritic, neuronal,
and other cell types (De Maio, 2011). eHsp70 is associated with immune
Modulating Hsp70 to Treat DPN 185
3. EXTRACELLULAR Hsp70
3.1 Import, Export, and Neuronal Support
Hsp70 export was unknowingly first characterized in heat-treated giant
squid axon when protein levels of a heat shock-like transferrin protein
increased in the axoplasm (Tytell et al., 1986). This transferrin protein
was specifically identified as Hsp70 when heat-treated rat embryonic cell
cultures (presumably fibroblasts) were shown to release Hsp70 into the
media, independent of an increase in cell death (Hightower & Guidon,
1989). Glial eHsp70 secretion has also been confirmed in models of heat-
treated glioblastoma cells (Guzhova et al., 2001).
The export mechanisms behind eHsp70 secretion are still not well
understood since Hsp70 lacks a secretory signal sequence and inhibiting
the secretory pathway has no effect on the release of eHsp70
(Hightower & Guidon, 1989). Some evidence has pointed toward an innate
ability of the protein to traverse the cell membrane (Multhoff, 2007), lipid
raft-mediated lysosomal release (Hunter-Lavin et al., 2004), and secretory-
like granule excretion (Evdonin et al., 2006). However, the largest collec-
tion of evidence points to exosome-dependent trafficking; heat-shocked
peripheral blood mononuclear cells increased eHsp70 levels and produced
Hsp70-loaded exosomes (Lancaster & Febbraio, 2005). Though further
study is needed to fully understand this Hsp70 export mechanism, exosomal
eHsp70 release has been demonstrated in other models including colon
and pancreatic cancer cell lines (Gastpar et al., 2005) as well as breast and
leukemic cancer cells (Bausero, Gastpar, Multhoff, & Asea, 2005).
186 S.M. Emery and R.T. Dobrowsky
et al., 2007; Ortega, Hinchado, Martin-Cordero, & Asea, 2009; Senf et al.,
2013). However, eHsp70 can also indirectly increase chemotaxis of immune
cells by stimulating T-cell-mediated beta-chemokine release of Rantes and
macrophage inflammatory protein-1β, which drive infiltration of numerous
innate and adaptive immune components (Lehner et al., 2000). Once B- and
T-lymphocytes have infiltrated the inflammatory site, they may use eHsp70
itself as a chemoattractant. In vitro experiments with and without heat shock
have demonstrated lymphocyte export of eHsp70 (Hunter-Lavin et al.,
2004). Similarly, increases in macrophage phagocytosis and neutrophil
microbicide activity have been reported after exogenous Hsp70 administra-
tion (Kovalchin et al., 2006; Ortega et al., 2006). eHsp70 also prompted
proinflammatory cytokine release from epithelial, innate immune, and adap-
tive immune cells, which may explain why eHsp70 is associated with inflam-
matory cytokine production in multiple mouse and human studies (Asea et al.,
2000, 2002; Basu, Binder, Ramalingam, & Srivastava, 2001; Chase et al.,
2007; Dvoriantchikova, Santos, Saeed, Dvoriantchikova, & Ivanov, 2014;
Dybdahl et al., 2005; Huang, Wang, Chen, Wang, & Zhang, 2013; Qiao,
Liu, & Li, 2008). Collectively, these data support a strong capacity of eHsp70
to initiate and escalate an immune-mediated inflammatory response.
A growing list of receptors seem to mediate the immunomodulatory
actions of eHsp70 by evoking a host of proinflammatory cytokines
(IL-1β, IL-6, IL-8, IL-12, TNF-α) through a CD14-dependent, Toll-like
receptor (TLR) 2/4 mechanism (Asea et al., 2000, 2002; Chase et al.,
2007; Dvoriantchikova et al., 2014; Dybdahl, 2001). TLR2 has also been
linked to eHsp70 stimulation of neutrophil chemotaxis (Ortega et al.,
2009). Similarly, CD94, a C-type lectin receptor, has been demonstrated
to mediate eHsp70 stimulation of IFN-γ release from NK cells (Gross,
Hansch, Gastpar, & Multhoff, 2003). However, a separate report indicated
that this response required coculturing the NK cells with dendritic cells that
produce NK G2D ligand after eHsp70 treatment (Qiao et al., 2008). The
differences in these studies might be clarified by their use of patient-derived
NK cells vs the YT leukemic NK cell line. CD40 was determined to be nec-
essary for mycobacterial-Hsp70-driven beta-chemokine release and human
eHsp70 was demonstrated to interact with CD40 (Lehner et al., 2000). The
CD40-eHsp70-dependent effects were expanded to include internalization
of eHsp70–peptide complexes for antigen presentation in macrophages
(Becker, 2002; Lehner et al., 2000; Wang et al., 2001). Surprisingly, an anal-
ogous study identified CD91, not CD40, as managing antigen presentation
in macrophages and dendritic cells through the MHC class I molecule by
188 S.M. Emery and R.T. Dobrowsky
4. INTRACELLULAR Hsp70
4.1 Chaperone Functions
The molecular chaperone activity of intracellular heat shock proteins has
been extensively characterized (De Maio, 2011). iHsp70 is an integral com-
ponent of this chaperone system and the complex cochaperone interactions
and molecular mechanisms that facilitate this process have been described in
numerous reviews (Becker & Craig, 1994; Kim et al., 2013; Saibil, 2013;
Stetler et al., 2010). A major function of this chaperone activity is to main-
tain intracellular proteostasis by assisting nascent peptide folding, refolding of
unfolded proteins, preventing inappropriate protein interactions, managing
protein aggregate formation, and degrading dysfunctional proteins (Saibil,
2013). These activities have particular relevance in neurodegenerative
diseases, which are often associated with aggregates of misfolded proteins,
including Alzheimer’s disease, Parkinson’s disease, ALS, Huntington’s
disease, and others (Turturici et al., 2011). It is believed that postmitotic
neurons affected in these pathologies are particularly vulnerable due to an
inability to diminish misfolded protein concentrations through cell division
(Muchowski & Wacker, 2005). Consequently, enhancing iHsp70 action
might prove a viable therapeutic approach to treating these pathologies.
In vitro, iHsp70 increases the solubility and microtubule interaction of
the tau protein, as well as early-stage aggregation of Aβ that leads to amyloid
plaque formation in Alzheimer’s disease (Dou et al., 2003; Evans, Wisen, &
Gestwicki, 2006). However, managing inappropriate protein interactions is
only part of the solution to minimizing misfolded protein stress. If a protein
cannot be refolded to a functional state, then it must be degraded to prevent
accumulation.
SOD1 is a cytosolic free radical scavenger protein that is integral to man-
aging oxidative stress and mutant SOD1 promotes mitochondrial dysfunc-
tion that contributes to the onset of familial ALS (Tan, Pasinelli, & Trotti,
2014). CHIP is a C-terminal Hsp70-interacting protein that facilitates
ubiquitination and degradation of misfolded iHsp70 client proteins like
mutant SOD1. Mutant SOD1 is also degraded by chaperone-mediated
autophagy where regulator proteins such as Bcl-2-associated athanogene
3 (BAG-3) and Hsp22 direct iHsp70’s chaperone functions to identify, traf-
fic, and present misfolded proteins for autophagic degradation (Kalmar,
Lu, & Greensmith, 2014). This orchestrated removal of dysfunctional pro-
tein is crucial for neurological homeostasis, as well as metabolic disorders
190 S.M. Emery and R.T. Dobrowsky
connection between Hsp70 and SOD1 expression was surmised since cere-
brum homogenates from these animals showed a marked decrease in SOD1
protein levels and activity (Choi et al., 2005). Interestingly, this regulation
may not be unidirectional, since SOD1-overexpressing transgenic mice
have elevated levels of Hsp70 mRNA following cerebral ischemia
(Kondo et al., 1996). Nevertheless, iHsp70 seems to have a capacity to mod-
ulate SOD1 expression. Evidence for Hsp70 regulation of SOD2 is not so
clear-cut, however. Hsp70 knockout mice also have decreased SOD2 activ-
ity, but these mice do not show changes in SOD2 protein expression levels
(Choi et al., 2005). This difference between SOD2 activity and expression
may be explained by two observations. First, iHsp70 has been found to traf-
fic SOD2 to the mitochondria (Afolayan et al., 2014) and second, SOD2
requires additional processing in the mitochondria before it becomes fully
functional (Candas & Li, 2014). As a result, genetic deletion of iHsp70
would prevent SOD2 from being efficiently localized to the mitochondria
for activation. Consistent with this premise, pharmacological induction of
Hsp70 in hyperglycemically stressed rat embryonic sensory neurons
increased mitochondrial SOD2 expression, as determined by quantitative
mass spectrometry (Zhang et al., 2012).
Another major role iHsp70 plays in managing oxidative stress involves
supporting proper mitochondrial function. Mitochondria are a major source
of ROS production within the cell, having a concentration 5- to 10-fold
higher than in the cytosol (Cadenas & Davies, 2000). Left unchecked, intra-
cellular stress can overload or impair the mitochondrial machinery, decrease
protein import, and ultimately result in mitochondrial dysfunction and sig-
nificant increases in oxidative stress (Baseler et al., 2011; Tomlinson &
Gardiner, 2008). For instance, hyperglycemic stress of embryonic sensory
neurons decreased the translation of numerous mitochondrial proteins
and mitochondrial oxygen consumption while increasing superoxide levels.
However, increasing iHsp70 levels with a C-terminal Hsp90 inhibitor
decreased superoxide production and improved mitochondrial bioenerget-
ics (Zhang et al., 2012). Further studies from our lab have demonstrated that
pharmacologically modulating Hsp70 has a beneficial effect on mitochon-
drial bioenergetics (Ma et al., 2014; Urban et al., 2012; Zhang et al.,
2012). Interestingly, RNA-Seq analysis indicated that the mitochondrial
transcriptome was unaltered by pharmacological modulation of Hsp70 in
diabetic mice, despite an improvement in mitochondrial bioenergetics.
Therefore iHsp70’s protection of mitochondrial bioenergetics may not be
significantly mediated through gene transcription (Ma et al., 2015).
192 S.M. Emery and R.T. Dobrowsky
Fig. 1 Modulating Hsp70 with KU-596 decreased the expression of txnip in diabetic DRG
in an Hsp70-dependent manner. Wild-type (A) and Hsp70 KO (B) mice were rendered
diabetic with streptozotocin and after 12 weeks of diabetes, treated with vehicle or
weekly doses of KU-596 at 20 mg/kg for 4 weeks. mRNA was harvested from the lumbar
DRG and used for RNA-Seq analysis. Shown are representative alignments to the mouse
txnip gene from nondiabetic, diabetic, and diabetic animals that received KU-596 ther-
apy. Dark blue (black in the print version) bars indicate sequences that mapped to the
forward strand, while dark red (dark gray in the print version) bars are sequences that
mapped to the reverse strand of the eight exons of the txnip gene. See Ma et al., 2015 for
additional details.
194 S.M. Emery and R.T. Dobrowsky
stress insult, these results suggest that Hsp70 attenuates intracellular oxidative
stress induced by diabetes, which correlates with an improvement in clini-
cally relevant measures of DPN.
4.3 Inflammation
Another major aspect of iHsp70 biology is its ability to regulate inflamma-
tory signaling. Heat shock prevents TNF-α production after ischemic stress
in the renal tubular epithelial cell line LLC-PK1 and in rat liver after induc-
tion of sepsis by cecal ligation and puncture (CLP; Chen, Kuo, Wang, Lu, &
Yang, 2005; Meldrum, 2003). In vivo, Hsp70 overexpression suppresses
increased expression of IL-6 and TNF-α induced by LPS, as well as elevation
of IL-6 and cytokine-induced neutrophil chemoattractant-1 in rat blood
after CLP (Dokladny, Lobb, Wharton, Ma, & Moseley, 2010; Weiss
et al., 2007). Each of these studies reported that an Hsp70-dependent sup-
pression of NF-κB activation mediated the antiinflammatory action. Using
DNA binding and nuclear localization, numerous studies have supported
this claim with heat shock (Feinstein et al., 1996), liposomal Hsp70 delivery
(Meldrum, 2003), and Hsp70 overexpression (Feinstein, Galea, & Reis,
1997). Mechanistically, this effect may be due to Hsp70-dependent decrease
in degradation of the NF-κB inhibitor (IκBα), which binds NF-κB to
inhibit its nuclear localization (Dokladny et al., 2010; Feinstein et al.,
1997; Wong, Ryan, & Wispe, 1997). Precipitating this action is Hsp70’s
ability to inhibit phosphorylation of the IκB kinase (IKK). IKK phosphor-
ylates IκBα, causing it to dissociate from NF-κB and be degraded. In the
absence of IKK phosphorylation, this process is blocked (Chan, Ou,
Wang, & Chan, 2004; Chung et al., 2008; Weiss et al., 2007). Immunopre-
cipitation studies demonstrate that Hsp70 associates with the IKK complex,
which hinders the IKKβ subunit of the complex from being integrated
(Weiss et al., 2007). Additional studies support Hsp70 binding to IKKγ
(NEMO), which prevents its trimerization and function as a structural com-
ponent of the IKK complex (Agou et al., 2002; Chen et al., 2005; Ran
et al., 2004).
A second Hsp70 mechanism for hindering NF-κB activation has also
been described since Hsp70 stabilized phosphorylated and ubiquitinated
IκBα. This interaction prevented the degradation of IκBα without affecting
the IKK complex (Weiss et al., 2007). Additionally, a separate report sug-
gests a third mechanism where Hsp70 directly upregulates IκBα expression
to suppress NF-κB activation (Wong, Ryan, Menendez, & Wispe, 1999).
Modulating Hsp70 to Treat DPN 195
2012; Urban et al., 2010). KU-32 and KU-596 are two such compounds
that were mentioned above and these drugs improved multiple clinical
indices of DPN in models of both Type 1 and Type 2 diabetes, in an
Hsp70-dependent manner (Ma et al., 2014, 2015; Urban et al., 2010,
2012). Similarly, an N-terminal Hsp90 inhibitor has proven beneficial in
treating diabetic nephropathy and the physiologic improvement in renal
function may also be Hsp70 dependent (Lazaro et al., 2015). As neither
of these diabetic complications has an etiology that centers on formation
of any one protein aggregate, clearly modulating Hsp70 may prove effica-
cious toward treating complex metabolic complications of diabetes.
Although all these compounds were shown to increase iHsp70, their effects
on eHsp70 levels were not examined. Consistent with many of the studies
described in Section 4, novologues diminished the level of oxidative stress,
improved mitochondrial bioenergetics, and decreased an array of genes in
DRG that were associated with inflammation (Ma et al., 2014, 2015;
Urban et al., 2012; Zhang et al., 2012). The improvement of DPN and
decrease in inflammation by KU-596 therapy was also associated with a
transcriptomic profile that was predicted to inhibit the NF-κB pathway
in DRG of diabetic wild-type mice but not diabetic Hsp70 KO mice
(Ma et al., 2015). These actions would be inconsistent with enhancing sys-
temic levels of eHsp70, but it is possible that a more localized exchange of
eHsp70 between Schwann cells and sensory neurons may contribute to the
dependence of drug effectiveness on Hsp70.
It is interesting that increasing Hsp70 in skeletal muscle using the HSF1
activator, BGP-15, improved fasting glucose and insulin signaling in obese
mice (Henstridge, Bruce, et al., 2014; Henstridge, Whitham, et al., 2014).
However, novologues improved DPN in an Hsp70-dependent manner
without correcting any metabolic parameters in models of both Type 1
and Type 2 diabetes. Similarly, the N-terminal Hsp90 inhibitor and
geldanamycin derivative (DMAG) improved diabetic nephropathy without
effecting metabolic measures of diabetes: weight loss, blood glucose, and
HbA1c (Lazaro et al., 2015). The reason for the difference between these
compounds is curious, but may lie in BGP-15 being a more direct activator
of HSF1. It is encouraging that no adverse effects were reported from a short
28-day clinical trial in patients given 200 or 400 mg BGP-15 daily, which
may be consistent with minimal drug effect on increasing eHsp70 activity
(Literáti-Nagy et al., 2009). As treatment of diabetic complications such
as DPN will require prolonged drug administration, it will be important
to assess any effects on eHsp70 as novel modulators of Hsp70 move forward
Modulating Hsp70 to Treat DPN 201
6. CONCLUDING REMARKS
In summary, results in animal models and small clinical trials clearly
provide compelling proof of concept that modulating molecular chaperones
and particularly iHsp70 provides a viable approach to improve insulin resis-
tance and diabetic complications. Although we are not putting forth the pre-
mise that directly targeting proteins and pathways that are linked to the
pathophysiological progression of DPN is a less than fruitful approach to dis-
ease management, it seems that altering any one pathway may be insufficient
to treat the human disease. A similar fate may befall an approach that only
modulates iHsp70 to treat DPN. However, since modulating Hsp70
improves oxidative stress, mitochondrial function, and to some extent
inflammation, increasing the ability of neurons and Schwann cells to tolerate
ongoing diabetic stress may facilitate the action of additional agents or
improve the ability of glycemic control to slow the rate of onset and dimin-
ish the severity of DPN, especially in Type 2 diabetic patients.
ACKNOWLEDGMENTS
This work was supported by Grant DK095911 from the National Institute of Diabetes,
Digestive, and Kidney Diseases to R.T.D. and a postdoctoral fellowship from the
Pharmaceutical Research and Manufacturers of America Foundation to S.M.E.
REFERENCES
Afolayan, A. J., Teng, R. J., Eis, A., Rana, U., Broniowska, K. A., Corbett, J. A., et al.
(2014). Inducible Hsp70 regulates superoxide dismutase-2 and mitochondrial oxidative
stress in the endothelial cells from developing lungs. American Journal of Physiology. Lung
Cellular and Molecular Physiology, 306, L351–L360.
Agou, F., Ye, F., Goffinont, S., Courtois, G., Yamaoka, S., Israel, A., et al. (2002). Nemo
trimerizes through its coiled-coil C-terminal domain. Journal of Biological Chemistry, 277,
17464–17475.
Arthur-Farraj, P. J., Latouche, M., Wilton, D. K., Quintes, S., Chabrol, E., Banerjee, A.,
et al. (2012). C-jun reprograms Schwann cells of injured nerves to generate a repair cell
essential for regeneration. Neuron, 75, 633–647.
Asea, A., Kraeft, S. K., Kurt-Jones, E. A., Stevenson, M. A., Chen, L. B., Finberg, R. W.,
et al. (2000). Hsp70 stimulates cytokine production through a CD14-dependent path-
way, demonstrating its dual role as a chaperone and cytokine. Nature Medicine, 6,
435–442.
202 S.M. Emery and R.T. Dobrowsky
Asea, A., Rehli, M., Kabingu, E., Boch, J. A., Bare, O., Auron, P. E., et al. (2002). Novel
signal transduction pathway utilized by extracellular Hsp70: Role of toll-like receptor
(TLR) 2 and TLR4. Journal of Biological Chemistry, 277, 15028–15034.
Baseler, W. A., Dabkowski, E. R., Williamson, C. L., Croston, T. L., Thapa, D.,
Powell, M. J., et al. (2011). Proteomic alterations of distinct mitochondrial subpopula-
tions in the Type 1 diabetic heart: Contribution of protein import dysfunction. American
Journal of Physiology. Regulatory, Integrative and Comparative Physiology, 300, R186–R200.
Basu, S., Binder, R. J., Ramalingam, T., & Srivastava, P. K. (2001). CD91 is a common
receptor for heat shock proteins GRP96, Hsp90, Hsp70, and calreticulin. Immunity,
14, 303–313.
Bausero, M. A., Gastpar, R., Multhoff, G., & Asea, A. (2005). Alternative mechanism by
which IFN-g enhances tumor recognition: Active release of heat shock protein 72. Jour-
nal of Immunology, 175, 2900–2912.
Becker, T. (2002). CD40, an extracellular receptor for binding and uptake of Hsp70-peptide
complexes. Journal of Cell Biology, 158, 1277–1285.
Becker, J., & Craig, E. A. (1994). Heat-shock proteins as molecular chaperones. European
Journal of Biochemistry, 219, 11–23.
Beckmann, R. P., Lovett, M., & Welch, W. J. (1992). Examining the function and regulation
of Hsp70 in cells subjected to metabolic stress. Journal of Cell Biology, 117, 1137–1150.
Bellmann, K., Jaattela, M., Wissing, D., Burkart, V., & Kolb, H. (1996). Heat shock protein
Hsp70 overexpression confers resistance against nitric oxide. FEBS Letters, 391, 185–188.
Bellmann, K., Wenz, A., Radons, J., Burkart, V., Kleemann, R., & Kolb, H. (1995). Heat
shock induces resistance in rat pancreatic islet cells against nitric oxide, oxygen radicals
and streptozotocin toxicity in vitro. Journal of Clinical Investigation, 95, 2840–2845.
Benarroch, E. E. (2011). Heat shock proteins: Multiple neuroprotective functions and impli-
cations for neurologic disease. Neurology, 76, 660–667.
Bocanegra, V., Manucha, W., Pena, M. R., Cacciamani, V., & Valles, P. G. (2010).
Caveolin-1 and Hsp70 interaction in microdissected proximal tubules from spontane-
ously hypertensive rats as an effect of losartan. Journal of Hypertension, 28, 143–155.
Broome, C. S., Kayani, A. C., Palomero, J., Dillmann, W. H., Mestril, R., Jackson, M. J.,
et al. (2006). Effect of lifelong overexpression of Hsp70 in skeletal muscle on age-related
oxidative stress and adaptation after nondamaging contractile activity. FASEB Journal,
20, 1549–1551.
Brownlee, M. (2001). Biochemistry and molecular cell biology of diabetic complications.
Nature, 414, 813–820.
Cadenas, E., & Davies, K. J. (2000). Mitochondrial free radical generation, oxidative stress,
and aging. Free Radical Biology and Medicine, 29, 222–230.
Callaghan, B. C., Cheng, H. T., Stables, C. L., Smith, A. L., & Feldman, E. L. (2012).
Diabetic neuropathy: Clinical manifestations and current treatments. Lancet Neurology,
11, 521–534.
Campisi, J., Leem, T. H., & Fleshner, M. (2003). Stress-induced extracellular Hsp72 is a
functionally significant danger signal to the immune system. Cell Stress & Chaperones,
8, 272–286.
Candas, D., & Li, J. J. (2014). MnSOD in oxidative stress response-potential regulation via
mitochondrial protein influx. Antioxidants & Redox Signaling, 20, 1599–1617.
Chan, J. Y., Ou, C. C., Wang, L. L., & Chan, S. H. (2004). Heat shock protein 70 confers
cardiovascular protection during endotoxemia via inhibition of Nuclear Factor-KappaB
activation and inducible nitric oxide synthase expression in the rostral ventrolateral
medulla. Circulation, 110, 3560–3566.
Chase, M. A., Wheeler, D. S., Lierl, K. M., Hughes, V. S., Wong, H. R., & Page, K. (2007).
Hsp72 induces inflammation and regulates cytokine production in airway epithelium
Modulating Hsp70 to Treat DPN 203
Dybdahl, B., Slordahl, S. A., Waage, A., Kierulf, P., Espevik, T., & Sundan, A. (2005). Myo-
cardial ischaemia and the inflammatory response: Release of heat shock protein 70 after
myocardial infarction. Heart, 91, 299–304.
Ebrahimi-Fakhari, D., Saidi, L. J., & Wahlster, L. (2013). Molecular chaperones and protein
folding as therapeutic targets in Parkinson’s disease and other synucleinopathies. Acta
Neuropathologica Communications, 1, 79.
Edwards, J. L., Vincent, A. M., Cheng, H. T., & Feldman, E. L. (2008). Diabetic neuropathy:
Mechanisms to management. Pharmacology and Therapeutics, 120, 1–34.
Evans, C. G., Chang, L., & Gestwicki, J. E. (2010). Heat shock protein 70 (Hsp70) as an
emerging drug target. Journal of Medicinal Chemistry, 53, 4585–4602.
Evans, C. G., Wisen, S., & Gestwicki, J. E. (2006). Heat shock proteins 70 and 90 inhibit
early stages of amyloid beta-(1-42) aggregation in vitro. Journal of Biological Chemistry,
281, 33182–33191.
Evdonin, A. L., Martynova, M. G., Bystrova, O. A., Guzhova, I. V., Margulis, B. A., &
Medvedeva, N. D. (2006). The release of Hsp70 from A431 carcinoma cells is mediated
by secretory-like granules. European Journal of Cell Biology, 85, 443–455.
Farmer, K. L., Li, C., & Dobrowsky, R. T. (2012). Diabetic peripheral neuropathy: Should a
chaperone accompany our therapeutic approach? Pharmacological Reviews, 64, 880–900.
Feinstein, D. L., Galea, E., Aquino, D. A., Li, G. C., Xu, H., & Reis, D. J. (1996). Heat shock
protein 70 suppresses astroglial-inducible nitric-oxide synthase expression by decreasing
NFkappaB activation. Journal of Biological Chemistry, 271, 17724–17732.
Feinstein, D. L., Galea, E., & Reis, D. J. (1997). Suppression of glial nitric oxide synthase
induction by heat shock: Effects on proteolytic degradation of IkappaB-alpha. Nitric
Oxide, 1, 167–176.
Garcia-Alcala, H., Santos Vichido, C. I., Islas Macedo, S., Genestier-Tamborero, C. N.,
Minutti-Palacios, M., Hirales Tamez, O., et al. (2015). Treatment with alpha-lipoic acid
over 16 weeks in Type 2 diabetic patients with symptomatic polyneuropathy who
responded to initial 4-week high-dose loading. Journal of Diabetes Research, 2015. 189857.
Gastpar, R., Gehrmann, M., Bausero, M. A., Asea, A., Gross, C., Schroeder, J. A., et al.
(2005). Heat shock protein 70 surface-positive tumor exosomes stimulate migratory
and cytolytic activity of natural killer cells. Cancer Research, 65, 5238–5247.
Gifondorwa, D. J., Robinson, M. B., Hayes, C. D., Taylor, A. R., Prevette, D. M.,
Oppenheim, R. W., et al. (2007). Exogenous delivery of heat shock protein 70 increases
lifespan in a mouse model of amyotrophic lateral sclerosis. Journal of Neuroscience, 27,
13173–13180.
Gil Lorenzo, A. F., Bocanegra, V., Benardon, M. E., Cacciamani, V., & Valles, P. G. (2014).
Hsp70 regulation on NOX4/p22Phox and cytoskeletal integrity as an effect of losartan in
vascular smooth muscle cells. Cell Stress & Chaperones, 19, 115–134.
Goeckeler, J. L., & Brodsky, J. L. (2010). Molecular chaperones and substrate ubiquitination
control the efficiency of endoplasmic reticulum-associated degradation. Diabetes Obe-
sity & Metabolism, 12(Suppl. 2), 32–38.
Gross, C., Hansch, D., Gastpar, R., & Multhoff, G. (2003). Interaction of heat shock protein
70 peptide with NK cells involves the NK receptor CD94. Biological Chemistry, 384,
267–279.
Guzhova, I., Kislyakova, K., Moskaliova, O., Fridlanskaya, I., Tytell, M., Cheetham, M.,
et al. (2001). In vitro studies show that Hsp70 can be released by glia and that exogenous
Hsp70 can enhance neuronal stress tolerance. Brain Research, 914, 66–73.
Haas, I. G., & Wabl, M. (1983). Immunoglobulin heavy chain binding protein. Nature, 306,
387–389.
Henstridge, D. C., Bruce, C. R., Drew, B. G., Tory, K., Kolonics, A., Estevez, E., et al.
(2014). Activating Hsp72 in rodent skeletal muscle increases mitochondrial number
and oxidative capacity and decreases insulin resistance. Diabetes, 63, 1881–1894.
Modulating Hsp70 to Treat DPN 205
Henstridge, D. C., Whitham, M., & Febbraio, M. A. (2014). Chaperoning to the metabolic
party: The emerging therapeutic role of heat-shock proteins in obesity and Type 2
diabetes. Molecular Metabolism, 3, 781–793.
Hightower, L. E., & Guidon, P. T., Jr. (1989). Selective release from cultured mammalian
cells of heat-shock (stress) proteins that resemble glia-axon transfer proteins. Journal of
Cellular Physiology, 138, 257–266.
Horn, P., Kalz, A., Lim, C. L., Pyne, D., Saunders, P., Mackinnon, L., et al. (2007). Exercise-
recruited NK cells display exercise-associated eHsp-70. Exercise Immunology Review, 13,
100–111.
Huang, C., Wang, J., Chen, Z., Wang, Y., & Zhang, W. (2013). 2-Phenylethynesulfonamide
prevents induction of pro-inflammatory factors and attenuates LPS-induced liver injury by
targeting NHE1-Hsp70 complex in mice. PloS One, 8, e67582.
Hunter-Lavin, C., Davies, E. L., Bacelar, M. M., Marshall, M. J., Andrew, S. M., &
Williams, J. H. (2004). Hsp70 release from peripheral blood mononuclear cells. Biochem-
ical and Biophysical Research Communications, 324, 511–517.
Hur, J., Sullivan, K. A., Pande, M., Hong, Y., Sima, A. A. F., Jagadish, H. V., et al. (2011).
The identification of gene expression profiles associated with progression of human
diabetic neuropathy. Brain, 134, 3222–3235.
Hutton, E. J., Carty, L., Laurá, M., Houlden, H., Lunn, M. P. T., Brandner, S., et al. (2011).
C-jun expression in human neuropathies: A pilot study. Journal of the Peripheral Nervous
System, 16, 295–303.
Ianaro, A., Lalenti, A., Maffia, P., Di Meglio, P., Di Rosa, M., & Santoro, M. G. (2003).
Anti-inflammatory activity of 15-deoxy-delta 12,14-PGJ2 and 2-cyclopenten-1-one:
Role of the heat shock response. Molecular Pharmacology, 64, 85–93.
Jaattela, M., & Wissing, D. (1993). Heat-shock proteins protect cells from monocyte cyto-
toxicity: Possible mechanism of self-protection. Journal of Experimental Medicine, 177,
231–236.
Johnson, J. D., & Fleshner, M. (2006). Releasing signals, secretory pathways, and immune
function of endogenous extracellular heat shock protein 72. Journal of Leukocyte Biology,
79, 425–434.
Jones, Q., Voegeli, T. S., Li, G., Chen, Y., & William Currie, R. (2011). Heat shock proteins
protect against ischemia and inflammation through multiple mechanisms. Inflammation &
Allergy Drug Targets, 10, 247–259.
Kalmar, B., Lu, C. H., & Greensmith, L. (2014). The role of heat shock proteins in
amyotrophic lateral sclerosis: The therapeutic potential of arimoclomol. Pharmacology
and Therapeutics, 141, 40–54.
Kamal, A., Thao, L., Sensintaffar, J., Zhang, L., Boehm, M. F., Fritz, L. C., et al. (2003).
A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors.
Nature, 425, 407–410.
Kim, Y. E., Hipp, M. S., Bracher, A., Hayer-Hartl, M., & Hartl, F. U. (2013). Molecular
chaperone functions in protein folding and proteostasis. Annual Review of Biochemistry,
82, 323–355.
Kondo, T., Murakami, K., Honkaniemi, J., Sharp, F. R., Epstein, C. J., & Chan, P. H.
(1996). Expression of Hsp70 mRNA is induced in the brain of transgenic mice over-
expressing human CuZn-superoxide dismutase following transient global cerebral ische-
mia. Brain Research, 737, 321–326.
Kovalchin, J. T., Wang, R., Wagh, M. S., Azoulay, J., Sanders, M., & Chandawarkar, R. Y.
(2006). In vivo delivery of heat shock protein 70 accelerates wound healing by
up-regulating macrophage-mediated phagocytosis. Wound Repair and Regeneration, 14,
129–137.
Krause, M., Heck, T. G., Bittencourt, A., Scomazzon, S. P., Newsholme, P., Curi, R., et al.
(2015). The chaperone balance hypothesis: The importance of the extracellular to
206 S.M. Emery and R.T. Dobrowsky
Ma, J., Farmer, K. L., Pan, P., Urban, M. J., Zhao, H., Blagg, B. S., et al. (2014). Heat shock
protein 70 is necessary to improve mitochondrial bioenergetics and reverse diabetic
sensory neuropathy following KU-32 therapy. Journal of Pharmacology and Experimental
Therapeutics, 348, 281–292.
Ma, J., Pan, P., Anyika, M., Blagg, B. S., & Dobrowsky, R. T. (2015). Modulating molecular
chaperones improves mitochondrial bioenergetics and decreases the inflammatory trans-
criptome in diabetic sensory neurons. ACS Chemical Neuroscience, 6, 1637–1648.
Madden, L. A., Sandstrom, M. E., Lovell, R. J., & McNaughton, L. (2008). Inducible
heat shock protein 70 and its role in preconditioning and exercise. Amino Acids, 34,
511–516.
Mansilla, M. J., Costa, C., Eixarch, H., Tepavcevic, V., Castillo, M., Martin, R., et al. (2014).
Hsp70 regulates immune response in experimental autoimmune encephalomyelitis. PloS
One, 9, e105737.
Meldrum, K. K. (2003). Liposomal delivery of heat shock protein 72 into renal tubular cells
blocks nuclear factor-kappaB activation, tumor necrosis factor-alpha production, and
subsequent ischemia-induced apoptosis. Circulation Research, 92, 293–299.
Miyata, Y., Nakamoto, H., & Neckers, L. (2013). The therapeutic target Hsp90 and cancer
hallmarks. Current Pharmaceutical Design, 19, 347–365.
Muchowski, P. J., & Wacker, J. L. (2005). Modulation of neurodegeneration by molecular
chaperones. Nature Reviews. Neuroscience, 6, 11–22.
Multhoff, G. (2007). Heat shock protein 70 (Hsp70): Membrane location, export and immu-
nological relevance. Methods, 43, 229–237.
Murakami, H., Pain, D., & Blobel, G. (1988). 70-kd Heat shock-related protein is one of at
least two distinct cytosolic factors stimulating protein import into mitochondria. Journal of
Cell Biology, 107, 2051–2057.
Neef, D. W., Jaeger, A. M., & Thiele, D. J. (2011). Heat shock transcription factor 1 as a
therapeutic target in neurodegenerative diseases. Nature Reviews. Drug Discovery, 10,
930–944.
Obrosova, I. G., Drel, V. R., Pacher, P., Ilnytska, O., Wang, Z. Q., Stevens, M. J., et al.
(2005). Oxidative-nitrosative stress and poly(ADP-ribose) polymerase (PARP) activa-
tion in experimental diabetic neuropathy: The relation is revisited. Diabetes, 54,
3435–3441.
Oglesbee, M. J., Herdman, A. V., Passmore, G. G., & Hoffman, W. H. (2005). Diabetic
ketoacidosis increases extracellular levels of the major inducible 70-kDa heat shock pro-
tein. Clinical Biochemistry, 38, 900–904.
Ortega, E., Giraldo, E., Hinchado, M. D., Martinez, M., Ibanez, S., Cidoncha, A., et al.
(2006). Role of Hsp72 and norepinephrine in the moderate exercise-induced stimulation
of neutrophils’ microbicide capacity. European Journal of Applied Physiology, 98, 250–255.
Ortega, E., Hinchado, M. D., Martin-Cordero, L., & Asea, A. (2009). The effect of stress-
inducible extracellular Hsp72 on human neutrophil chemotaxis: A role during acute
intense exercise. Stress, 12, 240–249.
Park, H. S., Lee, J. S., Huh, S. H., Seo, J. S., & Choi, E. J. (2001). Hsp72 functions as a natural
inhibitory protein of c-jun N-terminal kinase. EMBO Journal, 20, 446–456.
Parkinson, D. B., Bhaskaran, A., Arthur-Farraj, P., Noon, L. A., Woodhoo, A., Lloyd, A. C.,
et al. (2008). C-jun is a negative regulator of myelination. Journal of Cell Biology, 181,
625–637.
Peterson, L. B., & Blagg, B. S. (2009). To fold or not to fold: Modulation and consequences
of Hsp90 inhibition. Future Medicinal Chemistry, 1, 267–283.
Polla, B. S., Kantengwa, S., Francois, D., Salvioli, S., Franceschi, C., Marsac, C., et al. (1996).
Mitochondria are selective targets for the protective effects of heat shock against oxida-
tive injury. Proceedings of the National Academy of Sciences of the United States of America, 93,
6458–6463.
208 S.M. Emery and R.T. Dobrowsky
Qiao, Y., Liu, B., & Li, Z. (2008). Activation of NK cells by extracellular heat shock protein
70 through induction of NKG2D ligands on dendritic cells. Cancer Immunity, 8, 12.
Ran, R., Lu, A., Zhang, L., Tang, Y., Zhu, H., Xu, H., et al. (2004). Hsp70 promotes TNF-
mediated apoptosis by binding IKK gamma and impairing NF-kappaB survival signaling.
Genes and Development, 18, 1466–1481.
Richard, V., Kaeffer, N., & Thuillez, C. (1996). Delayed protection of the ischemic heart—
From pathophysiology to therapeutic applications. Fundamental and Clinical Pharmacology,
10, 409–415.
Robinson, M. B., Tidwell, J. L., Gould, T., Taylor, A. R., Newbern, J. M., Graves, J., et al.
(2005). Extracellular heat shock protein 70: A critical component for motoneuron sur-
vival. Journal of Neuroscience, 25, 9735–9745.
Rodrigues-Krause, J., Krause, M., O’Hagan, C., De Vito, G., Boreham, C., Murphy, C.,
et al. (2012). Divergence of intracellular and extracellular Hsp72 in Type 2 diabetes: Does
fat matter? Cell Stress & Chaperones, 17, 293–302.
Saibil, H. (2013). Chaperone machines for protein folding, unfolding and disaggregation.
Nature Reviews. Molecular Cell Biology, 14, 630–642.
Saleh, A., Roy Chowdhury, S. K., Smith, D. R., Balakrishnan, S., Tessler, L., Martens, C.,
et al. (2013). Ciliary neurotrophic factor activates NF-kappaB to enhance mitochondrial
bioenergetics and prevent neuropathy in sensory neurons of streptozotocin-induced dia-
betic rodents. Neuropharmacology, 65, 65–73.
Saleh, A., Schapansky, J., Smith, D. R., Young, N., Odero, G. L., Aulston, B., et al. (2013).
Normalization of NF-kappaB activity in dorsal root ganglia neurons cultured from dia-
betic rats reverses neuropathy-linked markers of cellular pathology. Experimental Neurol-
ogy, 241, 169–178.
Schmidt, O., Pfanner, N., & Meisinger, C. (2010). Mitochondrial protein import: From pro-
teomics to functional mechanisms. Nature Reviews Molecular Cell Biology, 11, 655–667.
Schulze, P. C., Yoshioka, J., Takahashi, T., He, Z., King, G. L., & Lee, R. T. (2004). Hyper-
glycemia promotes oxidative stress through inhibition of thioredoxin function by
thioredoxin-interacting protein. Journal of Biological Chemistry, 279, 30369–30374.
Senf, S. M., Howard, T. M., Ahn, B., Ferreira, L. F., & Judge, A. R. (2013). Loss of the
inducible Hsp70 delays the inflammatory response to skeletal muscle injury and severely
impairs muscle regeneration. PloS One, 8, e62687.
Singh, L. P. (2013). Thioredoxin interacting protein (TXNIP) and pathogenesis of diabetic
retinopathy. Journal of Clinical & Experimental Ophthalmology, 4. http://dx.doi.org/
10.4172/2155-9570.1000287.
Stavniichuk, R., Drel, V. R., Shevalye, H., Maksimchyk, Y., Kuchmerovska, T. M.,
Nadler, J. L., et al. (2011). Baicalein alleviates diabetic peripheral neuropathy through
inhibition of oxidative–nitrosative stress and p38 MAPK activation. Experimental Neurol-
ogy, 230, 106–113.
Stetler, R. A., Gan, Y., Zhang, W., Liou, A. K., Gao, Y., Cao, G., et al. (2010). Heat shock
proteins: Cellular and molecular mechanisms in the central nervous system. Progress in
Neurobiology, 92, 184–211.
Tan, W., Pasinelli, P., & Trotti, D. (2014). Role of mitochondria in mutant SOD1 linked
amyotrophic lateral sclerosis. Biochimica et Biophysica Acta, 1842, 1295–1301.
Theriault, J. R., Mambula, S. S., Sawamura, T., Stevenson, M. A., & Calderwood, S. K.
(2005). Extracellular Hsp70 binding to surface receptors present on antigen presenting
cells and endothelial/epithelial cells. FEBS Letters, 579, 1951–1960.
Tidwell, J. L., Houenou, L. J., & Tytell, M. (2004). Administration of Hsp70 in vivo inhibits
motor and sensory neuron degeneration. Cell Stress & Chaperones, 9, 88–98.
Tomlinson, D. R., & Gardiner, N. J. (2008). Glucose neurotoxicity. Nature Reviews.
Neuroscience, 9, 36–45.
Modulating Hsp70 to Treat DPN 209
Turturici, G., Sconzo, G., & Geraci, F. (2011). Hsp70 and its molecular role in nervous
system diseases. Biochemistry Research International, 2011. 618127.
Tytell, M., Greenberg, S. G., & Lasek, R. J. (1986). Heat shock-like protein is transferred
from glia to axon. Brain Research, 363, 161–164.
Urban, M. J., Li, C., Yu, C., Lu, Y., Krise, J. M., McIntosh, M. P., et al. (2010). Inhibiting
heat shock protein 90 reverses sensory hypoalgesia in diabetic mice. ASN Neuro, 2,
189–199.
Urban, M. J., Pan, P., Farmer, K. L., Zhao, H., Blagg, B. S., & Dobrowsky, R. T. (2012).
Modulating molecular chaperones improves sensory fiber recovery and mitochondrial
function in diabetic peripheral neuropathy. Experimental Neurology, 235, 388–396.
Vareniuk, I., Pavlov, I. A., Drel, V. R., Lyzogubov, V. V., Ilnytska, O., Bell, S. R., et al.
(2007). Nitrosative stress and peripheral diabetic neuropathy in leptin-deficient (ob/ob)
mice. Experimental Neurology, 205, 425–436.
Vihervaara, A., & Sistonen, L. (2014). HSF1 at a glance. Journal of Cell Science, 127, 261–266.
Walton-Diaz, A., Khan, S., Bourboulia, D., Trepel, J. B., Neckers, L., & Mollapour, M.
(2013). Contributions of co-chaperones and post-translational modifications towards
Hsp90 drug sensitivity. Future Medicinal Chemistry, 5, 1059–1071.
Wang, Y., Kelly, C. G., Karttunen, J. T., Whittall, T., Lehner, P. J., Duncan, L., et al. (2001).
CD40 is a cellular receptor mediating mycobacterial heat shock protein 70 stimulation of
CC-chemokines. Immunity, 15, 971–983.
Waza, M., Adachi, H., Katsuno, M., Minamiyama, M., Sang, C., Tanaka, F., et al. (2005).
17-AAG, an Hsp90 inhibitor, ameliorates polyglutamine-mediated motor neuron
degeneration. Nature Medicine, 11, 1088–1095.
Wei, W., Liu, Q., Tan, Y., Liu, L., Li, X., & Cai, L. (2009). Oxidative stress, diabetes, and
diabetic complications. Hemoglobin, 33, 370–377.
Weiss, Y. G., Bromberg, Z., Raj, N., Raphael, J., Goloubinoff, P., Ben-Neriah, Y., et al.
(2007). Enhanced heat shock protein 70 expression alters proteasomal degradation of
IkB kinase in experimental acute respiratory distress syndrome. Critical Care Medicine,
35, 2128–2138.
Wellen, K. E. (2005). Inflammation, stress, and diabetes. Journal of Clinical Investigation, 115,
1111–1119.
Whitham, M., & Fortes, M. B. (2008). Heat shock protein 72: Release and biological sig-
nificance during exercise. Frontiers in Bioscience, 13, 1328–1339.
Whiting, D. R., Guariguata, L., Weil, C., & Shaw, J. (2011). IDF Diabetes Atlas: Global
estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Research and Clinical
Practice, 94, 311–321.
Wiedemann, N., Frazier, A. E., & Pfanner, N. (2004). The protein import machinery of
mitochondria. Journal of Biological Chemistry, 279, 14473–14476.
Wong, H. R., Ryan, M. A., Menendez, I. Y., & Wispe, J. R. (1999). Heat shock activates the
I-kappaBalpha promoter and increases I-kappaBalpha mRNA expression. Cell Stress &
Chaperones, 4, 1–7.
Wong, H. R., Ryan, M., & Wispe, J. R. (1997). The heat shock response inhibits inducible
nitric oxide synthase gene expression by blocking I kappa-B degradation and NF-kappa
B nuclear translocation. Biochemical and Biophysical Research Communications, 231,
257–263.
Wright, E., Jr., Scism-Bacon, J. L., & Glass, L. C. (2006). Oxidative stress in type 2 diabetes:
The role of fasting and postprandial glycaemia. International Journal of Clinical Practice, 60,
308–314.
Wu, N., Zheng, B., Shaywitz, A., Dagon, Y., Tower, C., Bellinger, G., et al. (2013).
AMPK-dependent degradation of TXNIP upon energy stress leads to enhanced glucose
uptake via Glut1. Molecular Cell, 49, 1167–1175.
210 S.M. Emery and R.T. Dobrowsky
Yang, X. M., Baxter, G. F., Heads, R. J., Yellon, D. M., Downey, J. M., & Cohen, M. V.
(1996). Infarct limitation of the second window of protection in a conscious rabbit
model. Cardiovascular Research, 31, 777–783.
Young, J. C., Hoogenraad, N. J., & Hartl, F. U. (2003). Molecular chaperones Hsp90 and
Hsp70 deliver preproteins to the mitochondrial import receptor Tom70. Cell, 112,
41–50.
Zhang, L., Zhao, H., Blagg, B. S., & Dobrowsky, R. T. (2012). A C-terminal heat shock
protein 90 inhibitor decreases hyperglycemia-induced oxidative stress and improves
mitochondrial bioenergetics in sensory neurons. Journal of Proteome Research, 11,
129–137.
Ziegler, D., Ametov, A., Barinov, A., Dyck, P. J., Gurieva, I., Low, P. A., et al. (2006). Oral
treatment with α-lipoic acid improves symptomatic diabetic polyneuropathy. Diabetes
Care, 29, 2365–2370.
Ziegler, D., Low, P. A., Litchy, W. J., Boulton, A. J., Vinik, A. I., Freeman, R., et al. (2011).
Efficacy and safety of antioxidant treatment with alpha-lipoic acid over 4 years in diabetic
polyneuropathy: The Nathan 1 trial. Diabetes Care, 34, 2054–2060.
CHAPTER ELEVEN
Contents
1. Introduction 212
2. Nociceptive Ion Channels Control Neuronal Excitability 213
3. Alterations of Voltage-Gated and Ligand-Gated Ion Channels in Sensory Neurons
in Animal Models of Type 1 Diabetes 215
4. Alterations of CaV3.2 T-Type Channels in Nociceptive Sensory Neurons in Animal
Models of Type 2 Diabetes with Painful PDN 217
5. Posttranslational Modification of Pronociceptive Ion Channels in PDN 217
6. Diminished Inhibitory Drive in the Spinal Cord May Also Contribute to Painful PDN 221
7. Conclusions 221
Acknowledgments 222
References 223
Abstract
Pain-sensing sensory neurons (nociceptors) of the dorsal root ganglia (DRG) and dorsal
horn (DH) can become sensitized (hyperexcitable) in response to pathological condi-
tions such as diabetes, which in turn may lead to the development of painful peripheral
diabetic neuropathy (PDN). Because of incomplete knowledge about the mechanisms
underlying painful PDN, current treatment for painful PDN has been limited to some-
what nonspecific systemic drugs that have significant side effects or potential for abuse.
Recent studies have established that several ion channels in DRG and DH neurons are
dysregulated and make a previously unrecognized contribution to sensitization of pain
responses by enhancing excitability of nociceptors in animal models of type 1 and type
2 PDN. Furthermore, it has been reported that targeting posttranslational modification
of nociceptive ion channels such as glycosylation and methylglyoxal metabolism can
completely reverse mechanical and thermal hyperalgesia in diabetic animals with
PDN in vivo. Understanding details of posttranslational regulation of nociceptive chan-
nel activity may facilitate development of novel therapies for treatment of painful PDN.
We argue that pharmacological targeting of the specific pathogenic mechanism rather
than of the channel per se may cause fewer side effects and reduce the potential for
drug abuse in patients with diabetes.
#
International Review of Neurobiology, Volume 127 2016 Elsevier Inc. 211
ISSN 0074-7742 All rights reserved.
http://dx.doi.org/10.1016/bs.irn.2016.03.005
212 S.M. Todorovic
1. INTRODUCTION
Diabetes mellitus is a chronic, multifactorial disease characterized by
hyperglycemia. The World Health Organization estimates that 220 million
people worldwide currently suffer from diabetes. Diabetes mellitus type 1
(formerly called insulin-dependent diabetes or juvenile diabetes) is a form
of diabetes that results from autoimmune destruction of insulin-producing
β-cells in the pancreas. In contrast, type 2 diabetes (formerly called
noninsulin-dependent or adult-onset diabetes), the most common form
of diabetes, results from insulin resistance and is most often associated with
obesity. Peripheral diabetic neuropathy (PDN) is the most frequent compli-
cation of diabetes, and it has been estimated that 60–70% of diabetic patients
will develop PDN symptoms with prevalence increasing with the duration
of diabetes mellitus (Centers for Disease Control and Prevention, 2011).
The prevalence of painful PDN is often underestimated, but a recent large
community-based study with more than 15,000 patients with diabetes
reported painful symptoms in about one-third of all participants (Abbott,
Malik, van Ross, Kulkarni, & Boulton, 2011). These patients can suffer from
both stimulus-evoked painful episodes, including hyperalgesia (exaggerated
pain experience upon presentation of noxious stimuli) and/or allodynia
(normally nonnoxious stimuli that may evoke pain sensation), as well as
spontaneous pain in the form of burning or tingling. Interestingly, these
painful pathologies in diabetics usually occur in parallel with degeneration
of peripheral nerves (Abbott et al., 2011). Eventually, these painful symp-
toms usually subside as the disabling pain is replaced by complete loss of sen-
sation (insensate PDN). Both intractable pain and loss of sensation have
significant adverse effects on quality of life measures. Unfortunately, current
treatment options are unable to reverse these symptoms and are often asso-
ciated with serious side effects. For example, CaV2.2 (N-type) subtypes of
high-voltage-activated (HVA) calcium channels and their regulatory sub-
unit α2δ are considered a major cellular target for the anticonvulsants
gabapentin and pregabalin, which are commonly used to relieve diabetes-
induced pain in clinics. However, more than 50% of patients using
gabapentin or pregabalin experience side effects, such as excessive sedation,
ataxia, dizziness, euphoria, and weight gain, all of which limit their clinical
use (Edwards, Vincent, Cheng, & Feldman, 2008). Although opioids are
partially effective for treatment of painful PDN, they are not suitable for
chronic use since their long-term use is associated with serious side effects
including constipation, urinary retention, impaired cognitive function,
Painful Diabetic Neuropathy: Prevention or Suppression? 213
CaV3.2 T-channels typically decreases the threshold for action potential fir-
ing in DRG cells that express T-channels.
5. POSTTRANSLATIONAL MODIFICATION OF
PRONOCICEPTIVE ION CHANNELS IN PDN
Aberrant regulation of function of nociceptive ion channels in sensory
neurons via posttranslational modification and ensuing hyperexcitability
may be factors contributing to painful symptoms of PDN. One of the most
prevalent forms of posttranslational modification of proteins is glycosylation,
218 S.M. Todorovic
7. CONCLUSIONS
It is likely that multiple signaling pathways targeting nociceptive ion
channels may work in concert to promote hyperexcitability of sensory
222 S.M. Todorovic
ACKNOWLEDGMENTS
Our research is supported by American Diabetes Association National Award for
Basic Research 7-09-BS-190 (to S.M.T.) and research funds from the Department of
Anesthesiology at the University of Virginia, Charlottesville, VA, as well as Department
of Anesthesiology at the University of Colorado Anschutz Medical Campus, Aurora, CO.
Painful Diabetic Neuropathy: Prevention or Suppression? 223
REFERENCES
Abbott, C. A., Malik, R. A., van Ross, E. R. E., Kulkarni, J., & Boulton, A. J. M. (2011).
Prevalence and characteristics of painful diabetic neuropathy in a large community-based
diabetic population in the UK. Diabetes Care, 34, 2220–2224.
Andersson, D. A., Gentry, C., Light, E., Vastani, N., Vallortigara, J., Bierhaus, A., et al.
(2013). Methylglyoxal evokes pain by stimulating TRPA1. PloS One, 8(10), e77986.
Bierhaus, A., Fleming, T., Stoyanov, S., Leffler, A., Babes, A., Neacsu, C., et al. (2012).
Methylglyoxal modification of Nav1.8 facilitates nociceptive neuron firing and causes
hyperalgesia in diabetic neuropathy. Nature Medicine, 18(6), 926–933.
Brederson, J. D., Kym, P. R., & Szallasi, A. (2013). Targeting TRP channels for pain relief.
European Journal of Pharmacology, 716(1–3), 61–76.
Calcutt, N. A. (2013). Location, location, location? Is the pain of diabetic neuropathy gen-
erated by hyperactive sensory neurons? Diabetes, 62, 3658–3660.
Cao, X. H., Byun, H. S., Chen, S. R., & Pan, H. L. (2011). Diabetic neuropathy enhances
voltage-activated Ca2+ channel activity and its control by M4 muscarinic receptors in
primary sensory neurons. Journal of Neurochemistry, 119(3), 594–603.
Carbone, E., & Lux, H. D. (1984). A low-voltage activated, fully inactivating Ca2+ channel
in vertebrate sensory neurons. Nature, 310, 501–502.
Centers for Disease Control and Prevention. (2011). National diabetes fact sheet: national esti-
mates and general information on diabetes and prediabetes in the United States, 2011. Atlanta,
GA: US Department of Health and Human Services, Center for Disease Control and
Prevention. http://www.cdc.gov/diabetes/pubs/pdf/ndfs_2011.pdf.
Cohen, D. M. (2006). Regulation of TRP channels by N-linked glycosylation. Seminars in
Cell & Developmental Biology, 17(6), 630–637.
Dubreuil, A. S., Boukhaddaoui, H., Desmadryl, G., Martinez-Salgado, C., Moshourab, R.,
Lewin, G. R., et al. (2004). Role of T-type calcium current in identified d-hair mech-
anoreceptor neurons studied in vitro. The Journal of Neuroscience, 24, 8480–8484.
Eberhardt, M. J., Filipovic, M. R., Leffler, A., de la Roche, J., Kistner, K., Fischer, M. J.,
et al. (2012). Methylglyoxal activates nociceptors through transient receptor potential
channel A1 (TRPA1): A possible mechanism of metabolic neuropathies. The Journal
of Biological Chemistry, 287(34), 28291–28306.
Edwards, J. L., Vincent, A. M., Cheng, H. T., & Feldman, E. L. (2008). Diabetic neuropathy:
Mechanisms to management. Pharmacology & Therapeutics, 120, 1–34.
Gooch, C., & Podwall, D. (2004). The diabetic neuropathies. The Neurologist, 10, 311–322.
Hall, K. E., Liu, J., Sima, A. A., & Wiley, J. W. (2001). Impaired inhibitory G-protein func-
tion contributes to increased calcium currents in rats with diabetic neuropathy. Journal of
Neurophysiology, 86(2), 760–770.
Hall, K. E., Sima, A. A., & Wiley, J. W. (1995). Voltage-dependent calcium currents are
enhanced in dorsal root ganglion neurons from the Bio Bred/Worchester diabetic rat.
Journal of Physiology (London), 486(Pt. 2), 313–322.
Hall, K. E., Sima, A. A., & Wiley, J. W. (1996). Opiate-mediated inhibition of calcium
signaling is decreased in dorsal root ganglion neurons from the diabetic BB/W rat.
The Journal of Clinical Investigation, 97(5), 1165–1172.
Hansen, C. S., Jensen, T. M., Jensen, J. S., Nawroth, P., Fleming, T., Witte, D. R., et al.
(2015). The role of serum methylglyoxal on diabetic peripheral and cardiovascular auto-
nomic neuropathy: The ADDITION Denmark study. Diabetic Medicine, 32(6), 778–785.
Hoeijmakers, J. G., Faber, C. G., Merkies, I. S., & Waxman, S. G. (2014). Channelopathies,
painful neuropathy, and diabetes: Which way does the causal arrow point? Trends in
Molecular Medicine, 20(10), 544–550.
Hong, S., & Wiley, J. W. (2005). Early painful diabetic neuropathy is associated with differ-
ential changes in the expression and function of vanilloid receptor 1. The Journal of
Biological Chemistry, 280(1), 618–627.
224 S.M. Todorovic
Jack, M., & Wright, D. (2012). Role of advanced glycation end products and glyoxalase I in
diabetic peripheral sensory neuropathy. Translational Research, 159(5), 355–365.
Jacus, M. O., Uebele, V. N., Renger, J. J., & Todorovic, S. M. (2012). Presynaptic CaV3.2
channels regulate excitatory neurotransmission in nociceptive dorsal horn neurons. The
Journal of Neuroscience, 32(27), 9374–9382.
Jagodic, M. M., Pathirathna, S., Nelson, M. T., Mancuso, S., Joksovic, P. M.,
Rosenberg, E. R., et al. (2007). Cell-specific alterations of T-type calcium current in
painful diabetic neuropathy enhance excitability of sensory neurons. The Journal of
Neuroscience, 27(12), 3305–3316.
Jing, L., Chu, X. P., Jiang, Y. Q., Collier, D. M., Wang, B., Jiang, Q., et al. (2012).
N-glycosylation of acid-sensing ion channel 1a regulates its trafficking and acidosis-
induced spine remodeling. The Journal of Neuroscience, 32(12), 4080–4091.
Khomula, E. V., Viatchenko-Karpinski, V. Y., Borisyuk, A. L., Duzhyy, D. E., Belan, P. V.,
& Voitenko, N. V. (2013). Specific functioning of Cav3.2 T-type calcium and
TRPV1 channels under different types of STZ-diabetic neuropathy. Biochimica et
Biophysica Acta, 1832(5), 636–649.
Latham, J. R., Pathirathna, S., Jagodic, M. M., Choe, W. J., Levin, M. E., Nelson, M. T.,
et al. (2009). Selective T-type calcium channel blockade alleviates hyperalgesia in ob/ob
mice. Diabetes, 58(11), 2656–2665.
Lee-Kubli, C. A., & Calcutt, N. A. (2014). Altered rate-dependent depression of the spinal
H-reflex as an indicator of spinal disinhibition in models of neuropathic pain. Pain,
155(2), 250–260.
Messinger, R. B., Naik, A. K., Jagodic, M. M., Nelson, M. T., Lee, W. Y., Choe, W. J., et al.
(2009). In-vivo silencing of the Cav3.2 T-type calcium channels in sensory neurons
alleviates hyperalgesia in rats with streptozocin-induced diabetic neuropathy. Pain,
145(1–2), 184–195.
Moremen, K. W., Tiemeyer, M., & Naim, A. V. (2012). Vertebrate protein glycosylation:
Diversity, synthesis and function. Nature Reviews. Molecular Cell Biology, 13, 448–462.
Nelson, M. T., Joksovic, P. M., Perez-Reyes, E., & Todorovic, S. M. (2005). The endog-
enous redox agent L-cysteine induces T-type Ca2+ channel-dependent sensitization of a
novel subpopulation of rat peripheral nociceptors. The Journal of Neuroscience, 25,
8766–8775.
Nelson, M. T., Woo, J., Kang, H.-W., Barrett, P. Q., Vitko, J., Perez-Reyes, E., et al.
(2007). Reducing agents sensitize C-type nociceptors by relieving high-affinity zinc
inhibition of T-type calcium channels. The Journal of Neuroscience, 27(31), 8250–8260.
Obradovic, A. L. j., Hwang, S. M., Scarpa, J., Hong, S. J., Todorovic, S. M., & Jevtovic-
Todorovic, V. (2014). CaV3.2 T-type calcium channels in peripheral sensory neurons
are important for mibefradil-induced reversal of hyperalgesia and allodynia in rats with
painful diabetic neuropathy. PLoS One, 9(4), e9.
Obrosova, I. G. (2009). Diabetic painful and insensate neuropathy: Pathogenesis and poten-
tial treatments. Neurotherapeutics, 6(4), 638–647.
Orestes, P., Osuru, H. P., McIntire, W. E., Jacus, M. O., Salajegheh, R., Jagodic, M. M.,
et al. (2013). Reversal of neuropathic pain in diabetes by targeting glycosylation of
CaV3.2 T-type channels. Diabetes, 62(11), 3828–3838.
Pabbidi, R. M., Cao, D. S., Parihar, A., Pauza, M. E., & Premkumar, L. S. (2008). Direct role
of streptozotocin in inducing thermal hyperalgesia by enhanced expression of transient
receptor potential vanilloid 1 in sensory neurons. Molecular Pharmacology, 73(3), 995–1004.
Pabbidi, R. M., Yu, S. Q., Peng, S., Khardori, R., Pauza, M. E., & Premkumar, L. S. (2008).
Influence of TRPV1 on diabetes-induced alterations in thermal pain sensitivity. Molecular
Pain, 4, 9.
Pertusa, M., Madrid, R., Morenilla-Palao, C., Belmonte, C., & Viana, F. (2012).
N-glycosylation of TRPM8 ion channels modulates temperature sensitivity of cold
thermoreceptor neurons. The Journal of Biological Chemistry, 287(22), 18218–18229.
Painful Diabetic Neuropathy: Prevention or Suppression? 225
Shankarappa, S. A., Piedras-Renterı́a, E. S., & Stubbs, E. B., Jr. (2011). Forced-exercise
delays neuropathic pain in experimental diabetes: Effects on voltage-activated calcium
channels. Journal of Neurochemistry, 118(2), 224–236.
Shin, J. B., Martinez-Salgado, C., Heppenstall, P. A., & Lewin, G. R. (2003). A T-type
calcium channel required for normal function of a mammalian mechanoreceptor. Nature
Neuroscience, 6(7), 724–730.
Sivilotti, L., & Woolf, C. J. (1994). The contribution of GABAA and glycine receptors to
central sensitization: Disinhibition and touch-evoked allodynia in the spinal cord. Journal
of Neurophysiology, 72, 169–179.
Todorovic, S. M. (2015). Is diabetic pain caused by dysregulated ion channels in sensory
neurons? Diabetes, 64(12), 3987–3989.
Todorovic, S. M., & Jevtovic-Todorovic, V. (2011). T-type voltage-gated calcium channels
as targets for development of novel pain therapies. British Journal of Pharmacology, 163(3),
484–495.
Todorovic, S. M., & Jevtovic-Todorovic, V. (2014). Targeting of CaV3.2 T-type calcium
channels in peripheral sensory neurons for the treatment of painful diabetic neuropathy.
Pflügers Archiv, 466(4), 701–706.
Tyrrell, L., Renganathan, M., Dib-Hajj, S. D., & Waxman, S. G. (2001). Glycosylation alters
steady-state inactivation of sodium channel Nav1.9/NaN in dorsal root ganglion neu-
rons and is developmentally regulated. The Journal of Neuroscience, 21(24), 9629–9637.
Vlassara, H., Brownlee, M., & Cerami, A. (1981). Nonenzymatic glycosylation of peripheral
nerve protein in diabetes mellitus. Proceedings of the National Academy of Sciences of the
United States of America, 78(8), 5190–5192.
Weiss, N., Black, S. A. G., Bladen, C., Chen, L., & Zamponi, G. W. (2013). Surface expres-
sion and function of CaV3.2 T-type calcium channels are controlled by asparagines-
linked glycosylation. Pflugers Archiv—European Journal of Physiology, 465(8), 1159–1170.
White, G., Lovinger, D. M., & Weight, F. F. (1989). Transient low-threshold Ca2+ current
triggers burst firing through an afterdepolarizing potential in an adult mammalian neu-
ron. Proceedings of the National Academy of Sciences of the United States of America, 86,
6802–6806.
Woolf, C. J. (2011). Central sensitization: Implications for the diagnosis and treatment of
pain. Pain, 152(3 Suppl.), S2–S15.
Xu, G. Y., Li, G., Liu, N., & Huang, L. Y. (2011). Mechanisms underlying purinergic P2X3
receptor-mediated mechanical allodynia induced in diabetic rats. Molecular Pain, 7, 60.
CHAPTER TWELVE
Contents
1. NCV as an Endpoint in Animal Models 229
2. Treatment Paradigms 230
3. Insanity: Doing the Same Thing Over and Over Again and Expecting
a Different Result 231
References 232
#
International Review of Neurobiology, Volume 127 2016 Elsevier Inc. 229
ISSN 0074-7742 All rights reserved.
http://dx.doi.org/10.1016/bs.irn.2016.03.009
230 P. Fernyhough and N.A. Calcutt
2. TREATMENT PARADIGMS
The majority of preclinical studies have used a prevention paradigm.
Drugs are applied at the beginning of the study and forestall development
New Directions in Diabetic Neuropathy 231
of the preclinical drug screen as a precursor to clinical trials. Perhaps the most
pertinent development in recent years is that studies in rodent models of
diabetes are now using an array of small-fiber endpoints that replicate the
clinical condition to test ideas on etiology and screen new therapies
(Beiswenger, Calcutt, & Mizisin, 2008; Christianson, Riekhof, & Wright,
2003; Christianson, Ryals, Johnson, Dobrowsky, & Wright, 2007;
Davidson, Coppey, Holmes, & Yorek, 2012a, 2012b). Loss of epidermal
fibers, loss of sweat gland innervation, loss of thermal (hot and cold) sensa-
tion, and loss of corneal nerve fiber density all occur in both diabetic animals
and humans (Arezzo, Schaumburg, & Laudadio, 1986; Breiner, Lovblom,
Perkins, & Bril, 2014; Hossain, Sachdev, & Malik, 2005; Kennedy,
Wendelschafer-Crabb, & Johnson, 1996; Liu et al., 2015; Malik et al.,
2003; Zinman, Bril, & Perkins, 2004). Other advantages of working with
small-fiber endpoints arise from reports that onset of structural change
can be detected early in the course of disease in both diabetic animals
and humans (Azmi et al., 2015; Christianson et al., 2003, 2007; Mehra et
al., 2007; Petropoulos et al., 2015; Quattrini et al., 2007; Smith,
Ramachandran, Tripp, & Singleton, 2001). Corneal nerve fiber loss, which
can be detected by confocal microscopy, can be viewed noninvasively so as
to allow frequent iterative assessment before and after therapeutic interven-
tions in diabetic rodents and humans (Chen et al., 2013; Tavakoli et al.,
2013). It should also be remembered that the aspects of diabetic neuropathy
that most trouble patients reflect sensory and autonomic dysfunction. Once
fully validated, the introduction of measures of small-fiber nerve pathology
and dysfunction as primary goals and endpoints in clinical trials of treatments
for diabetic neuropathy may allow shorter, less costly, trials that have better
chances of gaining regulatory approval and improving the lives of patients.
REFERENCES
Arezzo, J. C., Schaumburg, H. H., & Laudadio, C. (1986). Thermal sensitivity tester. Device
for quantitative assessment of thermal sense in diabetic neuropathy. Diabetes, 35,
590–592.
Azmi, S., Ferdousi, M., Petropoulos, I. N., Ponirakis, G., Alam, U., Fadavi, H., et al. (2015).
Corneal confocal microscopy identifies small-fiber neuropathy in subjects with impaired
glucose tolerance who develop type 2 diabetes. Diabetes Care, 38, 1502–1508.
Beiswenger, K. K., Calcutt, N. A., & Mizisin, A. P. (2008). Dissociation of thermal
hypoalgesia and epidermal denervation in streptozotocin-diabetic mice. Neuroscience
Letters, 442, 267–272.
Breiner, A., Lovblom, L. E., Perkins, B. A., & Bril, V. (2014). Does the prevailing hypothesis
that small-fiber dysfunction precedes large-fiber dysfunction apply to type 1 diabetic
patients? Diabetes Care, 37, 1418–1424.
New Directions in Diabetic Neuropathy 233
Bril, V., Hirose, T., Tomioka, S., & Buchanan, R. (2009). Ranirestat for the management of
diabetic sensorimotor polyneuropathy. Diabetes Care, 32, 1256–1260.
Calcutt, N. A., Allendoerfer, K. L., Mizisin, A. P., Middlemas, A., Freshwater, J. D.,
Burgers, M., et al. (2003). Therapeutic efficacy of sonic hedgehog protein in experimen-
tal diabetic neuropathy. The Journal of Clinical Investigation, 111, 507–514.
Calcutt, N. A., Campana, W. M., Eskeland, N. L., Mohiuddin, L., Dines, K. C.,
Mizisin, A. P., et al. (1999). Prosaposin gene expression and the efficacy of a prosaposin-
derived peptide in preventing structural and functional disorders of peripheral nerve in
diabetic rats. Journal of Neuropathology and Experimental Neurology, 58, 628–636.
Chen, D. K., Frizzi, K. E., Guernsey, L. S., Ladt, K., Mizisin, A. P., & Calcutt, N. A. (2013).
Repeated monitoring of corneal nerves by confocal microscopy as an index of peripheral
neuropathy in type-1 diabetic rodents and the effects of topical insulin. Journal of the
Peripheral Nervous System, 18, 306–315.
Christianson, J. A., Riekhof, J. T., & Wright, D. E. (2003). Restorative effects of
neurotrophin treatment on diabetes-induced cutaneous axon loss in mice. Experimental
Neurology, 179, 188–199.
Christianson, J. A., Ryals, J. M., Johnson, M. S., Dobrowsky, R. T., & Wright, D. E. (2007).
Neurotrophic modulation of myelinated cutaneous innervation and mechanical sensory
loss in diabetic mice. Neuroscience, 145, 303–313.
Davidson, E. P., Coppey, L. J., Holmes, A., & Yorek, M. A. (2012a). Changes in corneal inner-
vation and sensitivity and acetylcholine-mediated vascular relaxation of the posterior ciliary
artery in a type 2 diabetic rat. Investigative Ophthalmology & Visual Science, 53, 1182–1187.
Davidson, E. P., Coppey, L. J., & Yorek, M. A. (2012b). Early loss of innervation of cornea
epithelium in streptozotocin-induced type 1 diabetic rats: Improvement with ilepatril
treatment. Investigative Ophthalmology & Visual Science, 53, 8067–8074.
DCCT. (1993). The effect of intensive treatment of diabetes on the development and
progression of long-term complications in insulin-dependent diabetes mellitus. The
Diabetes Control and Complications Trial Research Group. The New England Journal
of Medicine, 329, 977–986.
DCCT. (2002). Effect of intensive therapy on the microvascular complications of type 1
diabetes mellitus. JAMA, 287, 2563–2569.
Eliasson, S. G. (1964). Nerve conduction changes in experimental diabetes. The Journal of
Clinical Investigation, 43, 2353–2358.
Greene, D. A., De Jesus, P. V., Jr., & Winegrad, A. I. (1975). Effects of insulin and dietary
myoinositol on impaired peripheral motor nerve conduction velocity in acute
streptozotocin diabetes. The Journal of Clinical Investigation, 55, 1326–1336.
Hossain, P., Sachdev, A., & Malik, R. A. (2005). Early detection of diabetic peripheral neu-
ropathy with corneal confocal microscopy. Lancet, 366, 1340–1343.
Hounsom, L., & Tomlinson, D. R. (1997). Does neuropathy develop in animal models?
Clinical Neuroscience, 4, 380–389.
Jakobsen, J. (1976a). Axonal dwindling in early experimental diabetes. I. A study of cross sec-
tioned nerves. Diabetologia, 12, 539–546.
Jakobsen, J. (1976b). Axonal dwindling in early experimental diabetes. II. A study of isolated
nerve fibres. Diabetologia, 12, 547–553.
Kalichman, M. W., Dines, K. C., Bobik, M., & Mizisin, A. P. (1998). Nerve conduction
velocity, laser Doppler flow, and axonal caliber in galactose and streptozotocin diabetes.
Brain Research, 810, 130–137.
Kato, N., Mizuno, K., Makino, M., Suzuki, T., & Yagihashi, S. (2000). Effects of 15-month
aldose reductase inhibition with fidarestat on the experimental diabetic neuropathy in
rats. Diabetes Research and Clinical Practice, 50, 77–85.
Kennedy, W. R., Wendelschafer-Crabb, G., & Johnson, T. (1996). Quantitation of epider-
mal nerves in diabetic neuropathy. Neurology, 47, 1042–1048.
234 P. Fernyhough and N.A. Calcutt
Liu, Y., Billiet, J., Ebenezer, G. J., Pan, B., Hauer, P., Wei, J., et al. (2015). Factors influenc-
ing sweat gland innervation in diabetes. Neurology, 84, 1652–1659.
Malik, R. A., Kallinikos, P., Abbott, C. A., van Schie, C. H., Morgan, P., Efron, N., et al.
(2003). Corneal confocal microscopy: A non-invasive surrogate of nerve fibre damage
and repair in diabetic patients. Diabetologia, 46, 683–688.
Mehra, S., Tavakoli, M., Kallinikos, P. A., Efron, N., Boulton, A. J., Augustine, T., et al.
(2007). Corneal confocal microscopy detects early nerve regeneration after pancreas
transplantation in patients with type 1 diabetes. Diabetes Care, 30, 2608–2612.
Mizisin, A. P., Nelson, R. W., Sturges, B. K., Vernau, K. M., Lecouteur, R. A.,
Williams, D. C., et al. (2007). Comparable myelinated nerve pathology in feline and
human diabetes mellitus. Acta Neuropathologica, 113, 431–442.
Mizisin, A. P., Shelton, G. D., Burgers, M. L., Powell, H. C., & Cuddon, P. A. (2002).
Neurological complications associated with spontaneously occurring feline diabetes
mellitus. Journal of Neuropathology and Experimental Neurology, 61, 872–884.
Petropoulos, I. N., Green, P., Chan, A. W., Alam, U., Fadavi, H., Marshall, A., et al. (2015).
Corneal confocal microscopy detects neuropathy in patients with type 1 diabetes without
retinopathy or microalbuminuria. PLoS One, 10, e0123517.
Quattrini, C., Tavakoli, M., Jeziorska, M., Kallinikos, P., Tesfaye, S., Finnigan, J., et al.
(2007). Surrogate markers of small fiber damage in human diabetic neuropathy.
Diabetes, 56, 2148–2154.
Sima, A. A. (2004). Diabetic neuropathy in type 1 and type 2 diabetes and the effects of
C-peptide. Journal of the Neurological Sciences, 220, 133–136.
Smith, A. G., Ramachandran, P., Tripp, S., & Singleton, J. R. (2001). Epidermal nerve
innervation in impaired glucose tolerance and diabetes-associated neuropathy.
Neurology, 57, 1701–1704.
Tavakoli, M., Mitu-Pretorian, M., Petropoulos, I. N., Fadavi, H., Asghar, O., Alam, U.,
et al. (2013). Corneal confocal microscopy detects early nerve regeneration in diabetic
neuropathy after simultaneous pancreas and kidney transplantation. Diabetes, 62,
254–260.
Wahren, J., Foyt, H., Daniels, M., & Arezzo, J. C. (2016). Long-acting C-peptide and
neuropathy in type 1 diabetes: A 12-month clinical trial. Diabetes Care, 39, 596–602.
Yagihashi, S., Yamagishi, S., & Wada, R. (2007). Pathology and pathogenetic mechanisms of
diabetic neuropathy: Correlation with clinical signs and symptoms. Diabetes Research and
Clinical Practice, 77(Suppl. 1), S184–S189.
Zinman, L. H., Bril, V., & Perkins, B. A. (2004). Cooling detection thresholds in the
assessment of diabetic sensory polyneuropathy: Comparison of CASE IV and Medoc
instruments. Diabetes Care, 27, 1674–1679.
Zochodne, D. W., Verge, V. M., Cheng, C., Sun, H., & Johnston, J. (2001). Does diabetes
target ganglion neurones? Progressive sensory neurone involvement in long-term
experimental diabetes. Brain, 124, 2319–2334.
CHAPTER THIRTEEN
Contents
1. Introduction 236
2. Diabetic Peripheral Neuropathy 238
2.1 Clinical Presentation 238
2.2 Diagnosis 240
2.3 Falls Risk in Older Adults with Diabetes 241
3. Diabetic Autonomic Neuropathy 245
3.1 Clinical Manifestations 245
3.2 Cardiovascular Autonomic Neuropathy 247
3.3 Prevention and Reversibility of Autonomic Neuropathy 251
4. Measuring Diabetic Neuropathy: Established and Innovative Approaches 253
4.1 QOL Measures 253
4.2 Clinical Assessment Tools 256
4.3 Objective Measurements 256
5. Concluding Remarks 270
References 271
Abstract
Here we review some seldom-discussed presentations of diabetic neuropathy, includ-
ing large fiber dysfunction and peripheral autonomic dysfunction, emphasizing the
impact of sympathetic/parasympathetic imbalance. Diabetic neuropathy is the most
common complication of diabetes and contributes additional risks in the aging adult.
Loss of sensory perception, loss of muscle strength, and ataxia or incoordination lead to
a risk of falling that is 17-fold greater in the older diabetic compared to their young
nondiabetic counterparts. A fall is accompanied by lacerations, tears, fractures, and
worst of all, traumatic brain injury, from which more than 60% do not recover. Auto-
nomic neuropathy has been hailed as the “Prophet of Doom” for good reason. It is con-
ducive to increased risk of myocardial infarction and sudden death. An imbalance in the
autonomic nervous system occurs early in the evolution of diabetes, at a stage when
active intervention can abrogate the otherwise relentless progression. In addition to
hypotension, many newly recognized syndromes can be attributed to cardiac auto-
nomic neuropathy such as orthostatic tachycardia and bradycardia. Ultimately, this con-
stellation of features of neuropathy conspire to impede activities of daily living,
especially in the patient with pain, anxiety, depression, and sleep disorders. The resulting
reduction in quality of life may worsen prognosis and should be routinely evaluated and
addressed. Early neuropathy detection can only be achieved by assessment of both
large and small- nerve fibers. New noninvasive sudomotor function technologies
may play an increasing role in identifying early peripheral and autonomic neuropathy,
allowing rapid intervention and potentially reversal of small-fiber loss.
1. INTRODUCTION
Neuropathy is the most common and troublesome complication of
diabetes mellitus, leading to the greatest morbidity and mortality and
resulting in a huge economic burden for diabetes care (Holzer et al.,
1998; Vinik, Mitchell, et al., 1995) (Fig. 1). It is the most common form
of neuropathy in the developed countries of the world, accounts for more
hospitalizations than all the other diabetic complications combined, and is
DPN
Painful Impairment
Neuropathic
neuropathic disability
deficits
symptoms handicap
Mortality
Foot ulcers
Ataxia charcot
weakness
Cost
$37B
Fractures Surgery
Cost lacerations amputation
$200B traumatic brain 96,000/y
injury
Fig. 1 The clinical and financial implications of aging and diabetic peripheral neurop-
athy (DPN).
Alternative Neuropathy Assessments 237
2.2 Diagnosis
The diagnosis of DSPN should rest on the findings of the clinical and neu-
rological examinations, with consideration of the presence of neuropathic
symptoms (positive and negative, sensory, and motor) and signs (sensory def-
icit, allodynia and hyperalgesia, motor weakness, and absence of reflexes).
According to the American Academy of Neurology report on the case def-
inition of distal symmetric polyneuropathy, there is good evidence that:
(A) Symptoms alone have poor diagnostic accuracy in predicting the pres-
ence of polyneuropathy,
(B) Signs are better predictors than symptoms,
(C) Multiple signs are better predictors than a single sign, and
(D) Relatively simple examinations are as accurate as complex scoring sys-
tems (England et al., 2005).
The most recent position statement of the American Diabetes Association
recommends that all patients with diabetes be screened for neuropathy at
diagnosis in type 2 diabetes and 5 years after diagnosis in type 1 diabetes.
Neuropathy screening should be repeated annually and must include sensory
examination of the feet and ankle reflexes (American Diabetes Association,
2015). The basic neurological assessment comprises the general medical and
neurological history to exclude other potential causes of neuropathy, inspec-
tion of the feet, and neurological examination of sensation. As for all patients
with distal symmetric polyneuropathy (England et al., 2009), screening lab-
oratory tests may be considered in selected patients with DSPN, with serum
B12 with its metabolites and serum protein immunofixation electrophoresis
being those with the highest yield of abnormalities (England et al., 2009).
The neurological examination should focus on the lower extremities and
always include an accurate foot inspection for deformities, ulcers, fungal
infection, muscle wasting, hair distribution or loss, and the presence or
absence of pulses. Footwear should also be inspected at every visit. Sensory
modalities should be assessed using simple handheld devices (Table 1)
(Haanpaa et al., 2009). The most sensitive measure is the vibration detection
threshold, although sensitivity of 10-g Semmes-Weinstein monofilament
to identify feet at risk varies between 86% and 100% (Armstrong, Lavery,
Vela, Quebedeaux, & Fleischli, 1998; Kumar et al., 1991). Combinations
of more than one test have more than 87% sensitivity in detecting DSPN
(Boulton et al., 2005; Vinik, Suwanwalaikorn, et al., 1995). Longitudinal
studies show that these simple tests are good predictors of foot ulcer risk
(Abbott et al., 2002). Knee and ankle reflexes should also be tested
Alternative Neuropathy Assessments 241
(Boulton, Gries, & Jervell, 1998; Boulton et al., 2005) and assessment of
joint position and motor power may be indicated.
The following findings should alert the physician to consider causes for
DSPN other than diabetes and cause referral for a detailed neurological
work-up: (1) pronounced asymmetry of the neurological deficits; (2) predom-
inantly motor deficits, mononeuropathy, or cranial nerve involvement;
(3) rapid development or progression of the neuropathic impairments; (4) pro-
gression of the neuropathy despite optimal glycemic control; (5) symptoms
from the upper limbs; and (6) family history of nondiabetic neuropathy.
of these adverse events within a given year. Given the range of health issues
that can arise following a fall, prevention should be the first course of action.
The key to preventing falls is identification of persons at risk and
implementing appropriate interventions. This requires recognizing factors
that can lead to increased risk of falling. However, despite our understanding
of the seriousness of falls and the high cost of medical care, identifying a few
critical factors that are strongly predictive of falls in high-risk populations is
challenging, partly because over 400 factors are linked with falls in adult
populations (Close, Lord, Menz, & Sherrington, 2005). Something as simple
as an individual’s perception of threat around them when they move (often
referred to as a “fear of falling”) can be a significant health issue. Nearly
13 million (36%) of American adults aged 65 years or more have been found
to be moderately or very afraid of falling, illustrating that developing a fear of
suffering an adverse event is strongly linked with actual falls (Boyd &
Stevens, 2009).
2.3.2 Risk of Falling Is Increased for Older Adults with Type 2 Diabetes
Persons with type 2 diabetes are at increased risk of falling compared to
healthy adults of a similar age. A combination of age (>65 years) and diabetes
further increases the risk of falling 17-fold (Cavanagh, Derr, Ulbrecht,
Maser, & Orchard, 1992; Pijpers et al., 2012) due to factors such as sensory
neuropathy, declining cognitive function, and use of multiple prescription
medications (Close et al., 2005; Peron & Ogbonna, 2015; Schwartz et al.,
2002, 2008). The likelihood of suffering a fall increases dramatically with
increasing age and/or the emergence of type 2 diabetes, with risk being
increased significantly by the presence of diabetes alone (Clark, Lord, &
Webster, 1993; Close et al., 2005; Lord, 1996; Lord & Clark, 1996;
Pickering et al., 2007; Robinovitch et al., 2000; Sosnoff, Motl, &
Morrison, 2013; Sosnoff et al., 2011). Older persons with diabetes must con-
tend with both age-related declines in balance control, muscle strength,
walking ability, and proprioception (Morrison, Colberg, Mariano,
Parson, & Vinik, 2010; Morrison, Colberg, Parson, & Vinik, 2012;
Schwartz et al., 2002, 2008; Wallace et al., 2002) and health-related issues
associated with diabetes (Berlie & Garwood, 2010; Maurer, Burcham, &
Cheng, 2005; Richardson & Hurvitz, 1995; Tilling, Darawil, & Britton,
2006). Indeed, the additional range of potential risk factors in anyone
with diabetes is extensive, covering neuropathy, visual deficits, loss of coor-
dination, cognitive impairment, autonomic dysfunction with orthostatic
hypotension, tachycardia, bradycardia, pain, poor lower body function,
Alternative Neuropathy Assessments 243
Fig. 2 A clinical algorithm for the evaluation of and targeted intervention for the aging
patient at risk of falling.
Alternative Neuropathy Assessments 245
Banks, & Blair, 2007; Peterson et al., 2009; Robinovitch et al., 2000;
Sherrington, Lord, & Herbert, 2003) and studies have reported that
targeted interventions can improve balance and walking ability and also
reduce falls risk in diabetes (Allet et al., 2009; Morrison et al., 2010,
2012). Structured balance training can lead to improvements in posture
and/or gait function (Allet et al., 2010; Morrison et al., 2010, 2012) as well
as general gains from physical activity like faster reaction times, improve-
ments in sensory perception and lower limb strength, and better sympa-
thetic/parasympathetic balance (Colberg, 2006; Colberg, Stansberry,
McNitt, & Vinik, 2002; Herriott, Colberg, Parson, Nunnold, & Vinik,
2004; Morrison et al., 2010, 2012). Interestingly, recent studies have
reported that exercise can also lead to improvements in neuropathy symp-
toms, including increased nerve fiber branching (Kluding et al., 2012) and
improved sensory responses in the lower limbs (Balducci et al., 2006).
and increases heart rate, stroke volume, and peripheral vascular resistance,
thus contributing to LV dysfunction. Such sympathetic hyperactivity, in
combination with regional myocardial sympathetic denervation, has
been shown to lead to diminished coronary blood flow reserve and diastolic
dysfunction in diabetic patients with early microangiopathy (Dinh et al.,
2011; Sacre et al., 2010; Vinik & Erbas, 2013).
odds ratio for autonomic neuropathy (Gaede, Vedel, Parving, & Pedersen,
1999). The EDIC study, a longitudinal cohort follow-up study for the
DCCT in which patients with type 1 diabetes were randomized to conven-
tional or intensive glycemic control, demonstrated persistent beneficial
effects of past glucose control on microvascular complications despite the
loss of glycemic separation in type 1 diabetic patients (DCCT EDIC
Research Group, 2003). During the EDIC follow-up, CAN progressed
in both treatment groups, but the incidence and prevalence of CAN
remained lower in the formerly intensive group than in the formerly con-
ventional group, despite similar levels of glycemic control during EDIC. To
minimize the development of autonomic neuropathy, intensive glucose
control of type 1 diabetes should therefore be started as early as possible
(Pop-Busui et al., 2009). However, while glycemic control with a reduction
of HbA1c from 9.5 to 8.4 improved HRV in those patients with mild auto-
nomic abnormalities, this was not so in type 1 diabetics with advanced auto-
nomic abnormalities (Burger, Weinrauch, D’Elia, & Aronson, 1999).
The effects of glycemic control in type 2 diabetics are less definite. The VA
Cooperative Study showed no difference in the prevalence of autonomic neu-
ropathy after 2 years of intensive glycemic control in type 2 diabetic patients
(Azad et al., 1999). In the Steno-2 Study, where people with type 2 diabetes
received intensive multifactorial treatment that targeted hyperglycemia,
hypertension, dyslipidemia, and microalbuminuria, along with secondary pre-
vention of CVD with aspirin, the approach reduced autonomic dysfunction
by 63%. The glucose-lowering agents appeared to have the least effect when
compared with antihypertensive treatment, lipid-lowering agents, aspirin, and
vitamin–mineral supplements (Gaede et al., 1999). In addition, a survey of
evidence from clinical trials shows that early identification of autonomic neu-
ropathy permits timely initiation of therapy with the antioxidant alpha-lipoic
acid, which slows or reverses progression of CAN (Ziegler et al., 1997).
Early ACE inhibition or angiotensin receptor blockade improved both
DAN and LVDD after 1 year of treatment in asymptomatic patients with long-
term diabetes, with the combination being slightly better than monotherapies
and auguring well for the patient with established CAN (Didangelos et al.,
2006). Fluvastatin treatment also improves cardiac sympathetic neuropathy
in the diabetic rat heart, in association with attenuation of increased cardiac
oxidative stress (Matsuki et al., 2010). On the other hand, selective
COX-2 inactivation confers protection against sympathetic denervation dur-
ing experimental diabetes by reducing intramyocardial oxidative stress and
inflammation (Kellogg, Converso, Wiggin, Stevens, & Pop-Busui, 2009).
Alternative Neuropathy Assessments 253
patients are more likely to have slower NCVs than patients without
symptoms, these do not relate with the severity of symptoms. In a long-term
follow-up study of type 2 diabetes patients (Partanen et al., 1995), electro-
physiologic abnormalities in the lower limb increased from 8% at baseline to
42% after 10 years with a decrease in sensory and motor amplitudes
(indicating axonal destruction) being more pronounced than NCV slowing.
Using objective measures of sensory function such as the vibration percep-
tion threshold test, the rate of decline in function has been reported as 1–2
vibration units/year. However, the most recent clinical studies suggest
that there is a decline in this rate of progression. The advent of therapeutic
lifestyle change and the use of statins and ACE inhibitors likely contribute
to slowed progression of neuropathy and have drastically changed the
requirements for placebo-controlled studies (Casellini et al., 2007). It is also
important to recognize that diabetic neuropathy is a disorder wherein the
prevailing abnormality is loss of nerve fibers and that this is reflected by a
reduction in electromyogram amplitudes, not conduction velocity. There-
fore, changes in NCV may not be an appropriate means of monitoring
progress or deterioration of nerve function. Furthermore, small, unmyelin-
ated nerve fibers are affected early in diabetes and are not reflected in NCV
studies. Other methods of measuring neuropathy that do not depend on
NCV, such as QST (see earlier), or skin biopsy with quantification of
intraepidermal nerve fibers (IENF), are necessary to identify these patients
(Pittenger et al., 2004; Pittenger, Simmons, et al., 2005; Sinnreich
et al., 2005).
care (Bromm & Treede, 1991). CHEPS, with its rapidly heating thermode
(70°C/s), allows for repeated assessments without the risk of long-term
damage to the area of interest. Studies supporting reproducibility of CHEPS
after 6 months (Ruscheweyh, Emptmeyer, Putzer, Kropp, & Marziniak,
2013), especially in areas of interest (Kramer et al., 2012), lend further sup-
port for its use in repeated clinical assessments.
Various groups, including ours, have performed comparison studies using
CHEPS in neuropathic populations, and CHEPS intrapeak amplitudes (IA)
have proven to be a distinguishable marker differentiating neuropaths from
nonneuropaths (Atherton et al., 2007; Chao et al., 2010; Parson et al., 2013;
Wong & Chung, 2011). CHEPS IA also correlates with other assessments of
small nerve fibers, including skin-flare response and IENF density (Atherton
et al., 2007; Chao, Hsieh, Tseng, Chang, & Hsieh, 2008; Chao et al., 2010).
Assessment of structural changes may miss early nerve dysfunction in the
presence of normal IENF density. However, CHEPS provides a sensitive
(76.4%) and specific (80.6%) tool in assessing nerve fiber loss in the dorsal foot
with IENF as a standard for comparison, while providing a functional eval-
uation (Casanova-Molla, Grau-Junyent, Morales, & Valls-Sole, 2011).
In a comparison of diabetic and nondiabetic patients, CHEPS IAs of the
lumbosacral region, dorsal, and volar forearm negatively correlated with
HbA1c (Parson et al., 2013). Diabetic neuropaths also tend to have CHEPS
responses of longer latencies and lower amplitudes (Parson et al., 2013)
(Chao et al., 2010). Upon closer examination of patients with DSPN,
CHEPS responses tend to be higher in those with pain than those without
(Parson et al., 2013), which has led to further investigation of the tool for
studying pain models and assessing intervention.
The versatility of CHEPS to study function in conjunction with other
devices, such as fMRI and magnetoencephalography, further adds to its
value in providing a more global evaluation of future therapeutics and
broadening our understanding of the mechanism of pain (Gopalakrishnan,
Machado, Burgess, & Mosher, 2013; Kramer, Jutzeler, Haefeli, Curt, &
Freund, 2015; Shenoy et al., 2011). Thus, there is a great need to create
a standard for CHEPS testing and normative values. Endeavors by Dutch
(Lagerburg et al., 2015) and Taiwanese groups (Chen et al., 2006) to create
such values should further accelerate its validation in diabetic neuropathy
and its various subtypes. However, future normative values must be
obtained from a more universally diverse cohort to truly create a robust stan-
dard with meaningful clinical significance.
260 A.I. Vinik et al.
160
Lean controls, n = 19
140
Obese controls, n = 13
120 Relatives, n = 19
Skin perfusion units
IGT, n = 14
100 Type 2 diabetes, n = 22
80
60
40
20
0
Occlusion 32° C 44° C
Minutes
Fig. 3 Abnormalities in heat-mediated vasodilation of hairy skin in obese controls, rel-
atives of subjects with diabetes, impaired glucose tolerance (IGT), and type 2 diabetes
subjects, as compared with lean controls (Vinik et al., 2013).
tests can be used as a surrogate for the diagnosis of DAN in any system since it
is generally rare to find involvement of any other division of the ANS in the
absence of cardiovascular autonomic dysfunction. For example, if one enter-
tains the possibility that the patient has erectile dysfunction due to DAN,
then prior to embarking upon a sophisticated and expensive evaluation of
erectile status, a measure of heart rate and its variability in response to deep
breathing would—if normal—exclude the likelihood that the erectile dys-
function is a consequence of disease of the ANS. The cause thereof would
have to be sought elsewhere. Similarly it is extremely unusual to find
gastroparesis secondary to DAN in a patient with normal cardiovascular
autonomic reflexes.
Alternative Neuropathy Assessments 263
4.3.7 Sudorimetry
Small unmyelinated C- and Aδ-fibers in the periphery subserve somatic and
autonomic nerve functions such as warm, cold, and pain perception as well
as innervating sweat glands. The gold standard for their evaluation has been
skin biopsy and quantification of IENF density, an invasive procedure
(Pittenger et al., 2004; Vinik et al., 2013). On the other hand, the evaluation
of sweating has the appeal of quantifiable noninvasive determination of the
integrity of the ANS. Sudomotor nerves are thin unmyelinated C-fibers that
are primarily cholinergic at the ganglion although, epinephrine, norepi-
nephrine, vasoactive intestinal peptide, atrial natriuretic peptide, calcitonin
gene-related polypeptide, galanin, ATP, and substance P have also been
localized to periglandular nerves (Freeman & Chapleau, 2013). A variety
of techniques have capitalized on quantifying sweating (Illigens &
Gibbons, 2009) or the innervation of sweat glands (Gibbons, Illigens,
Wang, & Freeman, 2009; Wang & Gibbons, 2013) to capture the impact
of disordered regulation on structure and function of sweat glands as surro-
gates for the underlying autonomic dysfunction.
Alternative Neuropathy Assessments 265
Sudoscan
Symmetry
Conductances Autonomic
measured on neuropathy
hands and feet (LF HRV model)
Fig. 4 Display of electrochemical skin conductance (ESC) results for the assessment
of sudomotor function. Numeric ESC scores are provided in microSiemens and visually
displayed on a scale of normative ranges. Asymmetry represents the percent difference
in ESC measures between left and right limbs. The cardiac neuropathy risk score (pCN) is
calculated from the measured ESC and patient characteristics.
(2013), ESC in the feet had a very high negative predictive value (83%), and
significantly correlated with both symptoms and signs assessed using the
MNSI and the Utah early neuropathy symptom scale. ESC measurement
may also be a simple tool for early identification of peripheral autonomic
neuropathy and useful in screening for subclinical CAN. Some of the major
studies completed using ESC measurement in a clinical diagnostic capacity
are summarized in Table 6.
performed twice on three different devices demonstrated that, for feet ESC,
the mean coefficient of variation for repeatability was 2.8 1.6% in healthy
volunteers and 6.9 6.3% in patients with type 2 diabetes while coefficient
of variation for reproducibility were 3.1 1.5% and 6.9 6.3%, respectively
(submitted for publication).
Table 8 Change Between Baseline and 12-Month Follow-Up in Weight, Waist, VO2max,
and ESC Risk Score Value in 154 Women Included in a Lifestyle Intervention Program
(Raisanen, Eklund, Calvet, & Tuomilehto, 2014)
Without Follow- Low Weekly
Up of Training Activitya High Weekly
Level (n 5 72) (n 5 62) Activityb (n 5 20)
Mean SD Mean SD Mean SD p
Change in weight (kg) 0.9 3.4 1.6 4.0 3.3 4.8 NS
Change in waist (cm) 2.1 4.7 2.4 4.5 3.6 5.9 NS
Change in estimated +0.5 0.9 +0.8 0.9 +1.1 1.2 NS
VO2max (METs)
Change in hand ESC (μS) +5.0 8.4 +3.0 9.4 +8.4 12.3 0.043
Change in foot ESC (μS) +5.6 8.9 +4.9 8.9 +10.8 12.8 0.024
Change in ESC risk score (%) 5.1 5.3 4.7 6.4 8.5 6.8 0.027
a
Less than 150 min of moderate activity and 75 min of high activity.
b
More than 150 min of moderate activity or 75 min of high activity; moderate activity 3–7 METs, high
activity > 7 METs.
receiving insulin. Type 1 patients taking insulin therapy also had a nonsignif-
icant increase in their feet ESC (Calvet, Dupin, Winiecki, & Schwarz, 2013).
Raisanen et al. (2014) demonstrated that a 12-month lifestyle intervention
could improve small-fiber function in metabolic syndrome. Among 154
female participants with the lowest fitness level at baseline, those performing
the highest level of weekly activity showed the greatest improvement in ESC,
which was more pronounced than the changes in weight, waist circumfer-
ence, or VO2max (Table 8). These, and other ongoing studies, indicate that
ESC may provide information on the status of small nerve fiber function that
may not otherwise be known to or measurable by the physician.
5. CONCLUDING REMARKS
Diabetic neuropathy is the most common complication of diabetes
and contributes additional risks in the aging adult. For eons, the focus has
been on pain and loss of sensory perception leading to development of foot
ulcers and amputations. There is now recognition that a broader view of
neuropathy needs to be taken. Loss of sensory perception, loss of muscle
strength, and ataxia or incoordination lead to an increased risk of falling
Alternative Neuropathy Assessments 271
17-fold greater in the older diabetic than their young nondiabetic counter-
parts. A fall is accompanied by lacerations, tears, fractures and, worst of all,
traumatic brain injury from which more than 60% do not recover. There
clearly is an increased need for early recognition of people at risk and to
embark on preventive measures. Autonomic neuropathy has been hailed
as the “Prophet of Doom” (Vinik et al., 2011) for good reason. It is condu-
cive to increased risk of MI and sudden death. It is therefore critical that we
identify people with cardiac autonomic neuropathy and embark on preven-
tive strategies. It also has been recognized that an imbalance in the ANS
occurs early in the evolution of diabetes at a stage when active intervention
can abrogate the relentless progression. Many new and emerging syndromes
can be attributed to CAN such as orthostatic tachycardia and bradycardia in
addition to hypotension. While pain is a common and debilitating accom-
paniment of neuropathy, providers have failed to recognize the importance
of the accompanying sleep deprivation, anxiety, and depression which, if
unattended, compromise appropriate management of pain. Ultimately the
constellation of features of neuropathy conspires to reduce QOL and
ADL. Thus, meticulous evaluation of DSPN and DAN in the aging diabetic
patient at diagnosis and then on a yearly basis is critical, in addition to falls
risk assessment and determination of autonomic integrity. Early neuropathy
detection can only be achieved by assessment of small-nerve fibers. New
noninvasive sudomotor function technologies may play an increasing role
in identifying early peripheral and autonomic neuropathy, allowing rapid
intervention and potentially prevention or reversal of small-fiber loss.
REFERENCES
Abbott, C. A., Carrington, A. L., Ashe, H., Bath, S., Every, L. C., Griffiths, J., et al. (2002).
The North-West Diabetes Foot Care Study: Incidence of, and risk factors for, new dia-
betic foot ulceration in a community-based patient cohort. Diabetic Medicine, 19,
377–384.
Abbott, C. A., Vileikyte, L., Williamson, S., Carrington, A. L., & Boulton, A. J. (1998). Mul-
ticenter study of the incidence of and predictive risk factors for diabetic neuropathic foot
ulceration. Diabetes Care, 21, 1071–1075.
Airaksinen, K. E. J., & Koistinen, M. J. (1992). Association between silent coronary artery
disease, diabetes, and autonomic neuropathy: Fact or fallacy? Diabetes Care, 15, 288–292.
Albers, A. R., Krichavsky, M. Z., & Balady, G. J. (2006). Stress testing in patients with dia-
betes mellitus: Diagnostic and prognostic value. Circulation, 113, 583–592.
Allet, L., Armand, S., de Bie, R. A., Golay, A., Monnin, D., Aminian, K., et al. (2010). The
gait and balance of patients with diabetes can be improved: A randomised controlled trial.
Diabetologia, 53, 458–466.
Allet, L., Armand, S., de Bie, R. A., Golay, A., Pataky, Z., Aminian, K., et al. (2009). Clinical
factors associated with gait alterations in diabetic patients. Diabetic Medicine, 26,
1003–1009.
272 A.I. Vinik et al.
Buse, J. B., Bigger, J. T., Byington, R. P., Cooper, L. S., Cushman, W. C.,
Friedewald, W. T., et al. (2007). Action to Control Cardiovascular Risk In Diabetes
(ACCORD) trial: Design and methods. The American Journal of Cardiology, 99, 21i–33i.
Calvet, J. H., Dupin, J., Winiecki, H., & Schwarz, P. E. (2013). Assessment of small fiber
neuropathy through a quick, simple and non invasive method in a German diabetes out-
patient clinic. Experimental and Clinical Endocrinology & Diabetes, 121, 80–83.
Canto, J. G., Shlipak, M. G., Rogers, W. J., Malmgren, J. A., Frederick, P. D.,
Lambrew, C. T., et al. (2000). Prevalence, clinical characteristics, and mortality among
patients with myocardial infarction presenting without chest pain. JAMA, 283,
3223–3229.
Caputo, G. M., Cavanagh, P. R., Ulbrecht, J. S., Gibbons, G. W., & Karchmer, A. W.
(1994). Assessment and management of foot disease in patients with diabetes. The
New England Journal of Medicine, 331, 854–860.
Casanova-Molla, J., Grau-Junyent, J. M., Morales, M., & Valls-Sole, J. (2011). On the rela-
tionship between nociceptive evoked potentials and intraepidermal nerve fiber density in
painful sensory polyneuropathies. Pain, 152, 410–418.
Casellini, C. M., Barlow, P. M., Rice, A. L., Casey, M., Simmons, K., Pittenger, G., et al.
(2007). A 6-month, randomized, double-masked, placebo-controlled study evaluating
the effects of the protein kinase C-{beta} inhibitor Ruboxistaurin on skin microvascular
blood flow and other measures of diabetic peripheral neuropathy. Diabetes Care, 30,
896–902.
Casellini, C. M., Parson, H. K., Hodges, K., Edwards, J. F., Lieb, D. C., Wohlgemuth, S. D.,
et al. (2015). Bariatric surgery restores somatic and autonomic nerve function towards
normal in obese patients with and without diabetes. In AACE 24th annual scientific clinical
congress, May 13–17. Abstract.
Casellini, C. M., Parson, H. K., Richardson, M. S., Nevoret, M. L., & Vinik, A. I. (2013).
SUDOSCAN, a noninvasive tool for detecting diabetic small fiber neuropathy and auto-
nomic dysfunction. Diabetes Technology & Therapeutics, 15, 948–953.
Cavanagh, P. R., Derr, J. A., Ulbrecht, J. S., Maser, R. E., & Orchard, T. J. (1992). Problems
with gait and posture in neuropathic patients with insulin-dependent diabetes mellitus.
Diabetic Medicine, 9, 469–474.
Cavanagh, P. R., Simoneau, G. G., & Ulbrecht, J. S. (1993). Ulceration, unsteadiness, and
uncertainty: The biomechanical consequences of diabetes mellitus. Journal of Biochemistry,
26(Suppl. 1), 23–40.
Chao, C. C., Hsieh, S. C., Tseng, M. T., Chang, Y. C., & Hsieh, S. T. (2008). Patterns of
contact heat evoked potentials (CHEP) in neuropathy with skin denervation: Correla-
tion of CHEP amplitude with intraepidermal nerve fiber density. Clinical Neurophysiol-
ogy, 119, 653–661.
Chao, C. C., Tseng, M. T., Lin, Y. J., Yang, W. S., Hsieh, S. C., Lin, Y. H., et al. (2010).
Pathophysiology of neuropathic pain in type 2 diabetes: Skin denervation and contact
heat-evoked potentials. Diabetes Care, 33, 2654–2659.
Chen, I. A., Hung, S. W., Chen, Y. H., Lim, S. N., Tsai, Y. T., Hsiao, C. L., et al. (2006).
Contact heat evoked potentials in normal subjects. Acta Neurologica Taiwanica, 15,
184–191.
Chen, A. C., Niddam, D. M., & rendt-Nielsen, L. (2001). Contact heat evoked potentials as a
valid means to study nociceptive pathways in human subjects. Neuroscience Letters, 316,
79–82.
Chiles, N. S., Phillips, C. L., Volpato, S., Bandinelli, S., Ferrucci, L., Guralnik, J. M., et al.
(2014). Diabetes, peripheral neuropathy, and lower-extremity function. Journal of
Diabetes and Its Complications, 28, 91–95.
Clark, R. D., Lord, S. R., & Webster, I. W. (1993). Clinical parameters associated with falls
in an elderly population. Gerontology, 39, 117–123.
274 A.I. Vinik et al.
Close, J. C., Lord, S. L., Menz, H. B., & Sherrington, C. (2005). What is the role of falls? Best
Practice & Research. Clinical Rheumatology, 19, 913–935.
Colberg, S. (2006). The impact of exercise on insulin action in type 2 diabetes mellitus:
Relationship to prevention and control. Insulin, 1, 85–98.
Colberg, S. R., Parson, H. K., Nunnold, T., Holton, D. R., Swain, D. P., & Vinik, A. I.
(2005). Change in cutaneous perfusion following 10 weeks of aerobic training in type
2 diabetes. Journal of Diabetes and Its Complications, 19, 276–283.
Colberg, S., Stansberry, K., McNitt, P., & Vinik, A. (2002). Chronic exercise is associated
with enhanced cutaneous blood flow in type 2 diabetes. Journal of Diabetes and Its Com-
plications, 16, 139–145.
Colberg, S., Swain, D., & Vinik, A. (2003). Use of heart rate reserve and rating of perceived
exertion to prescribe exercise intensity in diabetic autonomic neuropathy. Diabetes Care,
26, 986–990.
Corriveau, H., Prince, F., Hebert, R., Raiche, M., Tessier, D., Maheux, P., et al. (2000).
Evaluation of postural stability in elderly with diabetic neuropathy. Diabetes Care, 23,
1187–1191.
DCCT EDIC Research Group. (2003). Sustained effect of intensive treatment of type 1 dia-
betes mellitus on development and progression of diabetic nephropathy. JAMA, 290,
2159–2167.
DCCT Research Group. (1993). The effect of intensive treatment of diabetes on the devel-
opment and progression of long-term complications in insulin dependent diabetes
mellitus. The New England Journal of Medicine, 329, 977–986.
DCCT Research Group. (1995). The effect of intensive diabetes therapy on the development
and progression of neuropathy. Annals of Internal Medicine, 122, 561–568.
Diabetes Control and Complications Trial Research Group. (1998). The effect of intensive
diabetes therapy on measures of autonomic nervous system function in the Diabetes
Control And Complications Trial (DCCT). Diabetologia, 41, 416–423.
Didangelos, T. P., Arsos, G. A., Karamitsos, D. T., Athyros, V. G., Georga, S. D., &
Karatzas, N. D. (2006). Effect of quinapril or losartan alone and in combination on left
ventricular systolic and diastolic functions in asymptomatic patients with diabetic auto-
nomic neuropathy. Journal of Diabetes and Its Complications, 20, 1–7.
Dinh, W., Futh, R., Lankisch, M., Bansemir, L., Nickl, W., Scheffold, T., et al. (2011).
Cardiovascular autonomic neuropathy contributes to left ventricular diastolic dysfunc-
tion in subjects with type 2 diabetes and impaired glucose tolerance undergoing coronary
angiography. Diabetic Medicine, 28, 311–318.
Dros, J., Wewerinke, A., Bindels, P. J., & van Weert, H. C. (2009). Accuracy of monofil-
ament testing to diagnose peripheral neuropathy: A systematic review. Annals of Family
Medicine, 7, 555–558.
Duckworth, W., Abraira, C., Moritz, T., Reda, D., Emanuele, N., Reaven, P. D., et al.
(2009). Glucose control and vascular complications in veterans with type 2 diabetes.
The New England Journal of Medicine, 360, 129–139.
Dyck, P. J., Dyck, P. J., Larson, T. S., O’Brien, P. C., & Velosa, J. A. (2000). Patterns of
quantitative sensation testing of hypoesthesia and hyperalgesia are predictive of diabetic
polyneuropathy: A study of three cohorts. Nerve growth factor study group. Diabetes
Care, 23, 510–517.
Dyck, P. J., Kratz, K. M., Karnes, J. L., Litchy, W. J., Klein, R., Pach, J. M., et al. (1993). The
prevalence by staged severity of various types of diabetic neuropathy, retinopathy, and
nephropathy in a population-based cohort: The Rochester Diabetic Neuropathy Study.
Neurology, 43, 817–824.
Dyck, P. J., Lais, A., Karnes, J. L., O’Brien, P., & Rizza, R. (1986). Fiber loss is primary
and multifocal in sural nerves in diabetic polyneuropathy. Annals of Neurology, 19, 425–439.
Alternative Neuropathy Assessments 275
Ebadi, H., Perkins, B. A., Katzberg, H. D., Lovblom, L. E., & Bril, V. (2012). Evaluation of
proxy tests for SFSN: Evidence for mixed small and large fiber dysfunction. PLoS One, 7,
e42208.
England, J. D., Gronseth, G. S., Franklin, G., Carter, G. T., Kinsella, L. J., Cohen, J. A., et al.
(2009). Practice parameter: Evaluation of distal symmetric polyneuropathy: Role of
autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report
of the American Academy of Neurology, American Association of Neuromuscular
and Electrodiagnostic Medicine, and American Academy of Physical Medicine and
Rehabilitation. Neurology, 72, 177–184.
England, J. D., Gronseth, G. S., Franklin, G., Miller, R. G., Asbury, A. K., Carter, G. T.,
et al. (2005). Distal symmetric polyneuropathy: A definition for clinical research: Report
of the American Academy of Neurology, the American Association of Electrodiagnostic
Medicine, and the American Academy of Physical Medicine and Rehabilitation.
Neurology, 64, 199–207.
Eranki, V. G., Santosh, R., Rajitha, K., Pillai, A., Sowmya, P., Dupin, J., et al. (2013).
Sudomotor function assessment as a screening tool for microvascular complications in
type 2 diabetes. Diabetes Research in Clinical Practice, 101, e11–e13.
Ewing, D. J., Boland, O., Neilson, J. M., Cho, C. G., & Clarke, B. F. (1991). Autonomic
neuropathy, QT interval lengthening, and unexpected deaths in male diabetic patient.
Diabetologia, 34, 182–185.
Ewing, D. J., & Clarke, B. F. (1986). Diabetic autonomic neuropathy: Present insights and
future prospects. Diabetes Care, 9, 648–665.
Feldman, E. L., Stevens, M. J., Thomas, P. K., Brown, M. B., Canal, N., & Greene, D. A.
(1994). A practical two-step quantitative clinical and electrophysiological assessment for
the diagnosis and staging of diabetic neuropathy. Diabetes Care, 17, 1281–1289.
Fioretti, S., Scocco, M., Ladislao, L., Ghetti, G., & Rabini, R. A. (2010). Identification
of peripheral neuropathy in type-2 diabetic subjects by static posturography and linear
discriminant analysis. Gait & Posture, 32, 317–320.
Freeman, R., & Chapleau, M. W. (2013). Testing the autonomic nervous system. Handbook
of Clinical Neurology, 115, 115–136.
Gaede, P., Vedel, P., Parving, H. H., & Pedersen, O. (1999). Intensified multifactorial inter-
vention in patients with type 2 diabetes mellitus and microalbuminuria: The steno type 2
randomized study. Lancet, 353, 617–622.
Gerritsen, J., Dekker, J. M., TenVoorde, B. J., Kostense, P. J., Heine, R. J., Bouter, L. M.,
et al. (2001). Impaired autonomic function is associated with increased mortality, espe-
cially in subjects with diabetes, hypertension, or a history of cardiovascular disease: The
Hoorn study. Diabetes Care, 24, 1793–1798.
Gibbons, C. H., Freeman, R., & Veves, A. (2010). Diabetic neuropathy: A cross-
sectional study of the relationships among tests of neurophysiology. Diabetes Care, 33,
2629–2634.
Gibbons, C. H., Illigens, B. M., Wang, N., & Freeman, R. (2009). Quantification of sweat
gland innervation: A clinical-pathologic correlation. Neurology, 72, 1479–1486.
Gin, H., Baudoin, R., Raffaitin, C. H., Rigalleau, V., & Gonzalez, C. (2011). Non-invasive
and quantitative assessment of sudomotor function for peripheral diabetic neuropathy
evaluation. Diabetes & Metabolism, 37, 527–532.
Gopalakrishnan, R., Machado, A. G., Burgess, R. C., & Mosher, J. C. (2013). The use of
contact heat evoked potential stimulator (CHEPS) in magnetoencephalography for pain
research. Journal of Neuroscience Methods, 220, 55–63.
Green, A. Q., Krishnan, S. T., & Rayman, G. (2009). C-fiber function assessed by the laser
Doppler imager flare technique and acetylcholine iontophoresis. Muscle & Nerve, 40,
985–991.
276 A.I. Vinik et al.
Haanpaa, M. L., Backonja, M. M., Bennett, M. I., Bouhassira, D., Cruccu, G.,
Hansson, P. T., et al. (2009). Assessment of neuropathic pain in primary care. The Amer-
ican Journal of Medicine, 122, S13–S21.
Harris, M., Eastman, R., & Cowie, C. (1993). Symptoms of sensory neuropathy in adults
with NIDDM in the U.S. population. Diabetes Care, 16, 1446–1452.
Herman, W. H., & Kennedy, L. (2005). Underdiagnosis of peripheral neuropathy in type 2
diabetes. Diabetes Care, 28, 1480–1481.
Herriott, M. T., Colberg, S. R., Parson, H. K., Nunnold, T., & Vinik, A. I. (2004). Effects of
8 weeks of flexibility and resistance training in older adults with type 2 diabetes. Diabetes
Care, 27, 2988–2989.
Holzer, S. E., Camerota, A., Martens, L., Cuerdon, T., Crystal, P., & Zagari, M. (1998).
Costs and duration of care for lower extremity ulcers in patients with diabetes. Clinical
Therapeutics, 20, 169–181.
Howe, T., Rochester, L., Jackson, A., Banks, P., & Blair, V. (2007). Exercise for improving
balance in older people. The Cochrane Database of Systematic Reviews, 17, CD004963.
Illigens, B. M., & Gibbons, C. H. (2009). Sweat testing to evaluate autonomic function.
Clinical Autonomic Research, 19, 79–87.
Itskovich, V. V., Fei, D. Y., & Harkins, S. W. (2000). Psychophysiological and psychophys-
ical responses to experimental pain induced by two types of cutaneous thermal stimuli.
The International Journal of Neuroscience, 105, 63–75.
Joint Task Force of the EFNS and the PNS. (2010). European Federation of Neurological
Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis
of small fiber neuropathy. Report of a joint task force of the European Federation of
Neurological Societies and the Peripheral Nerve Society. Peripheral Nervous System,
15, 79–92.
Karason, K., Molgaard, H., Wikstrand, J., & Sjostrom, L. (1999). Heart rate variability
in obesity and the effect of weight loss. The American Journal of Cardiology, 83,
1242–1247.
Katoulis, E. C., Ebdon-Parry, M., Lanshammar, H., Vileikyte, L., Kulkarni, J., &
Boulton, A. J. (1997). Gait abnormalities in diabetic neuropathy. Diabetes Care, 20,
1904–1907.
Kellogg, A. P., Converso, K., Wiggin, T., Stevens, M., & Pop-Busui, R. (2009). Effects of
cyclooxygenase-2 gene inactivation on cardiac autonomic and left ventricular function
in experimental diabetes. American Journal of Physiology. Heart and Circulatory Physiology,
296, H453–H461.
Kendall, D. M., Rooney, D. P., Smets, Y. F., Salazar Bolding, L., & Robertson, R. P. (1997).
Pancreas transplantation restores epinephrine response and symptom recognition during
hypoglycemia in patients with long-standing type I diabetes and autonomic neuropathy.
Diabetes, 46, 249–257.
Kluding, P. M., Pasnoor, M., Singh, R., Jernigan, S., Farmer, K., Rucker, J., et al. (2012).
The effect of exercise on neuropathic symptoms, nerve function, and cutaneous inner-
vation in people with diabetic peripheral neuropathy. Journal of Diabetes and Its Compli-
cations, 26, 424–429.
Kramer, J. L., Jutzeler, C. R., Haefeli, J., Curt, A., & Freund, P. (2015). Discrepancy
between perceived pain and cortical processing: A voxel-based morphometry and con-
tact heat evoked potential study. Clinical Neurophysiology, 127, 762–768.
Kramer, J. L., Taylor, P., Haefeli, J., Blum, J., Zariffa, J., Curt, A., et al. (2012). Test-retest
reliability of contact heat-evoked potentials from cervical dermatomes. Journal of Clinical
Neurophysiology, 29, 70–75.
Krishnan, S. T., & Rayman, G. (2004). The LDIflare: A novel test of C-fiber
function demonstrates early neuropathy in type 2 diabetes. Diabetes Care, 27(12),
2930–2935.
Alternative Neuropathy Assessments 277
Kumar, S., Fernando, D. J., Veves, A., Knowles, E. A., Young, M. J., & Boulton, A. J.
(1991). Semmes-Weinstein monofilaments: A simple, effective and inexpensive screen-
ing device for identifying diabetic patients at risk of foot ulceration. Diabetes Research and
Clinical Practice, 13, 63–67.
Lafond, D., Corriveau, H., & Prince, F. (2004). Postural control mechanisms during quiet
standing in patients with diabetic sensory neuropathy. Diabetes Care, 27, 173–178.
Lagerburg, V., Bakkers, M., Bouwhuis, A., Hoeijmakers, J. G., Smit, A. M., Van Den
Berg, S. J., et al. (2015). Contact heat evoked potentials: Normal values and use in
small-fiber neuropathy. Muscle & Nerve, 51, 743–749.
Lai, S., Ahmed, U., Bollineni, A., Lewis, R., & Ramchandren, S. (2014). Diagnostic accu-
racy of qualitative versus quantitative tuning forks: Outcome measure for neuropathy.
Journal of Clinical Neuromuscular Disease, 15, 96–101.
Lalli, P., Chan, A., Garven, A., Midha, N., Chan, C., Brady, S., et al. (2013). Increased gait
variability in diabetes mellitus patients with neuropathic pain. Journal of Diabetes and Its
Complications, 27, 248–254.
Lambert, E., Sari, C. I., Dawood, T., Nguyen, J., McGrane, M., Eikelis, N., et al. (2010).
Sympathetic nervous system activity is associated with obesity-induced subclinical organ
damage in young adults. Hypertension, 56, 351–358.
Lambert, G. W., Straznicky, N. E., Lambert, E. A., Dixon, J. B., & Schlaich, M. P. (2010).
Sympathetic nervous activation in obesity and the metabolic syndrome—Causes, con-
sequences and therapeutic implications. Pharmacology & Therapeutics, 126, 159–172.
Lauria, G., Bakkers, M., Schmitz, C., Lombardi, R., Penza, P., Devigili, G., et al. (2010).
Intraepidermal nerve fiber density at the distal leg: A worldwide normative reference
study. Journal of the Peripheral Nervous System, 15, 202–207.
Lauria, G., Hsieh, S., Johansson, O., Kennedy, W., Leger, J., Mellgren, S., et al. (2010).
European Federation of Neurological Societies/Peripheral Nerve Society Guideline
on the use of skin biopsy in the diagnosis of small fiber neuropathy. Report of a joint
task force of the European Federation of Neurological Societies and the Peripheral Nerve
Society. European Journal of Neurology, 17, 903–912.
Levitt, N. S., Stansberry, K. B., Wychanck, S., & Vinik, A. I. (1996). Natural progression of
autonomic neuropathy and autonomic function tests in a cohort of IDDM. Diabetes Care,
19, 751–754.
Lord, S. (1996). Instability and falls in elderly people. In C. Lafont, A. Baroni, & M. Allard
et al. (Eds.), Falls, gait, and balance disorders in the elderly (pp. 125–139). New York:
Springer.
Lord, S. R., & Clark, R. D. (1996). Simple physiological and clinical tests for the accurate
prediction of falling in older people. Gerontology, 42, 199–203.
Lykke, J. A., Tarnow, L., Parving, H. H., & Hilsted, J. (2008). A combined abnormality
in heart rate variation and QT corrected interval is a strong predictor of cardiovascular
death in type 1 diabetes. Scandinavian Journal of Clinical and Laboratory Investigation, 68,
654–659.
Malmberg, K., Norhammar, A., Wedel, H., & Ryden, L. (1999). Glycometabolic state at
admission: Important risk marker of mortality in conventionally treated patients with dia-
betes mellitus and acute myocardial infarction: Long-term results from the Diabetes and
Insulin-Glucose Infusion in Acute Myocardial Infarction (DIGAMI) study. Circulation,
99, 2626–2632.
Marchant, B., Umachandran, V., Stevenson, R., Kopelman, P. G., & Timmis, A. D. (1993).
Silent myocardial ischemia: Role of subclinical neuropathy in patients with and without
diabetes. Journal of the American College of Cardiology, 22, 1433–1437.
Margolis, J. R., Kannel, W. S., Feinleib, M., Dawber, T. R., & McNamara, P. M. (1973).
Clinical features of unrecognized myocardial infarction—Silent and symptomatic. Eigh-
teen year follow-up: The Framingham study. The American Journal of Cardiology, 32, 1–7.
278 A.I. Vinik et al.
Martin, C. L., Waberski, B. H., Pop-Busui, R., Cleary, P. A., Catton, S., Albers, J. W., et al.
(2010). Vibration perception threshold as a measure of distal symmetrical peripheral neu-
ropathy in type 1 diabetes: Results from the DCCT/EDIC study. Diabetes Care, 33,
2635–2641.
Maser, R., Lenhard, M., & DeCherney, G. (2000). Cardiovascular autonomic neuropathy:
The clinical significance of its determination. Endocrinologist, 10, 27–33.
Maser, R. E., Lenhard, M. J., Irgau, I., & Wynn, G. M. (2007). Impact of surgically induced
weight loss on cardiovascular autonomic function: One-year follow-up. Obesity (Silver
Spring), 15, 364–369.
Maser, R. E., Mitchell, B. D., Vinik, A. I., & Freeman, R. (2003). The association between
cardiovascular autonomic neuropathy and mortality in individuals with diabetes: A meta-
analysis. Diabetes Care, 26, 1895–1901.
Matsuki, A., Nozawa, T., Igarashi, N., Sobajima, M., Ohori, T., Suzuki, T., et al. (2010).
Fluvastatin attenuates diabetes-induced cardiac sympathetic neuropathy in association
with a decrease in oxidative stress. Circulation Journal, 74, 468–475.
Maurer, M. S., Burcham, J., & Cheng, H. (2005). Diabetes mellitus is associated with an
increased risk of falls in elderly residents of a long-term care facility. The Journals of
Gerontology. Series A, Biological Sciences and Medical Sciences, 60, 1157–1162.
Morrison, S., Colberg, S. R., Mariano, M., Parson, H. K., & Vinik, A. I. (2010). Balance
training reduces falls risk in older individuals with type 2 diabetes. Diabetes Care, 33,
748–750.
Morrison, S., Colberg, S. R., Parson, H. K., & Vinik, A. I. (2012). Relation between risk of
falling and postural sway complexity in diabetes. Gait & Posture, 35, 662–668.
Nather, A., Keng, L. W., Aziz, Z., Hj, O. C., Mc, F. B., & Lin, B. (2011). Assessment of
sensory neuropathy in patients with diabetic foot problems. Diabetic Foot & Ankle, 2.
Nault, I., Nadreau, E., Paquet, C., Brassard, P., Marceau, P., Marceau, S., et al. (2007).
Impact of bariatric surgery—Induced weight loss on heart rate variability. Metabolism,
56, 1425–1430.
Neil, H. A., Thompson, A. V., John, S., McCarthy, S. T., & Mann, J. I. (1989). Diabetic
autonomic neuropathy: The prevalence of impaired heart rate variability in a geograph-
ically define population. Diabetic Medicine, 6, 20–24.
Novella, S. P., Inzucchi, S. E., & Goldstein, J. M. (2001). The frequency of undiagnosed
diabetes and impaired glucose tolerance in patients with idiopathic sensory neuropathy.
Muscle & Nerve, 24, 1229–1231.
O’Brien, I. A., McFadden, J. P., & Corrall, R. J. (1991). The influence of autonomic neu-
ropathy on mortality in insulin-dependent diabetes. The Quarterly Journal of Medicine, 79,
495–502.
Opsommer, E., Masquelier, E., & Plaghki, L. (1999). Determination of nerve conduction
velocity of C-fibres in humans from thermal thresholds to contact heat (thermode)
and from evoked brain potentials to radiant heat (CO2 laser). Neurophysiologie Clinique,
29, 411–422.
Ozaki, R., Cheung, K. K., Wu, E., Kong, A., Yang, X., Lau, E., et al. (2011). A new
tool to detect kidney disease in Chinese type 2 diabetes patients: Comparison of
EZSCAN with standard screening methods. Diabetes Technology & Therapeutics, 13,
937–943.
Paisley, A. N., Abbott, C. A., van Schie, C. H. M., & Boulton, A. J. M. (2002). A comparison
of the Neuropen against standard quantitative sensory threshold measures for assessing
peripheral nerve function. Diabetic Medicine, 19, 400–405.
Papanas, N., Papatheodorou, K., Christakidis, D., Papazoglou, D., Giassakis, G.,
Piperidou, H., et al. (2005). Evaluation of a new indicator test for sudomotor function
(Neuropad) in the diagnosis of peripheral neuropathy in type 2 diabetic patients. Exper-
imental and Clinical Endocrinology & Diabetes, 113, 195–198.
Alternative Neuropathy Assessments 279
Parson, H. K., Nguyen, V. T., Boyd, A. L., & Vinik, A. (2009). CHEPS detects neuropathic
changes earlier than traditional clinical measures. Diabetes, 58(Suppl.), 829P.
Parson, H. K., Nguyen, V. T., Orciga, M. A., Boyd, A. L., Casellini, C. M., & Vinik, A. I.
(2013). Contact heat-evoked potential stimulation for the evaluation of small nerve fiber
function. Diabetes Technology & Therapeutics, 15, 150–157.
Partanen, J., Niskanen, L., Lehtinen, J., Mervaala, E., Siitonen, O., & Uusitupa, M. (1995).
Natural history of peripheral neuropathy in patients with non-insulin-dependent diabe-
tes mellitus. The New England Journal of Medicine, 333, 89–94.
Patel, A., MacMahon, S., Chalmers, J., Neal, B., Billot, L., Woodward, M., et al. (2008).
Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes.
The New England Journal of Medicine, 358, 2560–2572.
Peron, E. P., & Ogbonna, K. C. (2015). Diabetic medications and polypharmacy. Clinics of
Geriatric Medicine, 31, 17–27.
Perugini, R. A., Li, Y., Rosenthal, L., Gallagher-Dorval, K., Kelly, J. J., & Czerniach, D. R.
(2010). Reduced heart rate variability correlates with insulin resistance but not with
measures of obesity in population undergoing laparoscopic Roux-en-Y gastric bypass.
Surgery for Obesity and Related Diseases, 6, 237–241.
Peterson, M. J., Giuliani, C., Morey, M. C., Pieper, C. F., Evenson, K. R., Mercer, V., et al.
(2009). Physical activity as a preventative factor for frailty: The health, aging, and body
composition study. The Journals of Gerontology. Series A, Biological Sciences and Medical
Sciences, 64, 61–68.
Pfeifer, M. A., Weinberg, C. R., Cook, D. L., Reenan, A., Halter, J. B., & Ensinck, J. W.
(1984). Autonomic neural dysfunction in recently diagnosed diabetic subjects. Diabetes
Care, 7, 447–453.
Pickering, R. M., Grimbergen, Y. A., Rigney, U., Ashburn, A., Mazibrada, G., Wood, B.,
et al. (2007). A meta-analysis of six prospective studies of falling in Parkinson’s disease.
Movement Disorders, 22, 1892–1900.
Piestrzeniewicz, K., Luczak, K., Lelonek, M., Wranicz, J. K., & Goch, J. H. (2008).
Obesity and heart rate variability in men with myocardial infarction. Cardiology Journal,
15, 43–49.
Pijpers, E., Ferreira, I., de Jongh, R. T., Deeg, D. J., Lips, P., Stehouwer, C. D., et al. (2012).
Older individuals with diabetes have an increased risk of recurrent falls: Analysis of
potential mediating factors: The Longitudinal Ageing Study Amsterdam. Age and Ageing,
41, 358–365.
Pittenger, G., Mehrabyan, A., Simmons, K., Rice, A., Dublin, C., Barlow, P., et al. (2005).
Small fiber neuropathy is associated with the metabolic syndrome. Metabolic Syndrome and
Related Disorders, 3, 113–121.
Pittenger, G. L., Ray, M., Burcus, N. I., McNulty, P., Basta, B., & Vinik, A. I. (2004). Intra-
epidermal nerve fibers are indicators of small-fiber neuropathy in both diabetic and
nondiabetic patients. Diabetes Care, 27, 1974–1979.
Pittenger, G., Simmons, K., Anandacoomaraswamy, D., Rice, A., Barlow, P., & Vinik, A.
(2005). Topiramate improves intraepidermal nerve fiber morphology and quantitative
neuropathy measures in diabetic neuropathy patients. Journal of the Peripheral Nervous
System, 10(Suppl. 1), 1–108.
Pop-Busui, R., Low, P. A., Waberski, B. H., Martin, C. L., Albers, J. W., Feldman, E. L.,
et al. (2009). Effects of prior intensive insulin therapy on cardiac autonomic nervous sys-
tem function in type 1 diabetes mellitus: The Diabetes Control and Complications Trial/
Epidemiology of Diabetes Interventions and Complications study (DCCT/EDIC).
Circulation, 119, 2886–2893.
Quattrini, C., Tavakoli, M., Jeziorska, M., Kallinikos, P., Tesfaye, S., Finnigan, J., et al.
(2007). Surrogate markers of small fiber damage in human diabetic neuropathy.
Diabetes, 56, 2148–2154.
280 A.I. Vinik et al.
Raisanen, A., Eklund, J., Calvet, J.-H., & Tuomilehto, J. (2014). Sudomotor function as a
tool for cardiorespiratory fitness level evaluation: Comparison with VO2 max. Interna-
tional Journal of Environmental Research and Public Health, 11, 5839–5848.
Rathmann, W., Ziegler, D., Jahnke, M., Haastert, B., & Gries, F. A. (1993). Mortality in
diabetic patients with cardiovascular autonomic neuropathy. Diabetic Medicine, 10,
820–824.
Ravussin, E. (2010). Impact of six month caloric restriction on autonomic nervous system in
healthy, overweight individuals. Obesity, 18, 414–416.
Resnick, H. E., Stansberry, K. B., Harris, T. B., Tirivedi, M., Smith, K., Morgan, P., et al.
(2002). Diabetes, peripheral neuropathy, and old age disability. Muscle & Nerve, 25,
43–50.
Resnick, H., Vinik, A., Schwartz, A., Leveille, S., Brancati, F., Balfour, J., et al. (2000). Inde-
pendent effects of peripheral nerve dysfunction on lower-extremity physical function in
old age. Diabetes Care, 23, 1642–1647.
Richardson, J. K., & Hurvitz, E. A. (1995). Peripheral neuropathy: A true risk factor for falls.
The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences, 50,
M211–M215.
Richardson, J., Thies, S., & Ashton-Miller, J. (2008). An exploration of step time variability
on smooth and irregular surfaces in older persons with neuropathy. Clinical biomechanics,
23, 349–356.
Richardson, J. K., Thies, S. B., DeMott, T. K., & Ashton-Miller, J. A. (2005). Gait analysis in
a challenging environment differentiates between fallers and nonfallers among older
patients with peripheral neuropathy. Archives of Physical Medicine and Rehabilitation, 86,
1539–1544.
Robinovitch, S. N., Hsiao, E. T., Sandler, R., Cortez, J., Liu, Q., & Paiement, G. D. (2000).
Prevention of falls and fall-related fractures through biomechanics. Exercise and Sport
Sciences Reviews, 28, 74–79.
Ruscheweyh, R., Emptmeyer, K., Putzer, D., Kropp, P., & Marziniak, M. (2013). Repro-
ducibility of contact heat evoked potentials (CHEPs) over a 6 months interval. Clinical
Neurophysiology, 124, 2242–2247.
Sacre, J. W., Franjic, B., Jellis, C. L., Jenkins, C., Coombes, J. S., & Marwick, T. H. (2010).
Association of cardiac autonomic neuropathy with subclinical myocardial dysfunction in
type 2 diabetes. JACC. Cardiovascular Imaging, 3, 1207–1215.
Sadosky, A., McDermott, A. M., Brandenburg, N. A., & Strauss, M. (2008). A review of the
epidemiology of painful diabetic peripheral neuropathy, postherpetic neuralgia, and less
commonly studied neuropathic pain conditions. Pain Practice, 8, 45–56.
Schwartz, A. V., Hillier, T. A., Sellmeyer, D. E., Resnick, H. E., Gregg, E., Ensrud, K. E.,
et al. (2002). Older women with diabetes have a higher risk of falls: A prospective study.
Diabetes Care, 25, 1749–1754.
Schwartz, A. V., Vittinghoff, E., Sellmeyer, D. E., Feingold, K. R., de, R. N.,
Strotmeyer, E. S., et al. (2008). Diabetes-related complications, glycemic control, and
falls in older adults. Diabetes Care, 31, 391–396.
Selvarajah, D., Cash, T., Davies, J., Sankar, A., Rao, G., Grieg, M., et al. (2015).
SUDOSCAN: A simple, rapid, and objective method with potential for screening for
diabetic peripheral neuropathy. PloS One, 10, e0138224.
Sharma, S., Vas, P. R., & Rayman, G. (2015). Assessment of diabetic neuropathy using a
point-of-care nerve conduction device shows significant associations with the
LDIFLARE method and clinical neuropathy scoring. Journal of Diabetes Science and
Technology, 9, 123–131.
Shehab, D. K., Al-Jarallah, K. F., Abraham, M., Mojiminiyi, O. A., Al-Mohamedy, H., &
Abdella, N. A. (2012). Back to basics: Ankle reflex in the evaluation of peripheral
Alternative Neuropathy Assessments 281
neuropathy in type 2 diabetes mellitus. QJM: Monthly Journal of the Association of Physi-
cians, 105, 315–320.
Shenoy, R., Roberts, K., Papadaki, A., McRobbie, D., Timmers, M., Meert, T., et al.
(2011). Functional MRI brain imaging studies using the contact heat evoked potential
stimulator (CHEPS) in a human volunteer topical capsaicin pain model. Journal of Pain
Research, 4, 365–371.
Sherrington, C., Lord, S. R., & Herbert, R. D. (2003). A randomised trial of weight-bearing
versus non-weight-bearing exercise for improving physical ability in inpatients after hip
fracture. The Australian Journal of Physiotherapy, 49, 15–22.
Shtein, R. M., & Callaghan, B. C. (2013). Corneal confocal microscopy as a measure of dia-
betic neuropathy. Diabetes, 62, 25–26.
Shy, M. E., Frohman, E. M., So, Y., Arezzo, J. C., Cornblath, D. C., Giuliani, M. J., et al.
(2003). Quantitative sensory testing: Report on the therapeutic and technology
assessment subcommittee of the American Academy of Neurology. Neurology, 602,
898–906.
Simoneau, G. G., Ulbrecht, J., Derr, J. A., Becher, M. B., & Cavanaugh, P. R. (1994). Pos-
tural instability in patients with diabetic sensory neuropathy. Diabetes Care, 17,
1411–1421.
Singleton, J. R., Smith, A. G., & Bromberg, M. B. (2001a). Painful sensory polyneuropathy
associated with impaired glucose tolerance. Muscle & Nerve, 24, 1225–1228.
Singleton, J. R., Smith, A. G., & Bromberg, M. B. (2001b). Increased prevalence of impaired
glucose tolerance in patients with painful sensory neuropathy. Diabetes Care, 24,
1448–1453.
Sinnreich, M., Taylor, B. V., & Dyck, P. J. (2005). Diabetic neuropathies. Classification,
clinical features, and pathophysiological basis. The Neurologist, 11, 63–79.
Smith, A. G., Lessard, M., Reyna, S., Doudova, M., & Singleton, J. R. (2014). The diag-
nostic utility of SUDOSCAN for distal symmetric peripheral neuropathy. Journal of Dia-
betes and Its Complications, 28, 511–516.
Smith, A. G., Russell, J., Feldman, E. L., Goldstein, J., Peltier, A., Smith, S., et al. (2006).
Lifestyle intervention for pre-diabetic neuropathy. Diabetes Care, 29, 1294–1299.
Sosnoff, J., Motl, R., & Morrison, S. (2013). Multiple sclerosis and falls—An evolving tale.
US Neurology, 9, 30–34.
Sosnoff, J. J., Socie, M. J., Boes, M. K., Sandroff, B. M., Pula, J. H., Suh, Y., et al. (2011).
Mobility, balance and falls in persons with multiple sclerosis. PloS One, 6, e28021.
Stevens, J. A., Corso, P. S., Finkelstein, E. A., & Miller, T. R. (2006). The costs of fatal and
non-fatal falls among older adults. Injury Prevention, 12, 290–295.
Stewart, J. M., Medow, M. S., & Montgomery, L. D. (2003). Local vascular responses affect-
ing blood flow in postural tachycardia syndrome. American Journal of Physiology. Heart and
Circulatory Physiology, 285, H2749–H2756.
Straznicky, N. E., Lambert, G. W., McGrane, M. T., Masuo, K., Dawood, T., Nestel, P. J.,
et al. (2009). Weight loss may reverse blunted sympathetic neural responsiveness
to glucose ingestion in obese subjects with metabolic syndrome. Diabetes, 58,
1126–1132.
Strotmeyer, E. S., de, R. N., Schwartz, A. V., Faulkner, K. A., Resnick, H. E.,
Goodpaster, B. H., et al. (2008). The relationship of reduced peripheral nerve function
and diabetes with physical performance in older white and black adults: The health,
aging, and body composition (health ABC) study. Diabetes Care, 31, 1767–1772.
Strotmeyer, E. S., de, R. N., Schwartz, A. V., Resnick, H. E., Goodpaster, B. H.,
Faulkner, K. A., et al. (2009). Sensory and motor peripheral nerve function and
lower-extremity quadriceps strength: The health, aging and body composition study.
Journal of the American Geriatrics Society, 57, 2004–2010.
282 A.I. Vinik et al.
Sumner, C. J., Sheth, S., Griffin, J. W., Cornblath, D. R., & Polydefkis, M. (2003). The
spectrum of neuropathy in diabetes and impaired glucose tolerance. Neurology, 60,
108–111.
Taksande, B., Ansari, S., Jaikrishnan, A., & Karwasara, V. (2011). The diagnostic sensitivity,
specificity, and reproducibility of the clinical physical examination signs in patients of
diabetes mellitus for making diagnosis of peripheral neuropathy. Journal of Endocrinology
and Metabolism, 1, 21–26.
Tavakoli, M., Mitu-Pretorian, M., Petropoulos, I. N., Fadavi, H., Asghar, O., Alam, U., et al.
(2013). Corneal confocal microscopy detects early nerve regeneration in diabetic neurop-
athy after simultaneous pancreas and kidney transplantation. Diabetes, 62, 254–260.
Tavakoli, M., Quattrini, C., Abbott, C., Kallinikos, P., Marshall, A., Finnigan, J., et al.
(2010). Corneal confocal microscopy: A novel noninvasive test to diagnose and stratify
the severity of human diabetic neuropathy. Diabetes Care, 33, 1792–1797.
Tesfaye, S., Boulton, A. J., Dyck, P. J., Freeman, R., Horowitz, M., Kempler, P., et al.
(2010). Diabetic neuropathies: Update on definitions, diagnostic criteria, estimation of
severity, and treatments. Diabetes Care, 33, 2285–2293.
Thaisetthawatkul, P., Fernandes Filho, J. A., & Herrmann, D. N. (2013). Contribution of
QSART to the diagnosis of small fiber neuropathy. Muscle & Nerve, 48, 883–888.
Tilling, L. M., Darawil, K., & Britton, M. (2006). Falls as a complication of diabetes mellitus
in older people. Journal of Diabetes and Its Complications, 20, 158–162.
Tucker, M. E. (2014). Laser-doppler-imager flare detects early diabetic neuropathy. www.
medscape.com/viewarticle/832229.
Turcot, K., Allet, L., Golay, A., Hoffmeyer, P., & Armand, S. (2009). Investigation of stand-
ing balance in diabetic patients with and without peripheral neuropathy using acceler-
ometers. Clinical Biomechanics (Bristol, Avon), 24, 716–721.
Uccioli, L., Giacomini, P. G., Monticone, G., Magrini, A., Durola, L., Bruno, E., et al.
(1995). Body sway in diabetic neuropathy. Diabetes Care, 18, 339–344.
Vas, P. R., & Rayman, G. (2013a). Validation of the modified LDIFlare technique: A simple
and quick method to assess C-fiber function. Muscle & Nerve, 47, 351–356.
Vas, P. R., & Rayman, G. (2013b). The rate of decline in small fibre function assessed using
axon reflex-mediated neurogenic vasodilatation and the importance of age related centile
values to improve the detection of clinical neuropathy. PLoS One, 8, e69920.
Veglio, M., Borra, M., Stevens, L. K., Fuller, J. H., & Perin, P. C. (1999). The relation
between QTc interval prolongation and diabetic complications. The EURODIAB
IDDM Complications Study Group. Diabetologia, 42, 68–75.
Veresiu, A. I., Bondor, C. I., Florea, B., Vinik, E. J., Vinik, A. I., & Gavan, N. A. (2015).
Detection of undisclosed neuropathy and assessment of its impact on quality of life:
A survey in 25,000 Romanian patients with diabetes. Journal of Diabetes and Its Compli-
cations, 29, 644–649.
Vileikyte, L., Peyrot, M., Bundy, C., Rubin, R. R., Leventhal, H., Mora, P., et al. (2003).
The development and validation of a neuropathy- and foot ulcer-specific quality of life
instrument. Diabetes Care, 26, 2549–2555.
Vinik, A. (1999). Diabetic neuropathy: Pathogenesis and therapy. The American Journal of
Medicine, 107(2B), 17S–26S.
Vinik, A., Casellini, C., & Nevoret, M.-L. (2015). Diabetic neuropathies. In L. DeGroot
(Ed.), South Dartmouth, MA: MDText.com, Inc.
Vinik, A. I., & Erbas, T. (2001). Recognizing and treating diabetic autonomic neuropathy.
Cleveland Clinic Journal of Medicine, 68, 928–944.
Vinik, A., & Erbas, T. (2002). Neuropathy. In N. Ruderman, J. T. Devlin, S. Schneider, &
A. Kriska (Eds.), Handbook of exercise in diabetes (pp. 463–495). Alexandria, VA: American
Diabetes Association.
Alternative Neuropathy Assessments 283
Vinik, A. I., & Erbas, T. (2013). Diabetic autonomic neuropathy. In R. M. Buijs &
D. F. Swaab (Eds.), Handbook of clinical neurology (3rd ed., pp. 279–294). Amsterdam,
The Netherlands: Elsevier.
Vinik, A., Erbas, T., & Pfeifer, M. A. (2003). Diabetic autonomic neuropathy. In D. Porte, Jr,
R. S. Sherwin, & A. Baron (Eds.), Ellenberg & Rifkin’s diabetes mellitus (6th ed.,
pp. 789–804). New York, NY: McGraw-Hill.
Vinik, E. J., Hayes, R. P., Oglesby, A., Bastyr, E., Barlow, P., Ford-Molvik, S. L., et al.
(2005). The development and validation of the Norfolk QOL-DN, a new measure of
patients’ perception of the effects of diabetes and diabetic neuropathy. Diabetes Technol-
ogy & Therapeutics, 7, 497–508.
Vinik, E. J., Hayes, C., Oglesby, A., & Vinik, A. I. (2004). Identification of factors in the
nerve fiber specific quality of life (QOL-DN) inventory that reflect QOL and health sta-
tus. Diabetes, 53, A295.
Vinik, A. I., Maser, R. E., & Ziegler, D. (2010). Neuropathy: The crystal ball for cardiovas-
cular disease? Diabetes Care, 33, 1688–1690.
Vinik, A., Maser, R., & Ziegler, D. (2011). Autonomic imbalance: Prophet of doom or scope
for hope? Diabetic Medicine, 28, 643–651.
Vinik, A. I., Mitchell, B. D., Leichter, S. B., Wagner, A. L., O’Brian, J. T., & Georges, L. P.
(1995). Epidemiology of the complications of diabetes. In R. D. G. Leslie &
D. C. Robbins (Eds.), Diabetes: Clinical science in practice (pp. 221–287). Cambridge,
UK: Cambridge University Press.
Vinik, A. I., Nevoret, M., Casellini, C., & Parson, H. (2013). Neurovascular function and
sudorimetry in health and disease. Current Diabetes Reports, 13, 517–532.
Vinik, E., Paulson, J., Ford-Molvik, S., & Vinik, A. (2008). German-translated Norfolk qual-
ity of life (QOL-DN) identifies the same factors as the english version of the tool and
discriminates different levels of neuropathy severity. Journal of Diabetes Science and Tech-
nology, 2, 1075–1086.
Vinik, A. I., Perrot, S., Vinik, E. J., Pazdera, L., Jacobs, H., Stoker, M., et al. (2015). Long-
term safety and efficacy of repeated applications of capsaicin 8% patch (QUTENZA™) in
painful diabetic peripheral neuropathy: Pace study. AACE ePoster Library. May 13,
2015; 97789. http://aace.multilearning.com/aace/2015/eposter/97789/aaron.vinik.
long-term.safety.and.efficacy.of.repeated.applications.of.html?f=p6m3e813o10388
Accessed 24.03.16.
Vinik, A. I., Strotmeyer, E. S., Nakave, A. A., & Patel, C. V. (2008). Diabetic neuropathy in
older adults. Clinics in Geriatric Medicine, 24, 407–435. v.
Vinik, A. I., Suwanwalaikorn, S., Stansberry, K. B., Holland, M. T., McNitt, P. M., &
Colen, L. E. (1995). Quantitative measurement of cutaneous perception in diabetic neu-
ropathy. Muscle & Nerve, 18, 574–584.
Vinik, A., Ullal, J., Parson, H. K., & Casellini, C. M. (2006). Diabetic neuropathies: Clinical
manifestations and current treatment options. Nature Clinical Practice. Endocrinology &
Metabolism, 2, 269–281.
Vinik, E. J., Vinik, A. I., Paulson, J. F., Merkies, I. S., Packman, J., Grogan, D. R., et al.
(2014). Norfolk QOL-DN: Validation of a patient reported outcome measure in trans-
thyretin familial amyloid polyneuropathy. Journal of the Peripheral Nervous System, 19,
104–114.
Vinik, A. I., & Ziegler, D. (2007). Diabetic cardiovascular autonomic neuropathy.
Circulation, 115, 387–397.
Volpato, S., Leveille, S. G., Blaum, C., Fried, L. P., & Guralnik, J. M. (2005). Risk factors
for falls in older disabled women with diabetes: The women’s health and aging study.
The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences, 60,
1539–1545.
284 A.I. Vinik et al.
Volpato, S., Maraldi, C., & Fellin, R. (2010). Type 2 diabetes and risk for functional decline
and disability in older persons. Current Diabetes Reviews, 6, 134–143.
Wallace, C., Reiber, G. E., LeMaster, J., Smith, D. G., Sullivan, K., Hayes, S., et al. (2002).
Incidence of falls, risk factors for falls, and fall-related fractures in individuals with dia-
betes and a prior foot ulcer. Diabetes Care, 25, 1983–1986.
Wang, N., & Gibbons, C. H. (2013). Skin biopsies in the assessment of the autonomic ner-
vous system. Handbook of Clinical Neurology, 117, 371–378.
Watkins, P. J. (1993). Progression of diabetic autonomic neuropathy. Diabetic Medicine,
10(Suppl. 2), 77S–78S.
Wirta, O., Pasternack, A., Mustonen, J., & Laippala, P. (1997). Renal and cardiovascular pre-
dictors of 9 year total and sudden cardiac mortality in non-insulin-dependent diabetic
subjects. Nephrology, Dialysis, Transplantation, 12, 2612–2617.
Witte, D. R., Tesfaye, S., Chaturvedi, N., Eaton, S. E., Kempler, P., & Fuller, J. H. (2005).
Risk factors for cardiac autonomic neuropathy in type 1 diabetes mellitus. Diabetologia,
48, 164–171.
Witzke, K. A., & Vinik, A. I. (2005). Diabetic neuropathy in older adults. Reviews in Endo-
crine & Metabolic Disorders, 6, 117–127.
Wong, M. C., & Chung, J. W. (2011). Feasibility of contact heat evoked potentials for detec-
tion of diabetic neuropathy. Muscle & Nerve, 44, 902–906.
Yajnik, C. S., Kantikar, V. V., Pande, A. J., & Deslypere, J. P. (2012). Quick and simple
evaluation of sudomotor function for screening of diabetic neuropathy. ISRN Endocri-
nology, 2012, 103714.
Yajnik, C. S., Kantikar, V., Pande, A., Deslypere, J. P., Dupin, J., Calvet, J. H., et al. (2013).
Screening of cardiovascular autonomic neuropathy in patients with diabetes using non-
invasive quick and simple assessment of sudomotor function. Diabetes & Metabolism, 39,
126–131.
Yarnitsky, D., & Sprecher, E. (1994). Thermal testing: Normative data and repeatability for
various test algorithms. Journal of the Neurological Sciences, 125, 39–45.
Young, M. J., Boulton, A. J. M., MacLeod, A. F., Williams, D. R. R., & Sonksen, P. H.
(1993). A multicenter study of the prevalence of diabetic peripheral neuropathy in
the United Kingdom hospital clinic population. Diabetologia, 36, 150–154.
Zhivov, A., Blum, M., Guthoff, R., & Stachs, O. (2010). Real-time mapping of the sub-
epithelial nerve plexus by in vivo confocal laser scanning microscopy. The British Journal
of Ophthalmology, 94, 1133–1135.
Ziegler, D. (2008). Painful diabetic neuropathy: Treatment and future aspects. Diabetes/
Metabolism Research and Reviews, 24(Suppl. 1), S52–S57.
Ziegler, D., Cicmir, I., Mayer, P., Wiefels, K., & Gries, F. A. (1988). Somatic and autonomic
nerve function during the first year after diagnosis of type 1 (insulin-dependent) diabetes.
Diabetes Research, 7, 123–127.
Ziegler, D., & Gries, F. A. (1997). Alpha-lipoic acid in the treatment of diabetic peripheral
and cardiac autonomic neuropathy. Diabetes, 46(Suppl. 2), S62–S66.
Ziegler, D., Gries, F. A., Spuler, M., & Lessmann, F. (1992). The epidemiology of diabetic
neuropathy. Diabetic cardiovascular autonomic neuropathy multicenter study group.
Journal of Diabetes and Its Complications, 6, 49–57.
Ziegler, D., Papanas, N., Vinik, A. I., & Shaw, J. E. (2014). Epidemiology of polyneuropathy
in diabetes and prediabetes. In D. W. Zochodne & R. A. Malik (Eds.), Diabetes and The
Nervous System: 126 (3rd. series). Handbook of clinical neurology (pp. 3–22): Elsevier.
Ziegler, D., Schatz, H., Conrad, F., Gries, F. A., Ulrich, H., & Reichel, G. (1997). Effects of
treatment with the antioxidant alpha-lipoic acid on cardiac autonomic neuropathy in
NIDDM patients. A 4-month randomized controlled multicenter trial (DEKAN study).
deutsche kardiale autonome neuropathie. Diabetes Care, 20, 369–373.
Alternative Neuropathy Assessments 285
Ziegler, D., Strom, A., Nowotny, B., Zahiragic, L., Nowotny, P. J., Carstensen-Kirberg, M.,
et al. (2015). Effect of Low-energy diets differing in fiber, red meat, and coffee intake on
cardiac autonomic function in obese individuals with type 2 diabetes. Diabetes Care, 38,
1750–1757.
Ziegler, D., Zentai, C. P., Perz, S., Rathmann, W., Haastert, B., Doring, A., et al. (2008).
Prediction of mortality using measures of cardiac autonomic dysfunction in the diabetic
and nondiabetic population: The MONICA/KORA Augsburg Cohort Study. Diabetes
Care, 31, 556–561.
Ziegler, D., Zentai, C., Perz, S., Rathmann, W., Haastert, B., Meisinger, C., et al. (2004).
Diminished heart rate variability (HRV) and prolonged QTc interval, but not increased
QT dispersion (QTD) are predicators of mortality in the diabetic population (Abstract).
Diabetes, 53, A57.
Ziegler, D., Zentai, C., Perz, S., Rathmann, W., Haastert, B., Meisinger, C., et al. (2006).
Selective contribution of diabetes and other cardiovascular risk factors to cardiac auto-
nomic dysfunction in the general population. Experimental and Clinical Endocrinology &
Diabetes, 114, 153–159.
CHAPTER FOURTEEN
Contents
1. The Problem 288
2. What Can We Do? 291
3. Diagnostic Tests are Not Necessarily Good Surrogate End Points 293
4. Clinical Trials in Diabetic Neuropathy 294
4.1 Glycemic Control 295
4.2 Pancreas Transplantation 297
4.3 α-Lipoic Acid 298
4.4 Aldose Reductase Inhibitors 298
4.5 Vitamin B 299
4.6 Angiotensin-Converting Enzyme Inhibitors 299
4.7 Protein Kinase C Activation 300
4.8 C-Peptide 300
4.9 Actovegin 301
4.10 Disease Modification 301
4.11 Nerve Growth Factor 302
4.12 Other Growth Factors 303
4.13 Erythropoietin 303
4.14 Vitamin D 304
4.15 Angiotensin Axis 305
5. Can We Ever Succeed? 306
References 309
Abstract
As of March 2016, we continue to advocate the diagnosis of diabetic neuropathy using a
simple foot examination or monofilament, which identifies only those with severe neu-
ropathy and hence risk of foot ulceration. Given the fact that the 5-year mortality rate of
diabetic patients with foot ulceration is worse than that of most common cancers, surely
we should be identifying patients at an earlier stage of neuropathy to prevent its pro-
gression to a stage with such a high mortality? Of course, we lament that there is no
licensed treatment for diabetic neuropathy. Who is to blame? As researchers and carers,
we have a duty of care to our patients with diabetic neuropathy. So, we have to look
forward not backwards, and move away from our firmly entrenched views on the design
and conduct of clinical trials for diabetic neuropathy. Relevant organizations such as
Neurodiab, the American Diabetes Association and the Peripheral Nerve Society have
to acknowledge that they cannot continue to endorse a bankrupt strategy. The FDA
needs an open and self-critical dialogue with these organizations, to give pharmaceu-
tical companies at least a fighting chance to deliver effective new therapies for diabetic
neuropathy.
I have not failed, I've just found 10,000 ways that won't work
attr. Thomas Edison
1. THE PROBLEM
As the doctors say of a wasting disease, to start with it is easy to cure but difficult to
diagnose; after a time... it becomes easy to diagnose but difficult to cure.
Niccolo Machiavelli
Diabetic neuropathy affects at least 50% of patients with diabetes during their
lifetime. The human and economic burden of diabetic neuropathy and its
consequences in the form of painful neuropathy, foot ulceration, and ampu-
tation are considerable for patients and healthcare systems. This is true in
both in the developed (Abbott, Carrington, Ashe, et al., 2002; Boulton,
Vileikyte, Ragnarson-Tennvall, et al., 2005) and particularly in the devel-
oping world (Riaz et al., 2014) where the consequences of disability are
grave due to loss of employment and hence livelihood—not just for the
patient, but the whole family. The life expectancy of diabetic patients with
a foot ulcer (14.4 years) or Charcot foot (13.9 years) is markedly reduced
compared to a normative U.K. population (van Baal, Hubbard, Game,
et al., 2010). Moreover, by the time, a patient with diabetes has developed
a foot ulcer their 5-year mortality is greater than lymphoma (14%), breast
(10.6%), prostate (10%), and bladder (22%) cancer (Armstrong,
Wrobel, & Robbins, 2007; Marshall, Webb, Hall, et al., 2016) and is much
higher than colon cancer (11%), non-Hodgkin’s (31%), and Hodgkin’s lym-
phoma (14%) (Al-Hamadani, Habermann, Cerhan, et al., 2015). Indeed, it is
comparable to feared cancers such as lung cancer (48%) (Field, Duffy,
Baldwin, et al., 2016; Malvezzi, Carioli, Bertuccio, et al., 2016) (Fig. 1).
Furthermore, according to the World Health Organization and a recent sys-
tematic subnational analysis for the Global Burden of Disease in China, the
three leading causes of death in the world are ischemic heart disease, stroke,
and chronic obstructive pulmonary disease (COPD) (Zhou, Wang, Zhu,
et al., 2016). The 5-year mortality after a myocardial infarct (28%)
(Bata, Gregor, Wolf, et al., 2006), stroke (41.7%) (Hankey, Jamrozik,
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 289
Fig. 1 Five-year mortality of common cancers compared to diabetic patients with foot
ulceration.
Fig. 2 Five-year mortality for the three leading causes of death in the world compared
to patients with diabetic foot ulceration.
The FDA will carry out due process and endorse the low-risk consensus
view. The consequence of this approach has been failure after failure of clin-
ical trials in diabetic neuropathy, so that the field has the unenviable record
of a 100% failure rate. When future trials fail, and they will, investigators will
readily blame “big pharma” for enrolling the wrong types of patients, too
few patients, or choosing an inadequate study duration or the wrong end-
point. And thus the cycle continues.
Fig. 3 Skin biopsies immunostained for neuronal marker PGP 9.5 from a healthy subject
(A) and a patient with severe DPN (B). Note the almost complete depletion of IENFD (red
(gray in the print version) arrows) and only very small nerve fiber profiles (blue (dark gray
in the print version) arrows) scattered in the epidermis and a reduction of the subepi-
dermal nerve plexus (yellow (light gray in the print version) arrows) in (B). Original mag-
nification 200, scale bar ¼ 100 μm.
292 R.A. Malik
Fig. 4 Corneal confocal microscopy images of the sub-basal nerve plexus from a control
(A), a patient without DPN (B), and a patient with DPN (C) showing the reduction in cor-
neal nerves in those with DPN. Red (gray in the print version) arrows indicate main nerve
fibers; yellow (white in the print version) arrows indicate branches.
Johansson, et al., 2010; Lauria, Morbin, Lombardi, et al., 2003) and corneal
confocal microscopy (CCM), which allows noninvasive in vivo imaging of
corneal nerves (Fig. 4) (Malik, Kallinikos, Abbott, et al., 2003). Indeed a
robust body of data, which is conveniently ignored, shows that small fiber
damage occurs in patients with early diabetic neuropathy (Bitirgen,
Ozkagnici, Malik, et al., 2014; Malik, 2014a; Ziegler, Papanas, Zhivov,
et al., 2014) and subjects with IGT (Asghar, Petropoulos, Alam, et al.,
2014; Singleton, Smith, & Bromberg, 2001; Smith, Ramachandran,
Tripp, et al., 2001). Of clinical relevance to the hard end point sought by
the FDA (foot ulceration), an equally strong body of data shows that small
cutaneous fibers release an array of neuropeptides, which are key to the pro-
cess of wound healing (Ashrafi, Baguneid, & Bayat, 2015; Laverdet, Danigo,
Girard, et al., 2015) and hence repair after foot ulceration.
While estimation of IENF density (IENFD) by skin biopsy offers an
objective means of quantifying small fiber pathology, it is an invasive and
costly procedure that does not allow for the same nerves to be visualized
repeatedly (Lauria, Bakkers, et al., 2010; Lauria, Hsieh, et al., 2010;
Lauria, Morbin, Lombardi, et al., 2003). Our previous NIH and JDRF
funded studies have shown that CCM can quantify early small fiber axonal
damage in diabetic neuropathy (Petropoulos, Alam, Fadavi, et al., 2014),
reliably (Petropoulos, Manzoor, Morgan, et al., 2013), with high sensitivity
and specificity (Petropoulos et al., 2014). CCM has a better sensitivity and
comparable specificity for the diagnosis of diabetic neuropathy (Chen,
Graham, Dabbah, et al., 2015) and correlates with IENF loss (Quattrini,
Tavakoli, Jeziorska, et al., 2007). We have also shown that CCM can be
deployed in children with type 1 diabetes, that corneal nerve loss is evident
in young people with T1DM without retinopathy (Szalai, Deak, Modis,
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 293
There have been many clinical trials in subjects with diabetic neuropathy.
As discussed earlier, their failure may derive from many factors ranging
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 295
had no neuropathy at baseline. The apparent success of the DCCT has been
used to highlight the need for large numbers of patients without neuropathy
at baseline who are followed for at least 5 years using a combination of clin-
ical neurological examination, neurophysiology, and autonomic-function
testing in clinical trials of therapies for DPN.
In type 2 diabetes, the evidence of a benefit of improved glycemic con-
trol on neuropathy is less clear—or is it? The United Kingdom Prospective
Diabetes Study (Stratton, Adler, Neil, et al., 2000) reported a significant risk
reduction in amputation for every 1% reduction in mean HbA1c (Stratton
et al., 2000) and a lower rate of impaired VPT with intensive therapy versus
standard therapy. However, this effect only became significant after 15 years,
with no significant advantage observed at 3, 6, 9, and 12 years. What does
this mean for trials that incorporate vibration perception threshold as a pri-
mary or secondary end point over 1–2 years?
Intensive glycemic control in type 2 diabetes has been shown to reduce
microvascular complications in a 6-year randomized, prospective study
(Ohkubo, Kishikawa, Araki, et al., 1995). Does this mean that the minimum
duration of trials should be 6 years? Boussageon, Bejan-Angoulvant,
Saadatian-Elahi, et al. (2011) conducted a meta-analysis of seven trials
involving over 34,000 patients and did not find a reduction in the incidence
of DPN in patients managed through intensive glycemic control. Further-
more, a Cochrane review of 17 randomized trials concluded that tight gly-
cemic control prevented neuropathy in type 1 diabetes, driven principally by
the DCCT findings, but that it had no benefit in type 2 diabetes (Callaghan,
Little, et al., 2012). It should be noted that these meta-analyses have signif-
icant limitations as the targeted glucose levels, therapeutic strategies, out-
come measures, trial design, and duration of follow-up differ between
studies. The principal culprit for demonstrating a lack of benefit is, however,
that the majority of these studies utilized crude neuropathy end points such
as a monofilament and foot exam and vibration perception.
The VA Cooperative study of type 2 diabetes reported no difference in
the prevalence of autonomic neuropathy between the intensive and standard
therapy arms at 2 years (Duckworth et al., 2009). In contrast, the Steno-2
Trial reported that an intervention that integrated glucose control and
multiple cardiovascular risk factor management reduced the prevalence of
cardiac autonomic neuropathy but not unsurprisingly showed no benefit
for somatic neuropathy, as assessed using vibration perception thresholds
in patients with type 2 diabetes and microalbuminuria even after 7.8 years
of follow-up (Gaede, Lund-Andersen, Parving, et al., 2008).
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 297
4.5 Vitamin B
Benfotiamine is a fat-soluble derivative of thiamine that has been shown in
animal models to inhibit three major pathways implicated in oxidative stress
and vascular dysfunction in diabetes: the advanced glycation end-product
pathway, the hexosamine pathway, and the protein kinase C (PKC)–
diacylglycerol pathway (Hammes, Du, Edelstein, et al., 2003). Although
the multimodal effects of benfotiamine present an attractive option for
treating DPN, clinical trials are unclear about its efficacy. A randomized
placebo-controlled trial (Stracke, Gaus, Achenbach, et al., 2008) of 165
patients reported an improvement in patient-reported symptoms in the
per-protocol arm of the study, although there was no improvement in
the intent-to-treat group compared to placebo. Furthermore there was an
improvement in symptoms but no change in deficits between the placebo
and treatment arms of the study (Stracke et al., 2008). In a multicenter, ran-
domized, double-blind, placebo-controlled trial of 214 patients with type 2
diabetes randomly assigned to 24 weeks of treatment with either L-meth-
ylfolate calcium 3 mg, methylcobalamin 2 mg, and pyridoxal-50 -phosphate
35 mg, or placebo the primary end point, not unsurprisingly, VPT did not
change, while the secondary end points of the Neuropathy Total Symptom
Score (NTSS-6) and Short Form 36 (SF-36), did (Fonseca, Lavery, Thethi,
et al., 2013). In a recent “real-world” study, 544 patients reported a mean
reduction of 35% in NTSS-6 scores after 12 weeks on a medical food product
containing L-methylfolate-methylcobalamin-pyridoxal-5-phosphate (Trippe,
Barrentine, Curole, et al., 2016).
4.8 C-Peptide
C-peptide deficiency is an important contributing factor to the characteristic
functional and structural abnormalities of peripheral nerves (Sima, 2003).
C-peptide binds to cell membranes, resulting in stimulation of endothelial
nitric oxide synthase (eNOS) and Na+, K+-ATPase (Wahren, Ekberg, &
Jornvall, 2007). In the Joslin 50-Year Medallist Study (Sun, Keenan,
Cavallerano, et al., 2011), protection from complications (retinopathy,
nephropathy, and neuropathy) was thought to be due to the presence of
enriched protective factors against microvascular complications. In two
double-blind, placebo-controlled studies in type 1 DM patients, C-peptide
replacement or placebo was given with the patients’ regular insulin therapy
(Ekberg, Brismar, Johansson, et al., 2003, 2007). Sensory NCV assessed in
the sural nerve showed a significant improvement. A recent large phase III
trial has reported no significant improvement in nerve conduction param-
eters compared to placebo, although it should be noted that the placebo arm
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 301
4.9 Actovegin
Actovegin is a hemoderivative produced from calf blood by ultrafiltration
that exerts insulin-like activity, stimulating glucose transport, pyruvate
dehydrogenase, and glucose oxidation (Ziegler, Movsesyan, Mankovsky,
et al., 2009). In a multicenter randomized placebo-controlled double-blind
trial, 567 patients with type 2 diabetes received 20 intravenous infusions of
Actovegin (2000 mg day1) (n ¼ 281) or placebo (n ¼ 286) once daily
followed by three tablets of Actovegin (1800 mg day1) or placebo three
times daily for 140 days. TSS (P ¼ 0.0003), VPT (P ¼ 0.017), and NIS-LL
sensory function (P ¼ 0.021) improved significantly (Ziegler et al., 2009).
However, this is the only trial of Actovegin and comprehensive clinical trials
are needed to confirm its benefits in the treatment of DPN (Boulton
et al., 2013).
4.13 Erythropoietin
Erythropoietin (EPO) is produced in situ by cells under stress and has
been found to antagonize the production of proinflammatory molecules
304 R.A. Malik
and promote tissue healing. While EPO has been found to ameliorate
experimental DPN, its use is limited by serious adverse effects, in partic-
ular increased thrombotic risk (Brines et al., 2014). ARA 290, a
nonhematopoietic peptide designed from the structure of EPO, selectively
interacts with the EPO receptors that mediate tissue protection. In a recent
phase II trial to evaluate the activity of ARA 290 in type 2 diabetes and
DPN, ARA 290 (4 mg) or placebo were self-administered subcutaneously
daily for 28 days and the subjects followed for an additional month without
further treatment. Neuropathic symptoms were found to improve signifi-
cantly in the ARA 290 group. Additionally, a significant improvement in
corneal nerve morphology was found using CCM in patients randomized
to the ARA 290 arm of the trial relative to placebo. ARA 290 remains in
development for DPN and type 2 diabetes (Brines et al., 2014).
4.14 Vitamin D
There is an association between vitamin D deficiency and pain in the gen-
eral population (Hirani, 2012). A mechanistic link between vitamin D and
pain has been supported by a recent study reporting that nociceptive cal-
citonin gene-related peptide (CGRP)-positive neurons have a distinct
vitamin D phenotype with hormonally regulated ligand and receptor levels
(Tague & Smith, 2011). Vitamin D deficiency results in increased numbers
of axons containing CGRP. In cultured neurons, vitamin D receptor
(VDR) expression is increased in growth cones and sprouting appears
to be regulated by VDR-mediated rapid response signaling pathways
(Tague, Clarke, Winter, et al., 2011). NGF is known to be depleted in
experimental diabetes (Hellweg, Wohrle, Hartung, et al., 1991) and in
a study of patients with diabetic neuropathy, NGF immunostaining on
skin keratinocytes correlated with skin axon–reflex vasodilation, a measure
of small fiber neuropathy (Anand, Terenghi, Warner, et al., 1996). In an
experimental study, NGF expression was maintained in sciatic nerves of
diabetic animals treated with a vitamin D analog (CB1093). Similarly
Tacalcitol, active vitamin D3, induces NGF production in human epider-
mal keratinocytes (Fukuoka, Sakurai, Ohta, et al., 2001). Treatment with
vitamin D3 has been shown to reduce demyelination in a cuprizone
experimental model of demyelination (Wergeland, Torkildsen, Myhr,
et al., 2011) and in a separate spinal cord compression model it has been
shown to induce axonal regeneration (Bianco, Gueye, Marqueste,
et al., 2011).
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 305
will be positive for a drug that is supposed to repair nerves, or at the very least
limit further degeneration. Trials are also constrained by current FDA
guidance, which has not changed for 20 years, and continues to endorse
symptoms and signs, QST, and neurophysiology. In my opinion, future
clinical trials in patients with diabetic neuropathy must minimally:
• Enrol patients with mild or moderate neuropathy.
• Account for the progression rate of neuropathy in the placebo cohort.
• Use surrogate end points of small fiber repair, where pertinent to the
therapeutic approach.
• Operate in a regulatory environment that accepts small fiber repair as a
desirable primary end point.
We can continue to go from failure to failure, or we can have the courage to
question how we diagnose and conduct clinical trials in diabetic neuropathy
in order to make a difference for future generations of patients with diabetic
neuropathy. Many potentially efficacious therapies may well have been
aborted prematurely and need to be reconsidered using the correct end
points.
REFERENCES
Abbott, C. A., Carrington, A. L., Ashe, H., et al. (2002). The north-west diabetes foot care
study: Incidence of, and risk factors for, new diabetic foot ulceration in a community-
based patient cohort. Diabetic Medicine, 19, 377–384.
Abbott, C. A., Malik, R. A., van Ross, E. R., et al. (2011). Prevalence and characteristics of
painful diabetic neuropathy in a large community-based diabetic population in the U.K.
Diabetes Care, 34, 2220–2224.
Agudo, R., Valls-Solé, J., Recasens, M., et al. (2002). Effects of kidney and pancreas trans-
plantation on neurophysiological rates of polyneuropathy and autonomous cardiac func-
tion in diabetic patients with end-stage renal disease. Medicina Clı´nica (Barcelona), 118,
534–538.
Ajroud-Driss, S., Christiansen, M., Allen, J. A., et al. (2013). Phase 1/2 open-label dose-
escalation study of plasmid DNA expressing two isoforms of hepatocyte growth factor
in patients with painful diabetic peripheral neuropathy. Molecular Therapy, 21,
1279–1286.
Alamdari, A., Mozafari, R., Tafakhori, A., et al. (2015). An inverse association between
serum vitamin D levels with the presence and severity of impaired nerve conduction
velocity and large fiber peripheral neuropathy in diabetic subjects. Neurological Sciences,
36, 1121–1126.
Al-Hamadani, M., Habermann, T. M., Cerhan, J. R., et al. (2015). Non-Hodgkin lym-
phoma subtype distribution, geodemographic patterns, and survival in the US:
A longitudinal analysis of the National Cancer Data Base from 1998 to 2011. American
Journal of Hematology, 90, 790–795.
Almurdhi, M. M., Reeves, N. D., Bowling, F. L., et al. (2016). Reduced lower-limb muscle
strength and volume in patients with type 2 diabetes in relation to neuropathy, intramus-
cular fat, and vitamin D levels. Diabetes Care, 39, 441–447.
Anand, P., Terenghi, G., Warner, G., et al. (1996). The role of endogenous nerve growth
factor in human diabetic neuropathy. Nature Medicine, 2, 703–707.
310 R.A. Malik
Brines, M., Dunne, A. N., van Velzen, M., et al. (2014). ARA 290, a nonerythropoietic
peptide engineered from erythropoietin, improves metabolic control and neuropathic
symptoms in patients with type 2 diabetes. Molecular Medicine, 20, 658.
Brooks, B., Delaney-Robinson, C., Molyneaux, L., et al. (2008). Endothelial and neural reg-
ulation of skin microvascular blood flow in patients with diabetic peripheral neuropathy:
Effect of treatment with the isoform-specific protein kinase C beta inhibitor,
ruboxistaurin. Journal of Diabetes and its Complications, 22, 88–95.
Brown, M. T., Herrmann, D. N., Goldstein, M., et al. (2014). Nerve safety of tanezumab, a
nerve growth factor inhibitor for pain treatment. Journal of the Neurological Sciences, 345,
139–147.
Callaghan, B. C., Cheng, H. T., Stables, C. L., et al. (2012). Diabetic neuropathy: Clinical
manifestations and current treatments. Lancet Neurology, 11, 521–534.
Callaghan, B. C., Little, A. A., Feldman, E. L., et al. (2012). Enhanced glucose control for
preventing and treating diabetic neuropathy. Cochrane Database of Systematic Reviews, 6,
CD007543.
Calles-Escandon, J., Lovato, L. C., Simons-Morton, D. G., et al. (2010). Effect of intensive
compared with standard glycemia treatment strategies on mortality by baseline subgroup
characteristics: The Action to Control Cardiovascular Risk in Diabetes (ACCORD)
trial. Diabetes Care, 33, 721–727.
Celikbilek, A., Gocmen, A. Y., Tanik, N., et al. (2015). Decreased serum vitamin D levels are
associated with diabetic peripheral neuropathy in a rural area of Turkey. Acta Neurologica
Belgica, 115, 47–52.
Chalk, C., Benstead, T. J., & Moore, F. (2007). Aldose reductase inhibitors for the treatment
of diabetic polyneuropathy. Cochrane Database of Systematic Reviews, CD004572.
Chen, X., Graham, J., Dabbah, M. A., et al. (2015). Small nerve fiber quantification in
the diagnosis of diabetic sensorimotor polyneuropathy: Comparing corneal confocal
microscopy with intraepidermal nerve fiber density. Diabetes Care, 38, 1138–1144.
Dabbah, M., Graham, J., Petropoulos, I., et al. (2011). Automatic analysis of diabetic periph-
eral neuropathy using multi-scale quantitative morphology of nerve fibres in corneal
confocal microscopy imaging. Medical Image Analysis, 15, 738–747.
Davis, T. M., Yeap, B. B., Davis, W. A., et al. (2008). Lipid-lowering therapy and peripheral
sensory neuropathy in type 2 diabetes: The Fremantle Diabetes Study. Diabetologia, 51,
562–566.
Dehghani, C., Pritchard, N., Edwards, K., et al. (2016). Risk factors associated with corneal
nerve alteration in type 1 diabetes in the absence of neuropathy: A longitudinal In Vivo
corneal confocal microscopy study. Cornea, (in press).
Del Carro, U., Fiorina, P., Amadio, S., et al. (2007). Evaluation of polyneuropathy markers in
type 1 diabetic kidney transplant patients and effects of islet transplantation: Neurophys-
iological and skin biopsy longitudinal analysis. Diabetes Care, 30, 3063–3069.
Drury, P. L., Ting, R., Zannino, D., et al. (2011). Estimated glomerular filtration rate and
albuminuria are independent predictors of cardiovascular events and death in type 2 dia-
betes mellitus: The Fenofibrate Intervention and Event Lowering in Diabetes (FIELD)
study. Diabetologia, 54, 32–43.
Duckworth, W., Abraira, C., Moritz, T., et al. (2009). Glucose control and vascular
complications in veterans with type 2 diabetes. The New England Journal of Medicine,
360, 129–139.
Dyck, P. J. (2014). Methodology for conduct of epidemiologic surveys and randomized
controlled trials of diabetic polyneuropathy. Handbook of Clinical Neurology, 126,
335–338.
Dyck, P. J., Albers, J. W., Wolfe, J., et al. (2013). A trial of proficiency of nerve conduction:
Greater standardization still needed. Muscle & Nerve, 48, 369–374.
Dyck, P. J., Argyros, B., Russell, J. W., et al. (2014). Multicenter trial of the proficiency of
smart quantitative sensation tests. Muscle & Nerve, 49, 645–653.
312 R.A. Malik
Dyck, P. J., Norell, J. E., Tritschler, H., et al. (2007). Challenges in design of multicenter
trials: End points assessed longitudinally for change and monotonicity. Diabetes Care,
30, 2619–2625.
Dyck, P. J., Overland, C. J., Low, P. A., et al. (2010). Signs and symptoms versus nerve
conduction studies to diagnose diabetic sensorimotor polyneuropathy: Cl vs. NPhys trial.
Muscle & Nerve, 42, 157–164.
Dyck, P. J., Zimmerman, B. R., Vilen, T. H., et al. (1988). Nerve glucose, fructose, sorbitol,
myo-inositol, and fiber degeneration and regeneration in diabetic neuropathy. The New
England Journal of Medicine, 319, 542–548.
Ekberg, K., Brismar, T., Johansson, B. L., et al. (2003). Amelioration of sensory nerve
dysfunction by C-peptide in patients with type 1 diabetes. Diabetes, 52, 536–541.
Ekberg, K., Brismar, T., Johansson, B. L., et al. (2007). C-Peptide replacement therapy and
sensory nerve function in type 1 diabetic neuropathy. Diabetes Care, 30, 71–76.
Field, J. K., Duffy, S. W., Baldwin, D. R., et al. (2016). UK Lung Cancer RCT Pilot Screen-
ing Trial: Baseline findings from the screening arm provide evidence for the potential
implementation of lung cancer screening. Thorax, 71, 161–170.
Fioretto, P., Steffes, M. W., Sutherland, D. E., et al. (1998). Reversal of lesions of diabetic
nephropathy after pancreas transplantation. The New England Journal of Medicine, 339,
69–75.
Fiorina, P., & Secchi, A. (2007). Pancreatic islet cell transplant for treatment of diabetes.
Endocrinology and Metabolism Clinics of North America, 36, 999–1013. ix.
Fonseca, V. A., Lavery, L. A., Thethi, T. K., et al. (2013). Metanx in type 2 diabetes with
peripheral neuropathy: A randomized trial. The American Journal of Medicine, 126,
141–149.
Fried, L. F., Forrest, K. Y., Ellis, D., et al. (2001). Lipid modulation in insulin-dependent
diabetes mellitus: Effect on microvascular outcomes. Journal of Diabetes and its Complica-
tions, 15, 113–119.
Fukuoka, M., Sakurai, K., Ohta, T., et al. (2001). Tacalcitol, an active vitamin D3, induces
nerve growth factor production in human epidermal keratinocytes. Skin Pharmacology and
Applied Skin Physiology, 14, 226–233.
Gaede, P., Lund-Andersen, H., Parving, H. H., et al. (2008). Effect of a multifactorial inter-
vention on mortality in type 2 diabetes. The New England Journal of Medicine, 358, 580–591.
Goto, Y., Hotta, N., Shigeta, Y., et al. (1995). Effects of an aldose reductase inhibitor,
epalrestat, on diabetic neuropathy. Clinical benefit and indication for the drug assessed
from the results of a placebo-controlled double-blind study. Biomedicine & Pharmacother-
apy, 49, 269–277.
Greene, D. A., Arezzo, J. C., & Brown, M. B. (1999). Effect of aldose reductase inhibition on
nerve conduction and morphometry in diabetic neuropathy. Zenarestat Study Group.
Neurology, 53, 580–591.
Hammes, H. P., Du, X., Edelstein, D., et al. (2003). Benfotiamine blocks three major path-
ways of hyperglycemic damage and prevents experimental diabetic retinopathy. Nature
Medicine, 9, 294–299.
Hankey, G. J., Jamrozik, K., Broadhurst, R. J., et al. (2000). Five-year survival after first-ever
stroke and related prognostic factors in the Perth Community Stroke Study. Stroke, 31,
2080–2086.
Hellweg, R., Wohrle, M., Hartung, H. D., et al. (1991). Diabetes mellitus-associated
decrease in nerve growth factor levels is reversed by allogeneic pancreatic islet transplan-
tation. Neuroscience Letters, 125, 1–4.
Herman, W. H., Aubert, R. E., Engelgau, M. M., et al. (1998). Diabetes mellitus in Egypt:
Glycaemic control and microvascular and neuropathic complications. Diabetic Medicine,
15, 1045–1051.
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 313
Malik, R. A., Kallinikos, P., Abbott, C. A., et al. (2003). Corneal confocal microscopy:
A non-invasive surrogate of nerve fibre damage and repair in diabetic patients.
Diabetologia, 46, 683–688.
Malik, R. A., Williamson, S., Abbott, C., et al. (1998). Effect of angiotensin-converting-
enzyme (ACE) inhibitor trandolapril on human diabetic neuropathy: Randomised
double-blind controlled trial. Lancet, 352, 1978–1981.
Malvezzi, M., Carioli, G., Bertuccio, P., et al. (2016). European cancer mortality predictions
for the year 2016 with focus on leukaemias. Annals of Oncology.
Marshall, D. C., Webb, T. E., Hall, R. A., et al. (2016). Trends in UK regional cancer
mortality 1991–2007. British Journal of Cancer, 114, 340–347.
Martinez, C. H., Mannino, D. M., Jaimes, F. A., et al. (2015). Undiagnosed obstructive lung
disease in the United States. Associated factors and long-term mortality. Annals of the
American Thoracic Society, 12, 1788–1795.
Maser, R. E., Lenhard, M. J., & Pohlig, R. T. (2015). Vitamin D insufficiency is associated
with reduced parasympathetic nerve fiber function in type 2 diabetes. Endocrine Practice,
21, 174–181.
Mayfield, J. A., & Sugarman, J. R. (2000). The use of the Semmes-Weinstein monofilament
and other threshold tests for preventing foot ulceration and amputation in persons with
diabetes. The Journal of Family Practice, 49, S17–S29.
Mehra, S., Tavakoli, M., Kallinikos, P. A., et al. (2007). Corneal confocal microscopy detects
early nerve regeneration after pancreas transplantation in patients with type 1 diabetes.
Diabetes Care, 30, 2608–2612.
Navarro, X., Sutherland, D. E., & Kennedy, W. R. (1997). Long-term effects of pancreatic
transplantation on diabetic neuropathy. Annals of Neurology, 42, 727–736.
Ohkubo, Y., Kishikawa, H., Araki, E., et al. (1995). Intensive insulin therapy prevents
the progression of diabetic microvascular complications in Japanese patients with
non-insulin-dependent diabetes mellitus: A randomized prospective 6-year study.
Diabetes Research and Clinical Practice, 28, 103–117.
Oyibo, S. O., Prasad, Y. D., Jackson, N. J., et al. (2002). The relationship between blood
glucose excursions and painful diabetic peripheral neuropathy: A pilot study. Diabetic
Medicine, 19, 870–873.
Petropoulos, I. N., Alam, U., Fadavi, H., et al. (2014). Rapid automated diagnosis of diabetic
peripheral neuropathy with in vivo corneal confocal microscopy. Investigative Ophthal-
mology & Visual Science, 55, 2071–2078.
Petropoulos, I. N., Manzoor, T., Morgan, P., et al. (2013). Repeatability of in vivo corneal
confocal microscopy to quantify corneal nerve morphology. Cornea, 32, e83–e89.
Pfeifer, M. A., & Schumer, M. P. (1995). Clinical trials of diabetic neuropathy: Past, present,
and future. Diabetes, 44, 1355–1361.
Pop-Busui, R., Stevens, M. J., Raffel, D. M., et al. (2013). Effects of triple antioxidant ther-
apy on measures of cardiovascular autonomic neuropathy and on myocardial blood flow
in type 1 diabetes: A randomised controlled trial. Diabetologia, 56, 1835–1844.
Pritchard, N., Edwards, K., Russell, A. W., et al. (2015). Corneal confocal microscopy pre-
dicts 4-year incident peripheral neuropathy in type 1 diabetes. Diabetes Care, 38,
671–675.
Quattrini, C., Tavakoli, M., Jeziorska, M., et al. (2007). Surrogate markers of small fiber
damage in human diabetic neuropathy. Diabetes, 56, 2148–2154.
Rajamani, K., Colman, P. G., Li, L. P., et al. (2009). Effect of fenofibrate on amputation
events in people with type 2 diabetes mellitus (FIELD study): A prespecified analysis
of a randomised controlled trial. Lancet, 373, 1780–1788.
Reichard, P., Nilsson, B. Y., & Rosenqvist, U. (1993). The effect of long-term intensified
insulin treatment on the development of microvascular complications of diabetes
mellitus. The New England Journal of Medicine, 329, 304–309.
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 315
Reja, A., Tesfaye, S., Harris, N. D., et al. (1995). Is ACE inhibition with lisinopril helpful in
diabetic neuropathy? Diabetic Medicine, 12, 307–309.
Riaz, M., Miyan, Z., Zaidi, S. I., Alvi, S. F., Fawwad, A., Ahmadani, M. Y., et al. (2014).
Characteristics of a large cohort of patients with diabetes having at-risk feet and outcomes
in patients with foot ulceration referred to a tertiary care diabetes unit. International
Wound Journal. http://dx.doi.org/10.1111/iwj.12289[Epub ahead of print].
Rice, A. S., Dworkin, R. H., McCarthy, T. D., et al. (2014). EMA401, an orally adminis-
tered highly selective angiotensin II type 2 receptor antagonist, as a novel treatment for
postherpetic neuralgia: A randomised, double-blind, placebo-controlled phase 2 clinical
trial. Lancet, 383, 1637–1647.
Ropper, A. H., Gorson, K. C., Gooch, C. L., et al. (2009). Vascular endothelial growth factor
gene transfer for diabetic polyneuropathy: A randomized, double-blinded trial. Annals of
Neurology, 65, 386–393.
Ruggenenti, P., Lauria, G., Iliev, I. P., et al. (2011). Effects of manidipine and delapril in
hypertensive patients with type 2 diabetes mellitus: The delapril and manidipine for
nephroprotection in diabetes (DEMAND) randomized clinical trial. Hypertension, 58,
776–783.
Shehab, D., Al-Jarallah, K., Abdella, N., et al. (2015). Prospective evaluation of the effect of
short-term oral vitamin d supplementation on peripheral neuropathy in type 2 diabetes
mellitus. Medical Principles and Practice, 24, 250–256.
Shehab, D., Al-Jarallah, K., Mojiminiyi, O. A., et al. (2012). Does Vitamin D deficiency play
a role in peripheral neuropathy in Type 2 diabetes? Diabetic Medicine, 29, 43–49.
Shelton, D. (2014). Development of nerve growth factor (NGF) inhibition as a strategy for
treatment of pain. Journal of the Peripheral Nervous System, 19(Suppl. 2), S12–S13.
Sima, A. A. (2003). New insights into the metabolic and molecular basis for diabetic neurop-
athy. Cellular and Molecular Life Sciences, 60, 2445–2464.
Sima, A. A., Bril, V., Nathaniel, V., et al. (1988). Regeneration and repair of myelinated
fibers in sural-nerve biopsy specimens from patients with diabetic neuropathy treated
with sorbinil. The New England Journal of Medicine, 319, 548–555.
Singleton, J. R., Smith, A. G., & Bromberg, M. B. (2001). Increased prevalence of impaired
glucose tolerance in patients with painful sensory neuropathy. Diabetes Care, 24,
1448–1453.
Smith, A. G., Ramachandran, P., Tripp, S., et al. (2001). Epidermal nerve innervation in
impaired glucose tolerance and diabetes-associated neuropathy. Neurology, 57, 1701–1704.
Soderstrom, L. H., Johnson, S. P., Diaz, V. A., et al. (2012). Association between vitamin
D and diabetic neuropathy in a nationally representative sample: Results from
2001–2004 NHANES. Diabetic Medicine, 29, 50–55.
Stracke, H., Gaus, W., Achenbach, U., et al. (2008). Benfotiamine in diabetic poly-
neuropathy (BENDIP): Results of a randomised, double blind, placebo-controlled
clinical study. Experimental and Clinical Endocrinology & Diabetes, 116, 600–605.
Stratton, I. M., Adler, A. I., Neil, H. A., et al. (2000). Association of glycaemia with
macrovascular and microvascular complications of type 2 diabetes (UKPDS 35):
Prospective observational study. BMJ, 321, 405–412.
Sun, J. K., Keenan, H. A., Cavallerano, J. D., et al. (2011). Protection from retinopathy and
other complications in patients with type 1 diabetes of extreme duration: The Joslin
50-Year medalist study. Diabetes Care, 34, 968–974.
Szalai, E., Deak, E., Modis, L., Jr., et al. (2016). Early corneal cellular and nerve fiber pathol-
ogy in young patients with type 1 diabetes mellitus identified using corneal confocal
microscopy. Investigative Ophthalmology & Visual Science, 57, 853–858.
Tague, S. E., Clarke, G. L., Winter, M. K., et al. (2011). Vitamin D deficiency promotes
skeletal muscle hypersensitivity and sensory hyperinnervation. The Journal of Neuroscience,
31, 13728–13738.
316 R.A. Malik
Tague, S. E., & Smith, P. G. (2011). Vitamin D receptor and enzyme expression in dorsal
root ganglia of adult female rats: Modulation by ovarian hormones. Journal of Chemical
Neuroanatomy, 41, 1–12.
Tavakoli, M., Ferdousi, M., Petropoulos, I. N., et al. (2015). Normative values for corneal
nerve morphology assessed using corneal confocal microscopy: A multinational norma-
tive data set. Diabetes Care, 38, 838–843.
Tavakoli, A., & Liong, S. (2012). Pancreatic transplant in diabetes. Advances in Experimental
Medicine and Biology, 771, 420–437.
Tavakoli, M., Mitu-Pretorian, M., Petropoulos, I. N., et al. (2013). Corneal confocal micros-
copy detects early nerve regeneration in diabetic neuropathy after simultaneous pancreas
and kidney transplantation. Diabetes, 62, 254–260.
Tesfaye, S., Boulton, A. J., Dyck, P. J., et al. (2010). Diabetic neuropathies: Update on def-
initions, diagnostic criteria, estimation of severity, and treatments. Diabetes Care, 33,
2285–2293.
Tesfaye, S., Chaturvedi, N., Eaton, S. E. M., et al. (2005). Vascular risk factors and diabetic
neuropathy. New England Journal of Medicine, 352, 341–350.
Tesfaye, S., Tandan, R., Bastyr, E. J., 3rd., et al. (2007). Factors that impact symptomatic
diabetic peripheral neuropathy in placebo-administered patients from two 1-year clinical
trials. Diabetes Care, 30, 2626–2632.
The Diabetes Control and Complications Trial Research Group. (1993). The effect of
intensive treatment of diabetes on the development and progression of long-term com-
plications in insulin-dependent diabetes mellitus. The New England Journal of Medicine,
329, 977–986.
The Diabetes Control and Complications Trial Research Group. (1995). The effect of
intensive diabetes therapy on the development and progression of neuropathy. Annals
of Internal Medicine, 122, 561–568.
Trippe, B. S., Barrentine, L. W., Curole, M. V., et al. (2016). Nutritional management of
patients with diabetic peripheral neuropathy with L-methylfolate-methylcobalamin-
pyridoxal-5-phosphate: Results of a real-world patient experience trial. Current Medical
Research and Opinion, 32, 219–227.
van Baal, J., Hubbard, R., Game, F., et al. (2010). Mortality associated with acute Charcot
foot and neuropathic foot ulceration. Diabetes Care, 33, 1086–1089.
van Hecke, M. V., Dekker, J. M., Stehouwer, C. D., et al. (2005). Diabetic retinopathy
is associated with mortality and cardiovascular disease incidence: The EURODIAB
prospective complications study. Diabetes Care, 28, 1383–1389.
van Velzen, M., Heij, L., Niesters, M., et al. (2014). ARA 290 for treatment of small fiber
neuropathy in sarcoidosis. Expert Opinion on Investigational Drugs, 23, 541–550.
Verheyen, A., Peeraer, E., Lambrechts, D., et al. (2013). Therapeutic potential of VEGF and
VEGF-derived peptide in peripheral neuropathies. Neuroscience, 244, 77–89.
Vinik, A. I., Bril, V., Kempler, P., et al. (2005). Treatment of symptomatic diabetic periph-
eral neuropathy with the protein kinase C beta-inhibitor ruboxistaurin mesylate during a
1-year, randomized, placebo-controlled, double-blind clinical trial. Clinical Therapeutics,
27, 1164–1180.
Wahren, J., Ekberg, K., & Jornvall, H. (2007). C-peptide is a bioactive peptide. Diabetologia,
50, 503–509.
Wahren, J., Foyt, H., Daniels, M., et al. (2016). Long-acting C-peptide and neuropathy in
Type 1 diabetes: A 12-month clinical trial. Diabetes Care, (in press).
Walsh, J. W., Hoffstad, O. J., Sullivan, M. O., & Margolis, D. J. (2015). Association of dia-
betic foot ulcer and death in a population-based cohort from the United Kingdom. Dia-
betic Medicine. http://dx.doi.org/10.1111/dme.13054.
Wergeland, S., Torkildsen, O., Myhr, K. M., et al. (2011). Dietary vitamin D3 supplements
reduce demyelination in the cuprizone model. PloS One, 6, e26262.
Wherefore Art Thou, O Treatment for Diabetic Neuropathy? 317
Wiggin, T. D., Sullivan, K. A., Pop-Busui, R., et al. (2009). Elevated triglycerides correlate
with progression of diabetic neuropathy. Diabetes, 58, 1634–1640.
Zhou, M., Wang, H., Zhu, J., et al. (2016). Cause-specific mortality for 240 causes in China
during 1990–2013: A systematic subnational analysis for the Global Burden of Disease
Study 2013. Lancet, 387, 251–272.
Ziegler, D., & Luft, D. (2002). Clinical trials for drugs against diabetic neuropathy: Can we
combine scientific needs with clinical practicalities? International Review of Neurobiology,
50, 431–463.
Ziegler, D., Movsesyan, L., Mankovsky, B., et al. (2009). Treatment of symptomatic
polyneuropathy with actovegin in type 2 diabetic patients. Diabetes Care, 32, 1479–1484.
Ziegler, D., Nowak, H., Kempler, P., et al. (2004). Treatment of symptomatic diabetic
polyneuropathy with the antioxidant alpha-lipoic acid: A meta-analysis. Diabetic Medicine,
21, 114–121.
Ziegler, D., Papanas, N., Zhivov, A., et al. (2014). Early detection of nerve fiber loss by
corneal confocal microscopy and skin biopsy in recently diagnosed type 2 diabetes.
Diabetes, 63, 2454–2463.
INDEX
Note: Page numbers followed by “f ” indicate figures, and “t” indicate tables.
A B
ACE inhibitors, 299–300 BioBreeding/Worcester rat, 102
Actovegin, 301 BioBreeding Zucker diabetic rat
Advanced glycation endproducts (AGEs) (BBZDR)/Wor rat, 96
clinical application, antiglycation agents,
134–135 C
DPN, 132–134, 133f Calcitonin gene-related peptide (CGRP)-
glycation of proteins, 131–132 positive neurons, 304
sensory neurodegeneration, diabetes, Campenot Chambers, 76–77
163–164 Cardiovascular autonomic neuropathy
transgenic and knockout mice studies, (CAN). See also Neuropathy
134 DAN
AGE–RAGE signaling, 164 diabetes management, 251
Aldose reductase inhibition (ARI) diagnosis, 248
DPN, 115–116 exercise intolerance, 248
growth factor, 298–299 heart rate variation (HRV), 247
NCV, 4 increased risk, mortality, 250
Aldose reductase (AR) pathway. intraoperative cardiovascular liability,
See Polyol (aldose reductase 248–249
(AR)/sorbitol–fructose) pathway left ventricular diastolic dysfunction
α-lipoic acid, 298 (LVDD), 247–248
Altered bioenergetics and oxidative stress, myocardial infarction (MI), increased
70–72 mortality due to, 250
Altered insulin signaling orthostatic hypotension, 249
Alzheimer’s disease, 159 resting tachycardia, 248
dermal and epidermal axons, 160 silent myocardial ischemia/cardiac
DRG, 159 denervation syndrome, 249–250
sensory neurons, receptors, 156–157, 158f sudden death, 250
type 1 DM, 157–159 diagnostic tests, 261–262, 263t
type 2 DM, 157–159 Caspase-3 expression, 162–163
Angiotensin II type 2 receptor (AT2R), 305 CaV3.2 T-type channels, 217
Apoptosis. See Cell death Cell death, 69–70
Arabidopsis thaliana miR171, 168 C-peptide, 160–161, 300–301
Axonal degeneration and regeneration
“die-back” neuropathies, 73–74 D
4-HNE, 75–76 Diabetic autonomic neuropathy (DAN)
immortalized cell lines, 74–75 CAN
paclitaxel-induced axonal degeneration, diabetes management, 251
73–74, 74f diagnosis, 248
regenerative responses, 75 exercise intolerance, 248
reinnervation, distal target tissues, 73 heart rate variation (HRV), 247
unsupportive extracellular environment, 76 increased risk, mortality, 250
319
320 Index
329
330 Contents of Recent Volumes
Free Radicals, Calcium, and the Synaptic Vesicle Recycling at the Drosophila Neuromuscu-
Plasticity-Cell Death Continuum: Emerging lar Junction
Roles of the Trascription Factor NFκB Daniel T. Stimson and Mani Ramaswami
Mark P. Mattson
Ionic Currents in Larval Muscles of Drosophila
AP-I Transcription Factors: Short- and Long- Satpal Singh and Chun-Fang Wu
Term Modulators of Gene Expression in the Brain
Development of the Adult Neuromuscular
Keith Pennypacker
System
Ion Channels in Epilepsy Joyce J. Fernandes and Haig Keshishian
Istvan Mody
Controlling the Motor Neuron
Posttranslational Regulation of Ionotropic Gluta- James R. Trimarchi, Ping Jin, and Rodney
mate Receptors and Synaptic Plasticity K. Murphey
Xiaoning Bi, Steve Standley, and Michel Baudry
Heritable Mutations in the Glycine, GABAA, and
Nicotinic Acetylcholine Receptors Provide New
Insights into the Ligand-Gated Ion Channel
Volume 44
Receptor Superfamily Human Ego-Motion Perception
Behnaz Vafa and Peter R. Schofield A. V. van den Berg
INDEX Optic Flow and Eye Movements
M. Lappe and K.-P. Hoffman
The Role of MST Neurons during Ocular Track-
Volume 43 ing in 3D Space
K. Kawano, U. Inoue, A. Takemura, Y. Kodaka,
Early Development of the Drosophila Neuromus-
and F. A. Miles
cular Junction: A Model for Studying Neuronal
Networks in Development Visual Navigation in Flying Insects
Akira Chiba M. V. Srinivasan and S.-W. Zhang
Development of Larval Body Wall Muscles Neuronal Matched Filters for Optic Flow
Michael Bate, Matthias Landgraf, and Mar Ruiz Processing in Flying Insects
Gómez Bate H. G. Krapp
Development of Electrical Properties and Synaptic A Common Frame of Reference for the Analysis
Transmission at the Embryonic Neuromuscular of Optic Flow and Vestibular Information
Junction B. J. Frost and D. R. W. Wylie
Kendal S. Broadie
Optic Flow and the Visual Guidance of
Ultrastructural Correlates of Neuromuscular Locomotion in the Cat
Junction Development H. Sherk and G. A. Fowler
Mary B. Rheuben, Motojiro Yoshihara, and
Stages of Self-Motion Processing in Primate
Yoshiaki Kidokoro
Posterior Parietal Cortex
Assembly and Maturation of the Drosophila Larval F. Bremmer, J.-R. Duhamel, S. B. Hamed, and
Neuromuscular Junction W. Graf
L. Sian Gramates and Vivian Budnik
Optic Flow Analysis for Self-Movement
Second Messenger Systems Underlying Plasticity Perception
at the Neuromuscular Junction C. J. Duffy
Frances Hannan and Yi Zhong
Neural Mechanisms for Self-Motion Perception
Mechanisms of Neurotransmitter Release in Area MST
J. Troy Littleton, Leo Pallanck, and Barry R. A. Andersen, K. V. Shenoy, J. A. Crowell,
Ganetzky and D. C. Bradley
Contents of Recent Volumes 333
Volume 51 INDEX
Volume 56
Volume 55
Behavioral Mechanisms and the Neurobiology of
Section I: Virsu Vectors For Use in the Nervous Conditioned Sexual Responding
System Mark Krause
Non-Neurotropic Adenovirus: a Vector for Gene NMDA Receptors in Alcoholism
Transfer to the Brain and Gene Therapy of Neu- Paula L. Hoffman
rological Disorders
P. R. Lowenstein, D. Suwelack, J. Hu, X. Yuan, Processing and Representation of Species-Specific
M. Jimenez-Dalmaroni, S. Goverdhama, and Communication Calls in the Auditory System of
M.G. Castro Bats
George D. Pollak, Achim Klug, and Eric E. Bauer
Adeno-Associated Virus Vectors
E. Lehtonen and L. Tenenbaum Central Nervous System Control of Micturition
Gert Holstege and Leonora J. Mouton
Problems in the Use of Herpes Simplex Virus as a
Vector The Structure and Physiology of the Rat Auditory
L. T. Feldman System: An Overview
Manuel Malmierca
Lentiviral Vectors
J. Jakobsson, C. Ericson, N. Rosenquist, and Neurobiology of Cat and Human Sexual Behavior
C. Lundberg Gert Holstege and J. R. Georgiadis
Dopamine Transporter Network and Pathways Neuroimaging Studies in Bipolar Children and
Rajani Maiya and R. Dayne Mayfield Adolescents
Rene L. Olvera, David C. Glahn, Sheila
Proteomic Approaches in Drug Discovery
C. Caetano, Steven R. Pliszka, and Jair C. Soares
and Development
Holly D. Soares, Stephen A. Williams, Peter Chemosensory G-Protein-Coupled Receptor
J. Snyder, Feng Gao, Tom Stiger, Christian Rohlff, Signaling in the Brain
Athula Herath, Trey Sunderland, Karen Putnam, Geoffrey E. Woodard
and W. Frost White
Disturbances of Emotion Regulation after Focal
Section III: Informatics Brain Lesions
Antoine Bechara
Proteomic Informatics
Steven Russell, William Old, Katheryn Resing, The Use of Caenorhabditis elegans in Molecular
and Lawrence Hunter Neuropharmacology
Jill C. Bettinger, Lucinda Carnell, Andrew
Section IV: Changes in the Proteome by Disease
G. Davies, and Steven L. McIntire
Proteomics Analysis in Alzheimer’s Disease: New
INDEX
Insights into Mechanisms of Neurodegeneration
D. Allan Butterfield and Debra Boyd-Kimball
Proteomics and Alcoholism
Volume 63
Frank A. Witzmann and Wendy N. Strother Mapping Neuroreceptors at work: On the Defini-
tion and Interpretation of Binding Potentials after
Proteomics Studies of Traumatic Brain Injury
20 years of Progress
Kevin K. W. Wang, Andrew Ottens,
Albert Gjedde, Dean F. Wong, Pedro Rosa-Neto,
William Haskins, Ming Cheng Liu, Firas
and Paul Cumming
Kobeissy, Nancy Denslow, SuShing Chen, and
Ronald L. Hayes Mitochondrial Dysfunction in Bipolar Disorder:
From 31P-Magnetic Resonance Spectroscopic
Influence of Huntington’s Disease on the Human
Findings to Their Molecular Mechanisms
and Mouse Proteome
Tadafumi Kato
Claus Zabel and Joachim Klose
Large-Scale Microarray Studies of Gene Expres-
Section V: Overview of the Neuroproteome
sion in Multiple Regions of the Brain in Schizo-
Proteomics—Application to the Brain phrenia and Alzeimer’s Disease
Katrin Marcus, Oliver Schmidt, Heike Schaefer, Pavel L. Katsel, Kenneth L. Davis, and Vahram
Michael Hamacher, AndrÅ van Hall, and Helmut Haroutunian
E. Meyer
Regulation of Serotonin 2C Receptor PRE-
INDEX mRNA Editing By Serotonin
Claudia Schmauss
The Dopamine Hypothesis of Drug Addiction:
Volume 62 Hypodopaminergic State
Miriam Melis, Saturnino Spiga, and Marco Diana
GABAA Receptor Structure–Function Studies:
A Reexamination in Light of New Acetylcholine Human and Animal Spongiform Encephalopa-
Receptor Structures thies are Autoimmune Diseases: A Novel Theory
Myles H. Akabas and Its supporting Evidence
Bao Ting Zhu
Dopamine Mechanisms and Cocaine Reward
Aiko Ikegami and Christine L. Duvauchelle Adenosine and Brain Function
Bertil B. Fredholm, Jiang-Fan Chen, Rodrigo
Proteolytic Dysfunction in Neurodegenerative
A. Cunha, Per Svenningsson, and Jean-Marie Vaugeois
Disorders
Kevin St. P. McNaught INDEX
342 Contents of Recent Volumes
Effects of Genes and Stress on the Neurobiology of Artistic Changes in Alzheimer’s Disease
Depression Sebastian J. Crutch and Martin N. Rossor
J. John Mann and Dianne Currier
Section IV: Cerebrovascular Disease
Quantitative Imaging with the Micropet Small-
Stroke in Painters
Animal Pet Tomograph
H. Bäzner and M. Hennerici
Paul Vaska, Daniel J. Rubins, David L. Alexoff,
and Wynne K. Schiffer Visuospatial Neglect in Lovis Corinth’s Self-
Portraits
Understanding Myelination through Studying its
Olaf Blanke
Evolution
Rüdiger Schweigreiter, Betty I. Roots, Art, Constructional Apraxia, and the Brain
Christine Bandtlow, and Robert M. Gould Louis Caplan
INDEX Section V: Genetic Diseases
Neurogenetics in Art
Alan E. H. Emery
Volume 74 A Naı̈ve Artist of St Ives
Evolutionary Neurobiology and Art F. Clifford Rose
C. U. M. Smith
Van Gogh’s Madness
Section I: Visual Aspects F. Clifford Rose
Perceptual Portraits Absinthe, The Nervous System and Painting
Nicholas Wade Tiina Rekand
The Neuropsychology of Visual Art: Conferring Section VI: Neurologists as Artists
Capacity
Anjan Chatterjee Sir Charles Bell, KGH, FRS, FRSE
(1774–1842)
Vision, Illusions, and Reality Christopher Gardner-Thorpe
Christopher Kennard
Section VII: Miscellaneous
Localization in the Visual Brain
Peg Leg Frieda
George K. York
Espen Dietrichs
Section II: Episodic Disorders
The Deafness of Goya (1746–1828)
Neurology, Synaesthesia, and Painting F. Clifford Rose
Amy Ione
INDEX
Fainting in Classical Art
Philip Smith
Migraine Art in the Internet: A Study of 450
Contemporary Artists
Klaus Podoll
Volume 75
Introduction on the Use of the Drosophila Embry-
Sarah Raphael’s Migraine with Aura as Inspiration
onic/Larval Neuromuscular Junction as a Model
for the Foray of Her Work into Abstraction
System to Study Synapse Development and
Klaus Podoll and Debbie Ayles
Function, and a Brief Summary of Pathfinding
The Visual Art of Contemporary Artists with and Target Recognition
Epilepsy Catalina Ruiz-Cañada and Vivian Budnik
Steven C. Schachter
Development and Structure of Motoneurons
Section III: Brain Damage Matthias Landgraf and Stefan Thor
Creativity in Painting and Style in Brain- The Development of the Drosophila Larval Body
Damaged Artists Wall Muscles
Julien Bogousslavsky Karen Beckett and Mary K. Baylies
Contents of Recent Volumes 347
Organization of the Efferent System and Structure ID, Ego, and Temporal Lobe Revisited
of Neuromuscular Junctions in Drosophila Shirley M. Ferguson and Mark Rayport
Andreas Prokop
Section II: Stereotaxic Studies
Development of Motoneuron Electrical Proper-
Olfactory Gustatory Responses Evoked by
ties and Motor Output
Electrical Stimulation of Amygdalar Region in
Richard A. Baines
Man Are Qualitatively Modifiable by Interview
Transmitter Release at the Neuromuscular Junction Content: Case Report and Review
Thomas L. Schwarz Mark Rayport, Sepehr Sani, and Shirley M. Ferguson
Vesicle Trafficking and Recycling at the Neuro- Section III: Controversy in Definition of Behav-
muscular Junction: Two Pathways for Endocytosis ioral Disturbance
Yoshiaki Kidokoro
Pathogenesis of Psychosis in Epilepsy. The
Glutamate Receptors at the Drosophila Neuromus- “Seesaw” Theory: Myth or Reality?
cular Junction Shirley M. Ferguson and Mark Rayport
Aaron DiAntonio
Section IV: Outcome of Temporal Lobectomy
Scaffolding Proteins at the Drosophila Neuromus-
Memory Function After Temporal Lobectomy for
cular Junction
Seizure Control: A Comparative Neuropsy chi-
Bulent Ataman, Vivian Budnik, and Ulrich Thomas
atric and Neuropsychological Study
Synaptic Cytoskeleton at the Neuromuscular Shirley M. Ferguson, A. John McSweeny, and Mark
Junction Rayport
Catalina Ruiz-Cañada and Vivian Budnik
Life After Surgery for Temporolimbic Seizures
Plasticity and Second Messengers During Synapse Shirley M. Ferguson, Mark Rayport, and Carolyn
Development A. Schell
Leslie C. Griffith and Vivian Budnik
Appendix I
Retrograde Signaling that Regulates Synaptic Mark Rayport
Development and Function at the Drosophila Neu-
Appendix II: Conceptual Foundations of Studies
romuscular Junction
of Patients Undergoing Temporal Lobe Surgery
Guillermo Marques and Bing Zhang
for Seizure Control
Activity-Dependent Regulation of Transcription Mark Rayport
During Development of Synapses
INDEX
Subhabrata Sanyal and Mani Ramaswami
Experience-Dependent Potentiation of Larval
Neuromuscular Synapses
Christoph M. Schuster
Volume 77
Regenerating the Brain
Selected Methods for the Anatomical Study of
David A. Greenberg and Kunlin Jin
Drosophila Embryonic and Larval Neuromuscular
Junctions Serotonin and Brain: Evolution, Neuroplasticity,
Vivian Budnik, Michael Gorczyca, and Andreas and Homeostasis
Prokop Efrain C. Azmitia
INDEX
Therapeutic Approaches to Promoting Axonal
Regeneration in the Adult Mammalian Spinal Cord
Volume 76 Sari S. Hannila, Mustafa M. Siddiq, and Marie
T. Filbin
Section I: Physiological Correlates of Freud’s
Evidence for Neuroprotective Effects of Antipsy-
Theories
chotic Drugs: Implications for the Pathophysio-
The ID, the Ego, and the Temporal Lobe logy and Treatment of Schizophrenia
Shirley M. Ferguson and Mark Rayport Xin-Min Li and Haiyun Xu
348 Contents of Recent Volumes
Neurogenesis and Neuroenhancement in the Patho- Schizophrenia and the α7 Nicotinic Acetylcholine
physiology and Treatment of Bipolar Disorder Receptor
Robert J. Schloesser, Guang Chen, and Husseini Laura F. Martin and Robert Freedman
K. Manji
Histamine and Schizophrenia
Neuroreplacement, Growth Factor, and Small Jean-Michel Arrang
Molecule Neurotrophic Approaches for Treating
Cannabinoids and Psychosis
Parkinson’s Disease
Deepak Cyril D’Souza
Michael J. O’Neill, Marcus J. Messenger, Viktor
Lakics, Tracey K. Murray, Eric H. Karran, Philip Involvement of Neuropeptide Systems in Schizo-
G. Szekeres, Eric S. Nisenbaum, and Kalpana phrenia: Human Studies
M. Merchant Ricardo Cáceda, Becky Kinkead, and Charles
B. Nemeroff
Using Caenorhabditis elegans Models of Neuro-
degenerative Disease to Identify Neuroprotective Brain-Derived Neurotrophic Factor in Schizo-
Strategies phrenia and Its Relation with Dopamine
Brian Kraemer and Gerard D. Schellenberg Olivier Guillin, Caroline Demily, and Florence
Thibaut
Neuroprotection and Enhancement of Neurite
Outgrowth With Small Molecular Weight Com- Schizophrenia Susceptibility Genes: In Search of a
pounds From Screens of Chemical Libraries Molecular Logic and Novel Drug Targets for a
Donard S. Dwyer and Addie Dickson Devastating Disorder
Joseph A. Gogos
INDEX
INDEX
Volume 78
Neurobiology of Dopamine in Schizophrenia
Olivier Guillin, Anissa Abi-Dargham, and Marc Volume 79
Laruelle
The Destructive Alliance: Interactions of
The Dopamine System and the Pathophysiology Leukocytes, Cerebral Endothelial Cells, and the
of Schizophrenia: A Basic Science Perspective Immune Cascade in Pathogenesis of Multiple
Yukiori Goto and Anthony A. Grace Sclerosis
Alireza Minagar, April Carpenter, and J. Steven
Glutamate and Schizophrenia: Phencyclidine,
Alexander
N-methyl-D-aspartate Receptors, and Dopamine–
Glutamate Interactions Role of B Cells in Pathogenesis of Multiple
Daniel C. Javitt Sclerosis
Behrouz Nikbin, Mandana Mohyeddin Bonab,
Deciphering the Disease Process of Schizophrenia:
Farideh Khosravi, and Fatemeh Talebian
The Contribution of Cortical GABA Neurons
David A. Lewis and Takanori Hashimoto The Role of CD4 T Cells in the Pathogenesis of
Multiple Sclerosis
Alterations of Serotonin Transmission in
Tanuja Chitnis
Schizophrenia
Anissa Abi-Dargham The CD8 T Cell in Multiple Sclerosis: Suppressor
Cell or Mediator of Neuropathology?
Serotonin and Dopamine Interactions in Rodents
Aaron J. Johnson, Georgette L. Suidan, Jeremiah
and Primates: Implications for Psychosis and Anti-
McDole, and Istvan Pirko
psychotic Drug Development
Gerard J. Marek Immunopathogenesis of Multiple Sclerosis
Smriti M. Agrawal and V. Wee Yong
Cholinergic Circuits and Signaling in the Patho-
physiology of Schizophrenia Molecular Mimicry in Multiple Sclerosis
Joshua A. Berman, David A. Talmage, and Lorna Jane E. Libbey, Lori L. McCoy, and Robert
W. Role S. Fujinami
Contents of Recent Volumes 349
Life and Death of Neurons in the Aging Recruitment and Retention in Clinical Trials of
Cerebral Cortex the Elderly
John H. Morrison and Patrick R. Hof Flavia M. Macias, R. Eugene Ramsay, and
A. James Rowan
An In Vitro Model of Stroke-Induced Epilepsy:
Elucidation of the Roles of Glutamate and Treatment of Convulsive Status Epilepticus
Calcium in the Induction and Maintenance of David M. Treiman
Stroke-Induced Epileptogenesis Treatment of Nonconvulsive Status Epilepticus
Robert J. DeLorenzo, David A. Sun, Robert Matthew C. Walker
E. Blair, and Sompong Sambati
Antiepileptic Drug Formulation and Treatment
Mechanisms of Action of Antiepileptic Drugs in the Elderly: Biopharmaceutical Considerations
H. Steve White, Misty D. Smith, and Barry E. Gidal
Karen S. Wilcox
INDEX
Epidemiology and Outcomes of Status Epilepticus
in the Elderly
Alan R. Towne
New Insights into the Roles of Metalloproteinases Differential Modulation of Type 1 and Type 2
in Neurodegeneration and Neuroprotection Cannabinoid Receptors Along the Neuroimmune
A. J. Turner and N. N. Nalivaeva Axis
Sergio Oddi, Paola Spagnuolo, Monica Bari,
Relevance of High-Mobility Group Protein Antonella D’Agostino, and Mauro Maccarrone
Box 1 to Neurodegeneration
Silvia Fossati and Alberto Chiarugi Effects of the HIV-1 Viral Protein Tat on Central
Neurotransmission: Role of Group I Meta-
Early Upregulation of Matrix Metalloproteinases botropic Glutamate Receptors
Following Reperfusion Triggers Neuro-
Elisa Neri, Veronica Musante, and Anna Pittaluga
inflammatory Mediators in Brain Ischemia in Rat
Diana Amantea, Rossella Russo, Micaela Gliozzi, Evidence to Implicate Early Modulation of Inter-
Vincenza Fratto, Laura Berliocchi, G. Bagetta, leukin-1β Expression in the Neuroprotection
G. Bernardi, and M. Tiziana Corasaniti Afforded by 17β-Estradiol in Male Rats Under-
gone Transient Middle Cerebral Artery Occlusion
The (Endo)Cannabinoid System in Multiple Olga Chiappetta, Micaela Gliozzi, Elisa Siviglia,
Sclerosis and Amyotrophic Lateral Sclerosis
Diana Amantea, Luigi A. Morrone, Laura
Diego Centonze, Silvia Rossi, Alessandro Berliocchi, G. Bagetta, and M. Tiziana Corasaniti
Finazzi-Agrò, Giorgio Bernardi, and Mauro
Maccarrone A Role for Brain Cyclooxygenase-2 and Prosta-
glandin-E2 in Migraine: Effects of Nitroglycerin
Chemokines and Chemokine Receptors: Multi- Cristina Tassorelli, Rosaria Greco, Marie Therese
purpose Players in Neuroinflammation
Armentero, Fabio Blandini, Giorgio Sandrini, and
Richard M. Ransohoff, LiPing Liu, and Astrid Giuseppe Nappi
E. Cardona
The Blockade of K+-ATP Channels has Neuro-
Systemic and Acquired Immune Responses in protective Effects in an In Vitro Model of Brain
Alzheimer’s Disease Ischemia
Markus Britschgi and Tony Wyss-Coray
Robert Nisticò, Silvia Piccirilli, L. Sebastianelli,
Neuroinflammation in Alzheimer’s Disease and Giuseppe Nisticò, G. Bernardi, and N. B. Mercuri
Parkinson’s Disease: Are Microglia Pathogenic
Retinal Damage Caused by High Intraocular
in Either Disorder?
Pressure-Induced Transient Ischemia is Prevented
Joseph Rogers, Diego Mastroeni, Brian Leonard,
by Coenzyme Q10 in Rat
Jeffrey Joyce, and Andrew Grover
Carlo Nucci, Rosanna Tartaglione, Angelica
Cytokines and Neuronal Ion Channels in Health Cerulli, R. Mancino, A. Spanò, Federica Cavaliere,
and Disease Laura Rombolà, G. Bagetta, M. Tiziana
Barbara Viviani, Fabrizio Gardoni, and Marina Corasaniti, and Luigi A. Morrone
Marinovich
Evidence Implicating Matrix Metalloproteinases
Cyclooxygenase-2, Prostaglandin E2, and Micro- in the Mechanism Underlying Accumulation of
glial Activation in Prion Diseases IL-1β and Neuronal Apoptosis in the Neocortex
Luisa Minghetti and Maurizio Pocchiari of HIV/gp120-Exposed Rats
Rossella Russo, Elisa Siviglia, Micaela Gliozzi,
Glia Proinflammatory Cytokine Upregulation as a
Diana Amantea, Annamaria Paoletti,
Therapeutic Target for Neurodegenerative
Laura Berliocchi, G. Bagetta, and
Diseases: Function-Based and Target-Based
M. Tiziana Corasaniti
Discovery Approaches
Linda J. Van Eldik, Wendy L. Thompson, Neuroprotective Effect of Nitroglycerin in a
Hantamalala Ralay Ranaivo, Heather A. Behanna, Rodent Model of Ischemic Stroke: Evaluation
and D. Martin Watterson of Bcl-2 Expression
Rosaria Greco, Diana Amantea, Fabio Blandini,
Oxidative Stress and the Pathogenesis of Neuro-
Giuseppe Nappi, Giacinto Bagetta, M. Tiziana
degenerative Disorders
Corasaniti, and Cristina Tassorelli
Ashley Reynolds, Chad Laurie, R. Lee Mosley, and
Howard E. Gendelman INDEX
Contents of Recent Volumes 353
Involvement of the Prefrontal Cortex in Problem Bv8/Prokineticins and their Receptors: A New
Solving Pronociceptive System
Hajime Mushiake, Kazuhiro Sakamoto, Naohiro Lucia Negri, Roberta Lattanzi, Elisa Giannini,
Saito, Toshiro Inui, Kazuyuki Aihara, and Jun Michela Canestrelli, Annalisa Nicotra,
Tanji and Pietro Melchiorri
Contents of Recent Volumes 355
Bidirectional Interfaces with the Peripheral Section Four: Brain-Machine Interfaces and
Nervous System Space
Silvestro Micera and Xavier Navarro Adaptive Changes of Rhythmic EEG Oscillations
in Space: Implications for Brain–Machine
Interfacing Insect Brain for Space Applications
Interface Applications
Giovanni Di Pino, Tobias Seidl,
G. Cheron, A. M. Cebolla, M. Petieau,
Antonella Benvenuto, Fabrizio Sergi, Domenico
A. Bengoetxea, E. Palmero-Soler, A. Leroy, and
Campolo, Dino Accoto, Paolo Maria Rossini,
B. Dan
and Eugenio Guglielmelli
Validation of Brain–Machine Interfaces During
Section Two: Meet the Brain
Parabolic Flight
Meet the Brain: Neurophysiology
Jose del R. Millán, Pierre W. Ferrez, and Tobias
John Rothwell
Seidl
Fundamentals of Electroencefalography, Magne-
Matching Brain–Machine Interface Performance
toencefalography, and Functional Magnetic
to Space Applications
Resonance Imaging
Luca Citi, Oliver Tonet, and Martina Marinelli
Claudio Babiloni, Vittorio Pizzella, Cosimo Del
Gratta, Antonio Ferretti, and Gian Luca Romani Brain–Machine Interfaces for Space
Applications—Research, Technological Devel-
Implications of Brain Plasticity to Brain–Machine
opment, and Opportunities
Interfaces Operation: A Potential Paradox?
Leopold Summerer, Dario Izzo, and Luca Rossini
Paolo Maria Rossini
INDEX
Section Three: Brain Machine Interfaces, A New
Brain-to-Environment Communication Channel
An Overview of BMIs
Francisco Sepulveda Volume 87
Neurofeedback and Brain–Computer Interface: Peripheral Nerve Repair and Regeneration
Clinical Applications Research: A Historical Note
Niels Birbaumer, Ander Ramos Murguialday, Bruno Battiston, Igor Papalia, Pierluigi Tos, and
Cornelia Weber, and Pedro Montoya Stefano Geuna
Flexibility and Practicality: Graz Brain–Computer Development of the Peripheral Nerve
Interface Approach Suleyman Kaplan, Ersan Odaci, Bunyami Unal,
Reinhold Scherer, Gernot R. Müller-Putz, and Bunyamin Sahin, and Michele Fornaro
Gert Pfurtscheller
Histology of the Peripheral Nerve and Changes
On the Use of Brain–Computer Interfaces Out- Occurring During Nerve Regeneration
side Scientific Laboratories: Toward an Applica- Stefano Geuna, Stefania Raimondo, Giulia Ronchi,
tion in Domotic Environments Federica Di Scipio, Pierluigi Tos, Krzysztof Czaja,
F. Babiloni, F. Cincotti, M. Marciani, S. Salinari, and Michele Fornaro
L. Astolfi, F. Aloise, F. De Vico Fallani, and
Methods and Protocols in Peripheral Nerve
D. Mattia
Regeneration Experimental Research:
Brain–Computer Interface Research at the Part I—Experimental Models
Wadsworth Center: Developments in Noninva- Pierluigi Tos, Giulia Ronchi, Igor Papalia,
sive Communication and Control Vera Sallen, Josette Legagneux, Stefano Geuna, and
Dean J. Krusienski and Jonathan R. Wolpaw Maria G. Giacobini-Robecchi
Watching Brain TV and Playing Brain Ball: Methods and Protocols in Peripheral Nerve
Exploring Novel BCL Strategies Using Real– Regeneration Experimental Research: Part
Time Analysis of Human Intercranial Data II—Morphological Techniques
Karim Jerbi, Samson Freyermuth, Lorella Minotti, Stefania Raimondo, Michele Fornaro, Federica Di
Philippe Kahane, Alain Berthoz, and Jean-Philippe Scipio, Giulia Ronchi, Maria G. Giacobini-
Lachaux Robecchi, and Stefano Geuna
Contents of Recent Volumes 357
Deciphering Rett Syndrome With Mouse Genet- Part III—Transcranial Sonography in other
ics, Epigenomics, and Human Neurons Movement Disorders and Depression
Jifang Tao, Hao Wu, and Yi Eve Sun
Transcranial Sonography in Brain Disorders with
INDEX Trace Metal Accumulation
Uwe Walter
Transcranial Sonography in Dystonia
Volume 90 Alexandra Gaenslen
Part I: Introduction Transcranial Sonography in Essential Tremor
Heike Stockner and Isabel Wurster
Introductory Remarks on the History and Current
Applications of TCS VII—Transcranial Sonography in Restless Legs
Matthew B. Stern Syndrome
Jana Godau and Martin Sojer
Method and Validity of Transcranial Sonography
in Movement Disorders Transcranial Sonography in Ataxia
David Školoudı´k and Uwe Walter Christos Krogias, Thomas Postert and Jens Eyding
Transcranial Sonography—Anatomy Transcranial Sonography in Huntington’s Disease
Heiko Huber Christos Krogias, Jens Eyding and Thomas Postert
Transcranial Sonography in Depression
Part II: Transcranial Sonography in Parkinsons Milija D. Mijajlovic
Disease
Transcranial Sonography in Relation to SPECT Part IV: Future Applications and Conclusion
and MIBG Transcranial Sonography-Assisted Stereotaxy and
Yoshinori Kajimoto, Hideto Miwa and Tomoyoshi Follow-Up of Deep Brain Implants in Patients
Kondo with Movement Disorders
Diagnosis of Parkinson’s Disease—Transcranial Uwe Walter
Sonography in Relation to MRI Conclusions
Ludwig Niehaus and Kai Boelmans Daniela Berg
Early Diagnosis of Parkinson’s Disease INDEX
Alexandra Gaenslen and Daniela Berg
Transcranial Sonography in the Premotor Diag-
nosis of Parkinson’s Disease
Stefanie Behnke, Ute Schroder and Daniela Berg Volume 91
Pathophysiology of Transcranial Sonography Sig- The Role of microRNAs in Drug Addiction:
nal Changes in the Human Substantia Nigra A Big Lesson from Tiny Molecules
K. L. Double, G. Todd and S. R. Duma Andrzej Zbigniew Pietrzykowski
Transcranial Sonography for the Discrimination of The Genetics of Behavioral Alcohol Responses in
Idiopathic Parkinson’s Disease from the Atypical Drosophila
Parkinsonian Syndromes Aylin R. Rodan and Adrian Rothenfluh
A. E. P. Bouwmans, A. M. M. Vlaar, K. Srulijes,
Neural Plasticity, Human Genetics, and Risk for
W. H. Mess AND W. E. J. Weber
Alcohol Dependence
Transcranial Sonography in the Discrimination of Shirley Y. Hill
Parkinson’s Disease Versus Vascular Parkinsonism
Using Expression Genetics to Study the Neurobi-
Pablo Venegas-Francke
ology of Ethanol and Alcoholism
TCS in Monogenic Forms of Parkinson’s Disease Sean P. Farris, Aaron R. Wolen and Michael
Kathrin Brockmann and Johann Hagenah F. Miles
360 Contents of Recent Volumes
Genetic Variation and Brain Gene Expression in Neuroimaging of Dreaming: State of the Art and
Rodent Models of Alcoholism: Implications for Limitations
Medication Development Caroline Kusse, Vincenzo Muto, Laura Mascetti,
Karl Bj€ork, Anita C. Hansson and Luca Matarazzo, Ariane Foret, Anahita Shaffii-Le
W. olfgang H. Sommer Bourdiec and Pierre Maquet
Identifying Quantitative Trait Loci (QTLs) and Memory Consolidation, The Diurnal Rhythm of
Genes (QTGs) for Alcohol-Related Phenotypes Cortisol, and The Nature of Dreams: A New
in Mice Hypothesis
Lauren C. Milner and Kari J. Buck Jessica D. Payne
Glutamate Plasticity in the Drunken Amygdala: Characteristics and Contents of Dreams
The Making of an Anxious Synapse Michael Schredl
Brian A. Mccool, Daniel T. Christian, Marvin
Trait and Neurobiological Correlates of Individ-
R. Diaz and Anna K. Läck
ual Differences in Dream Recall and Dream
Ethanol Action on Dopaminergic Neurons in Content
the Ventral Tegmental Area: Interaction with Mark Blagrove and Edward F. Pace-Schott
Intrinsic Ion Channels and Neurotransmitter
Consciousness in Dreams
Inputs
David Kahn and Tzivia Gover
Hitoshi Morikawa and Richard
A. Morrisett The Underlying Emotion and the Dream: Relat-
ing Dream Imagery to the Dreamer’s Underlying
Alcohol and the Prefrontal Cortex
Emotion can Help Elucidate the Nature of
Kenneth Abernathy, L. Judson Chandler and John
Dreaming
J. Woodward
Ernest Hartmann
BK Channel and Alcohol, A Complicated Affair
Dreaming, Handedness, and Sleep Architecture:
Gilles Erwan Martin
Interhemispheric Mechanisms
A Review of Synaptic Plasticity at Purkinje Neu- Stephen D. Christman and Ruth E. Propper
rons with a Focus on Ethanol-Induced Cerebellar
To What Extent Do Neurobiological Sleep-
Dysfunction
Waking Processes Support Psychoanalysis?
C. Fernando Valenzuela, Britta Lindquist and
Claude Gottesmann
Paula A. Zflmudio-Bulcock
The Use of Dreams in Modern Psychotherapy
INDEX
Clara E. Hill and Sarah Knox
INDEX
Volume 92
The Development of the Science of Dreaming Volume 93
Claude Gottesmann
Underlying Brain Mechanisms that Regulate
Dreaming as Inspiration: Evidence from Religion, Sleep-Wakefulness Cycles
Philosophy, Literature, and Film Irma Gvilia
Kelly Bulkeley
What Keeps Us Awake?—the Role of Clocks and
Developmental Perspective: Dreaming Across the Hourglasses, Light, and Melatonin
Lifespan and What This Tells Us Christian Cajochen, Sarah Chellappa and Christina
Melissa M. Burnham and Christian Conte Schmidt
REM and NREM Sleep Mentation Suprachiasmatic Nucleus and Autonomic Nervous
Patrick Mcnamara, Patricia Johnson, Deirdre System Influences on Awakening From Sleep
McLaren, Erica Harris,Catherine Beauharnais and Andries Kalsbeek, Chun-xia Yi, Susanne E.
Sanford Auerbach la Fleur, Ruud m. Buijs, and Eric Fliers
Contents of Recent Volumes 361
Volume 97 Volume 98
Behavioral Pharmacology of Orofacial Movement
An Introduction to Dyskinesia—the Clinical
Disorders
Spectrum
Noriaki Koshikawa, Satoshi Fujita and Kazunori
Ainhi Ha and Joseph Jankovic
Adachi
L-dopa-induced Dyskinesia—Clinical Presenta-
Regulation of Orofacial Movement: Dopamine
tion, Genetics, And Treatment
Receptor Mechanisms and Mutant Models
L.K. Prashanth, Susan Fox and Wassilios
John L. Waddington, Gerard J. O’Sullivan and
G. Meissner
Katsunori Tomiyama
Experimental Models of L-DOPA-induced
Regulation of Orofacial Movement: Amino Acid
Dyskinesia
Mechanisms and Mutant Models
Tom H. Johnston and Emma L. Lane
Katsunori Tomiyama, Colm M.P. O’Tuathaigh,
and John L. Waddington Molecular Mechanisms of L-DOPA-induced
Dyskinesia
The Trigeminal Circuits Responsible for
Gilberto Fisone and Erwan Bezard
Chewing
Karl-Gunnar Westberg and Arlette Kolta New Approaches to Therapy
Jonathan Brotchie and Peter Jenner
Ultrastructural Basis for Craniofacial Sensory
Processing in the Brainstem Surgical Approach to L-DOPA-induced
Yong Chul Bae and Atsushi Yoshida Dyskinesias
Tejas Sankar and Andres M. Lozano
Mechanisms of Nociceptive Transduction and
Transmission: A Machinery for Pain Sensation Clinical and Experimental Experiences of
and Tools for Selective Analgesia Graft-induced Dyskinesia
Alexander M. Binshtok Emma L. Lane
Contents of Recent Volumes 363
Multimodal Drugs and their Future for Abnormalities in Metabolism and Hypothalamic–
Alzheimer’s and Parkinson’s Disease Pituitary–Adrenal Axis Function in Schizophrenia
Cornelis J. Van der Schyf and Werner J. Geldenhuys Paul C. Guest, Daniel Martins-de-Souza,
Natacha Vanattou-Saifoudine, Laura W. Harris
Neuroprotective Profile of the Multitarget Drug
and Sabine Bahn
Rasagiline in Parkinson’s Disease
Orly Weinreb, Tamar Amit, Peter Riederer, Immune and Neuroimmune Alterations in Mood
Moussa B.H. Youdim and Silvia A. Mandel Disorders and Schizophrenia
Roosmarijn C. Drexhage, Karin Weigelt, Nico van
Rasagiline in Parkinson’s Disease
Beveren, Dan Cohen, Marjan A. Versnel, Willem
L.M. Chahine and M.B. Stern
A. Nolen and Hemmo A. Drexhage
Selective Inhibitors of Monoamine Oxidase Type
Behavioral and Molecular Biomarkers in Transla-
B and the “Cheese Effect”
tional Animal Models for Neuropsychiatric
John P.M. Finberg and Ken Gillman
Disorders
A Novel Anti-Alzheimer’s Disease Drug, Ladostigil: Zoltán Sarnyai, Murtada Alsaif, Sabine Bahn,
Neuroprotective, Multimodal Brain-Selective Agnes Ernst, Paul C. Guest, Eva Hradetzky,
Monoamine Oxidase and Cholinesterase Inhibitor Wolfgang Kluge, Viktoria Stelzhammer and
Orly Weinreb, Tamar Amit, Orit Bar-Am and Hendrik Wesseling
Moussa B.H. Youdim
Stem Cell Models for Biomarker Discovery in
Novel MAO-B Inhibitors: Potential Therapeutic Brain Disease
Use of the Selective MAO-B Inhibitor PF9601N Alan Mackay-Sim, George Mellick and Stephen
in Parkinson’s Disease Wood
Mercedes Unzeta and Elisenda Sanz
The Application of Multiplexed Assay Systems for
INDEX Molecular Diagnostics
Emanuel Schwarz, Nico J.M. VanBeveren,
Paul C. Guest, Rauf Izmailov and
Volume 101 Sabine Bahn
General Overview: Biomarkers in Neuroscience Algorithm Development for Diagnostic Bio-
Research marker Assays
Michaela D. Filiou and Christoph W. Turck Rauf Izmailov, Paul C. Guest, Sabine Bahn and
Emanuel Schwarz
Imaging Brain Microglial Activation Using
Positron Emission Tomography and Translocator Challenges of Introducing New Biomarker Prod-
Protein-Specific Radioligands ucts for Neuropsychiatric Disorders into the
David R.J. Owen and Paul M. Matthews Market
The Utility of Gene Expression in Blood Cells for Sabine Bahn, Richard Noll, Anthony Barnes,
Diagnosing Neuropsychiatric Disorders Emanuel Schwarz and Paul C. Guest
Christopher H. Woelk, Akul Singhania, Josue Toward Personalized Medicine in the Neuropsy-
Perez-Santiago, Stephen J. Glatt and Ming chiatric Field
T. Tsuang Erik H.F. Wong, Jayne C. Fox, Mandy
Proteomic Technologies for Biomarker Studies in Y.M. Ng and Chi-Ming Lee
Psychiatry: Advances and Needs Clinical Utility of Serum Biomarkers for Major
Daniel Martins-de-Souza, Paul C. Guest, Psychiatric Disorders
Natacha Vanattou-Saifoudine, Laura W. Harris Nico J.M. van Beveren and Witte
and Sabine Bahn J.G. Hoogendijk
Converging Evidence of Blood-Based Biomarkers
The Future: Biomarkers, Biosensors, Neu-
for Schizophrenia: An update
roinformatics, and E-Neuropsychiatry
Man K. Chan, Paul C. Guest, Yishai Levin,
Christopher R. Lowe
Yagnesh Umrania, Emanuel Schwarz, Sabine Bahn
and Hassan Rahmoune SUBJECT INDEX
Contents of Recent Volumes 365
Neurophysiology of Deep Brain Stimulation Bone Marrow Mesenchymal Stem Cell Trans-
Manuela Rosa, Gaia Giannicola, Sara Marceglia, plantation for Improving Nerve Regeneration
Manuela Fumagalli, Sergio Barbieri, and Alberto Priori Júlia Teixeira Oliveira, Klauss Mostacada, Silmara
de Lima, and Ana Maria Blanco Martinez
Neurophysiology of Cortical Stimulation
Jean-Pascal Lefaucheur Perspectives of Employing Mesenchymal Stem
Cells from the Wharton’s Jelly of the Umbilical
Neural Mechanisms of Spinal Cord Stimulation
Cord for Peripheral Nerve Repair
Robert D. Foreman and Bengt Linderoth
Jorge Ribeiro, Andrea Gartner, Tiago Pereira,
Magnetoencephalography and Neuromodulation Raquel Gomes, Maria Ascensão Lopes,
Alfons Schnitzler and Jan Hirschmann Carolina Gonçalves, Artur Varejão, Ana Lúcia
Luı´s, and Ana Colette Maurı´cio
Current Challenges to the Clinical Translation of
Brain Machine Interface Technology Adipose-Derived Stem Cells and Nerve Regener-
Charles W. Lu, Parag G. Patil, and Cynthia A. ation: Promises and Pitfalls
Chestek Alessandro Faroni, Giorgio Terenghi, and
Adam J. Reid
Nanotechnology in Neuromodulation
Russell J. Andrews The Pros and Cons of Growth Factors and Cyto-
kines in Peripheral Axon Regeneration
Optogenetic Neuromodulation
Lars Klimaschewski, Barbara Hausott, and Doychin
Paul S. A. Kalanithi and Jaimie M. Henderson
N. Angelov
Diffusion Tensor Imaging and Neuromodulation:
Role of Inflammation and Cytokines in Peripheral
DTI as Key Technology for Deep Brain
Nerve Regeneration
Stimulation
P. Dubový, R. Jancˇálek, and T. Kubek
Volker Arnd Coenen, Thomas E. Schlaepfer, Niels
Allert, and Burkhard Mädler Ghrelin: A Novel Neuromuscular Recovery Pro-
moting Factor?
DBS and Electrical Neuro-Network Modulation
Raimondo Stefania, Ronchi Giulia, Geuna Stefano,
to Treat Neurological Disorders
Pascal Davide, Reano Simone, Filigheddu Nicoletta,
Amanda Thompson, Takashi Morishita, and
and Graziani Andrea
Michael S. Okun
Neuregulin 1 Role in Schwann Cell Regulation
Neuromodulation in Psychiatric Disorders
and Potential Applications to Promote Peripheral
Yasin Temel, Sarah A. Hescham, Ali Jahanshahi,
Nerve Regeneration
Marcus L. F. Janssen, Sonny K. H. Tan, Jacobus
Giovanna Gambarotta, Federica Fregnan, Sara
J. van Overbeeke, Linda Ackermans, Mayke
Gnavi, and Isabelle Perroteau
Oosterloo, Annelien Duits, Albert F. G. Leentjens,
and LeeWei Lim Extracellular Matrix Components in Peripheral
Nerve Regeneration
Ethical Aspects of Neuromodulation
Francisco Gonzalez-Perez, Esther Udina, and
Christiane Woopen
Xavier Navarro
SUBJECT INDEX
SUBJECT INDEX
Volume 108
Tissue Engineering and Regenerative Medicine:
Volume 109
Past, Present, and Future The Use of Chitosan-Based Scaffold to Enhance
António J. Salgado, Joaquim M. Oliveira, Albino Regeneration in the Nervous System
Martins, Fábio G. Teixeira, Nuno A. Silva, Sara Gnavi, Christina Barwig, Thomas Freier,
Nuno M. Neves, Nuno Sousa, and Rui L. Reis Kirsten Haarstert-Talini, Claudia Grothe, and
Stefano Geuna
Tissue Engineering and Peripheral Nerve Recon-
struction: An Overview Interfaces with the Peripheral Nerve for the Con-
Stefano Geuna, S. Gnavi, I. Perroteau, trol of Neuroprostheses
Pierluigi Tos, and B. Battiston Jaume del Valle and Xavier Navarro
368 Contents of Recent Volumes
The Use of Shock Waves in Peripheral Nerve The Neuropathology of Neurodegeneration with
Regeneration: New Perspectives? Brain Iron Accumulation
Thomas Hausner and Antal Nógrádi Michael C. Kruer
Phototherapy and Nerve Injury: Focus on Muscle Imaging of Iron
Response Petr Dusek, Monika Dezortova, and Jens Wuerfel
Shimon Rochkind, Stefano Geuna, and Asher
The Role of Iron Imaging in Huntington’s Disease
Shainberg
S.J.A. van den Bogaard, E.M. Dumas, and
Electrical Stimulation for Promoting Peripheral R.A.C. Roos
Nerve Regeneration
Lysosomal Storage Disorders and Iron
Kirsten Haastert-Talini and Claudia Grothe
Jose Miguel Bras
Role of Physical Exercise for Improving Post-
Manganese and the Brain
traumatic Nerve Regeneration
Karin Tuschl, Philippa B. Mills, and Peter T. Clayton
Paulo A.S. Armada-da-Silva, Cátia Pereira,
SandraAmado, and António P. Veloso Update on Wilson Disease
Aggarwal Annu and Bhatt Mohit
The Role of Timing in Nerve Reconstruction
Lars B. Dahlin An Update on Primary Familial Brain Calcification
R.R. Lemos, J.B.M.M. Ferreira, M.P. Keasey,
Future Perspectives in Nerve Repair and
and J.R.M. Oliveira
Regeneration
Pierluigi Tos, Giulia Ronchi, Stefano Geuna, and INDEX
Bruno Battiston
INDEX
Volume 111
Volume 110 History of Acupuncture Research
Yi Zhuang, Jing-jing Xing, Juan Li, Bai-Yun Zeng,
The Relevance of Metals in the Pathophysiology of and Fan-rong Liang
Neurodegeneration, Pathological Considerations
Effects of Acupuncture Needling with Specific
Kurt A. Jellinger
Sensation on Cerebral Hemodynamics and
Pantothenate Kinase-Associated Neurodegener- Autonomic Nervous Activity in Humans
ation (PKAN) and PLA2G6-Associated Neuro- Kouich Takamoto, Susumu Urakawa, Kazushige
degeneration (PLAN): Review of Two Major Sakai, Taketoshi Ono, and Hisao Nishijo
Neurodegeneration with Brain Iron Accumula-
Acupuncture Point Specificity
tion (NBIA) Phenotypes
Jing-jing Xing, Bai-Yun Zeng, Juan Li, Yi Zhuang,
Manju A. Kurian and Susan J. Hayflick
and Fan-rong Liang
Mitochondrial Membrane Protein-Associated
Acupuncture Stimulation Induces Neurogenesis
Neurodegeneration (MPAN)
in Adult Brain
Monika Hartig, Holger Prokisch, Thomas Meitinger,
Min-Ho Nam, Kwang Seok Ahn, and Seung-Hoon
and Thomas Klopstock
Choi
BPAN: The Only X-Linked Dominant NBIA
Acupuncture and Neurotrophin Modulation
Disorder
Marzia Soligo, Stefania Lucia Nori, Virginia Protto,
T.B. Haack, P. Hogarth, A. Gregory, P. Prokisch,
Fulvio Florenzano, and Luigi Manni
and S.J. Hayflick
Acupuncture Stimulation and Neuroendocrine
Neuroferritinopathy
Regulation
M.J. Keogh, C.M. Morris, and P.F. Chinnery
Jung-Sheng Yu, Bai-Yun Zeng, and
Aceruloplasminemia: An Update Ching-Liang Hsieh
Satoshi Kono
Current Development of Acupuncture Research
Therapeutic Advances in Neurodegeneration with in Parkinson’s Disease
Brain Iron Accumulation Bai-Yun Zeng, Sarah Salvage, and
Giovanna Zorzi and Nardo Nardocci Peter Jenner
Contents of Recent Volumes 369
Acupuncture Therapy for Stroke Patients Animal Models Recapitulating the Multifactorial
Xin Li and Qiang Wang Origin of Tourette Syndrome
Simone Macrı`, Martina Proietti Onori, Veit
Effects of Acupuncture Therapy on
Roessner, and Giovanni Laviola
Alzheimer’s Disease
Bai-Yun Zeng, Sarah Salvage, and Peter Jenner Neuroendocrine Aspects of Tourette Syndrome
Davide Martino, Antonella Macerollo, and
Acupuncture Therapy for Psychiatric Illness
James F. Leckman
Karen Pilkington
Clinical Pharmacology of Dopamine-Modulating
Acupuncture for the Treatment of Insomnia
Agents in Tourette’s Syndrome
Kaicun Zhao
Sabine Mogwitz, Judith Buse, Stefan Ehrlich, and
Acupuncture for the Treatment of Drug Veit Roessner
Addiction
Clinical Pharmacology of Nondopaminergic
Cai-Lian Cui, Liu-Zhen Wu, and Yi-jing Li
Drugs in Tourette Syndrome
Acupuncture Regulation of Blood Pressure: Andreas Hartmann
Two Decades of Research
Antiepileptic Drugs and Tourette Syndrome
John C. Longhurst and Stephanie Tjen-A-Looi
Andrea E. Cavanna and Andrea Nani
Effect and Mechanism of Acupuncture on
Clinical Pharmacology of Comorbid Obsessive–
Gastrointestinal Diseases
Compulsive Disorder in Tourette Syndrome
Toku Takahashi
Valeria Neri and Francesco Cardona
INDEX
Clinical Pharmacology of Comorbid Attention
Deficit Hyperactivity Disorder in Tourette
Syndrome
Volume 112 Renata Rizzo and Mariangela Gulisano
Emerging Treatment Strategies in Tourette
An Introduction to the Clinical Phenomenology
Syndrome: What’s in the Pipeline?
of Tourette Syndrome
Davide Martino, Namrata Madhusudan, Panagiotis C. Termine, C. Selvini, G. Rossi, and
U. Balottin
Zis, and Andrea E. Cavanna
Tics and Other Stereotyped Movements as Side
Functional Neuroanatomy of Tics
Effects of Pharmacological Treatment
Irene Neuner, Frank Schneider, and N. Jon Shah
Marcos Madruga-Garrido and Pablo Mir
Functional Imaging of Dopaminergic Neurotrans-
INDEX
mission in Tourette Syndrome
Bàrbara Segura and Antonio P. Strafella
Nondopaminergic Neurotransmission in the
Pathophysiology of Tourette Syndrome
Volume 113
Patrick T. Udvardi, Ester Nespoli, Autism Spectrum Disorder and the Cerebellum
Francesca Rizzo, Bastian Hengerer, and Esther B.E. Becker and Catherine J. Stoodley
Andrea G. Ludolph
Contribution of Long Noncoding RNAs to
Reinforcement Learning and Tourette Syndrome Autism Spectrum Disorder Risk
Stefano Palminteri and Mathias Pessiglione Brent Wilkinson and Daniel B. Campbell
Genetic Susceptibility and Neurotransmitters in Identifying Essential Cell Types and Circuits in
Tourette Syndrome Autism Spectrum Disorders
Peristera Paschou, Thomas V. Fernandez, Susan E. Maloney, Michael A. Rieger, and Joseph
Frank Sharp, Gary A. Heiman, and D. Dougherty
Pieter J. Hoekstra
Connecting Signaling Pathways Underlying
Pharmacological Animal Models of Tic Communication to ASD Vulnerability
Disorders Stephanie Lepp, Ashley Anderson, and Genevieve
Kevin W. McCairn and Masaki Isoda Konopka
370 Contents of Recent Volumes
Neuroimmune Basis of Alcoholic Brain Damage Adenosine Receptors and Huntington’s Disease
Fulton T. Crews and Ryan P. Vetreno Chien-fei Lee and Yijuang Chern
Converging Actions of Alcohol on Liver and Adenosine Receptors and Epilepsy: Current Evi-
Brain Immune Signaling dence and Future Potential
Gyongyi Szabo and Dora Lippai Susan A. Masino, Masahito Kawamura, Jr., and
David N. Ruskin
Opportunities for the Development of
Neuroimmune Therapies in Addiction Adenosine Receptor Control of Cognition in
Lara A. Ray, Daniel Roche, Keith Heinzerling, and Normal and Disease
Steve Shoptaw Jiang-Fan Chen
Use of Addictive Substances and NeuroHIV Adenosine Receptors in Cerebral Ischemia
Sulie L. Chang, Kaitlyn P. Connaghan, Yufeng Alessia Melani, Anna Maria Pugliese, and Felicita
Wei, and Ming D. Li Pedata
INDEX Roles of Adenosine and its Receptors in Sleep–
Wake Regulation
Zhi-Li Huang, Ze Zhang, and Wei-Min Qu
Volume 119 Involvement of Adenosine A2A Receptors in
Adenosine Receptor Neurobiology: Overview Depression and Anxiety
Jiang-Fan Chen, Chien-fei Lee, and Yijuang Chern Koji Yamada, Minoru Kobayashi, and Tomoyuki
Kanda
Adenosine Receptor PET Imaging in Human
Brain The Adenosine Neuromodulation System in
Masahiro Mishina and Kiich Ishiwata Schizophrenia
Daniel Rial, Diogo R. Lara, and Rodrigo A. Cunha
An Overview of Adenosine A2A Receptor Antag-
onists in Parkinson’s Disease INDEX
Peter Jenner
Mode of Action of Adenosine A2A Receptor Volume 120
Antagonists as Symptomatic Treatment for Par-
The Story of “Speed” from “Cloud Nine” to
kinson’s Disease
Brain Gain
Akihisa Mori
Andrew Lees, Katrin Sikk, and Pille Taba
Adenosine Receptors and Dyskinesia in
Amphetamine-Type Stimulants: The Early His-
Pathophysiology
tory of Their Medical and Non-Medical Uses
Masahiko Tomiyama
Nicolas Rasmussen
Clinical/Pharmacological Aspect of Adenosine
Miracle or Menace?
A2A Receptor Antagonist for Dyskinesia
Mike Jay
Tomoyuki Kanda and Shin-ichi Uchida
Psychostimulants: Basic and Clinical Pharmacology
Interaction of Adenosine Receptors with Other
Andrew C. McCreary, Christian P. Müller, and
Receptors from Therapeutic Perspective in Par-
Małgorzata Filip
kinson’s Disease
Nicolas Morin and Thérèse Di Paolo Epigenetic Mechanisms of Psychostimulant-
Induced Addiction
Effects of the Adenosine A2A Receptor Antagonist
Anti Kalda and Alexander Zharkovsky
on Cognitive Dysfunction in Parkinson’s Disease
Shin-ichi Uchida, Takako Kadowaki-Horita, and Experimental Models on Effects of Psycho-
Tomoyuki Kanda stimulants
Sulev Kõks
Clinical Nonmotor Aspect of A2A Antagonist in
PD Treatment Neurologic Complications of Psychomotor Stim-
Masahiro Nomoto, Masahiro Nagai, and Noriko ulant Abuse
Nishikawa Juan Sanchez-Ramos
Contents of Recent Volumes 373
Structural Insights into GIRK Channel Function The Role of Depression in the Uptake and Main-
Ian W. Glaaser and Paul A. Slesinger tenance of Cigarette Smoking
Janet Audrain-McGovern, Adam M. Leventhal,
Localization and Targeting of GIRK Channels in
and David R. Strong
Mammalian Central Neurons
Rafael Luján and Carolina Aguado Part IV: Parkinson’s Disease
Nicotine and Nicotinic Receptor Drugs: Potential
GIRK Channel Plasticity and Implications for
for Parkinson’s Disease and Drug-Induced Move-
Drug Addiction
ment Disorders
Ezequiel Marron Fernandez de Velasco,
Maryka Quik, Tanuja Bordia, Danhui Zhang, and
Nora McCall, and Kevin Wickman
Xiomara A. Perez
GIRK Channels: A Potential Link Between
Part V: Alzheimer’s Disease
Learning and Addiction
Nicotinic Cholinergic Mechanisms in Alzheimer’s
Megan E. Tipps and Kari J. Buck
Disease
Behavioral and Genetic Evidence for GIRK Jianxin Shen and Jie Wu
Channels in the CNS: Role in Physiology, Path-
INDEX
ophysiology, and Drug Addiction
Jody Mayfield, Yuri A. Blednov, and R. Adron
Harris
INDEX Volume 125
The Endocannabinoid Signaling System in the
CNS: A Primer
Volume 124 Cecilia J. Hillard
Evidence for a Role of Adolescent Endo-
Part I: Introductory Chapter
cannabinoid Signaling in Regulating HPA Axis
Neuronal Nicotinic Acetylcholine Receptor Stress Responsivity and Emotional Behavior
Structure and Function and Response to Nicotine Development
John A. Dani Tiffany T.-Y. Lee and Boris B. Gorzalka
Part II: Schizophrenia The Endocannabinoid System and Its Role in
The Role of Nicotine in Schizophrenia Regulating the Intrinsic Neural Circuitry of the
Robert E. Featherstone and Steven J. Siegel Gastrointestinal Tract
Samantha M. Trautmann and Keith A. Sharkey
Neuronal α7 Nicotinic Receptors as a Target for
the Treatment of Schizophrenia Endocannabinoid Mechanisms Influencing Nausea
Tanya L. Wallace and Daniel Bertrand Martin A. Sticht, Erin M. Rock, Cheryl L.
Limebeer, and Linda A. Parker
Role of the Neuregulin Signaling Pathway in
Nicotine Dependence and Co-morbid Disorders Endocannabinoid Regulation of Neuroendocrine
Miranda L. Fisher, Anu Loukola, Jaakko Kaprio, Systems
and Jill R. Turner Jeffrey G. Tasker, Chun Chen, Marc O. Fisher,
Xin Fu, Jennifer R. Rainville, and Grant L. Weiss
Effective Cessation Strategies for Smokers with
Schizophrenia The Role of the Brain’s Endocannabinoid System
A. Eden Evins and Corinne Cather in Pain and Its Modulation by Stress
Louise Corcoran, Michelle Roche, and David P. Finn
Part III: Mood Disorders
Role of the Brain’s Reward Circuitry in Depres- Endocannabinoid Signaling in Motivation,
sion: Transcriptional Mechanisms Reward, and Addiction: Influences on
Eric J. Nestler Mesocorticolimbic Dopamine Function
Claudia Sagheddu, Anna Lisa Muntoni, Marco
Nicotine Addiction and Psychiatric Disorders
Pistis, and Miriam Melis
Munir Gunes Kutlu, Vinay Parikh, and
Thomas J. Gould INDEX
Contents of Recent Volumes 375