Programmed Cell Death in Aging
Programmed Cell Death in Aging
Author manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Author Manuscript
Abstract
Programmed cell death (PCD) pathways, including apoptosis and regulated necrosis, are required
Author Manuscript
for normal cell turnover and tissue homeostasis. Mis-regulation of PCD is increasingly implicated
in aging and aging-related disease. During aging the cell turnover rate declines for several highly-
mitotic tissues. Aging-associated disruptions in systemic and inter-cell signaling combined with
cell-autonomous damage and mitochondrial malfunction result in increased PCD in some cell
types, and decreased PCD in other cell types. Increased PCD during aging is implicated in
immune system decline, skeletal muscle wasting (sarcopenia), loss of cells in the heart, and
neurodegenerative disease. In contrast, cancer cells and senescent cells are resistant to PCD,
enabling them to increase in abundance during aging. PCD pathways limit life span in fungi, but
whether PCD pathways normally limit adult metazoan life span is not yet clear. PCD is regulated
by a balance of negative and positive factors, including the mitochondria, which are particularly
subject to aging-associated malfunction.
Author Manuscript
Keywords
apoptosis; necrosis; aging; sarcopenia; mitochondria; senescence
The canonical form of PCD is apoptosis(Elmore, 2007; Kerr et al., 1972), sometimes called
type I PCD, in which the cell undergoes a characteristic series of molecular, biochemical and
Address for correspondence: John Tower, PhD 1050 Childs Way, RRI 210 Los Angeles, CA 90089-2910 Phone: 213-740-5384
[email protected].
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Tower Page 2
mitochondria by activating “Bcl-2 family” members Bad and Bax to form a pore structure in
the mitochondrial membrane(Westphal et al., 2014). The anti-apoptotic “Bcl-2 family”
proteins(e.g., Bcl-2, Bcl-x and Bcl-xL) can bind to the pro-apoptotic proteins to inhibit pore
formation. In normal cells the mitochondria undergo dynamic changes in morphology
regulated in part by the fission factor Drp1(Otera et al., 2013). During apoptosis Drp1 directs
dramatic fission that is required formitochondrial membrane permeabilization (Frank et al.,
2001). In addition, the cell cytoplasm also contains one or more IAPs (inhibitor of apoptosis
proteins), which are a conserved family of caspase inhibitors. Recent studies in mice reveal
that in vivo inhibition of caspase activity does not prevent cell death, but rather shifts the
mechanism towards a more necrotic phenotype (Galluzzi et al., 2015).
Apoptosis is initiated in response to death signals that can be either intrinsic or extrinsic.
Author Manuscript
The intrinsic apoptosis pathway is promoted by cellular stresses including DNA damage,
activated oncogenes, hypoxia, oxidative stress and irradiation. These stimuli shift the
balance of cytoplasmic activities to favor the pro-apoptotic factors by altering protein
expression and/or stability, resulting in mitochondrial membrane permeabilization and the
release of mitochondrial pro-apoptotic factors into the cytoplasm (Figure 1). These
mitochondrial factors include cytochrome c, Smac/DIABLO and the HtrA2/Omi serine
protease. The cytochrome c binds and activates the cytoplasmic proteins Apaf-1 and
procaspase-9. Together these proteins form a structure called the apoptosome, which in turn
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 3
leads to cleavage of procaspase-9 to form the active caspase-9 protease. Smac/DIABLO and
Author Manuscript
HtrA2/Omi inactivate specific IAPs, thereby further tipping the balance towards apoptosis.
Subsequently another group of proteins is released from the mitochondria, including AIF,
endonuclease G (EndoG) and CAD, which act in the nucleus to promote DNA fragmentation
and nuclear condensation. As discussed below, AIF and EndoG also participate in several
forms of caspase-independent PCD. In many cell types the intrinsic pathway can be
activated by the withdrawal of specific hormones that normally act to suppress the apoptotic
pathway(Raff et al., 1994).
The extrinsic pathway for activation of apoptosis involves signaling through transmembrane
receptors of the tumor necrosis factor (TNF) receptor family. These receptors share an
intracellular protein domain involved in signaling called the “death domain”, and are
sometime called “death receptors”. For example, in cells that express TNFR1 (TNF receptor
1), the hormone TNF-alpha activates the receptor and downstream signaling resulting in
Author Manuscript
In certain cell types the initiation of PCD is associated with up-regulation of cell cycle
markers, leading to the suggestion that PCD in these cells may involve a partial entry into
the cell cycle. For example, serum withdrawal from quiescent cultures of mouse 3T3 cells
caused apoptosis associated with up-regulation of G1 phase proteins c-myc, c-jun, c-fos and
cdc-2, as well as BrdU incorporation and PCNA expression, indicative of an abortive G1
traverse(Pandey and Wang, 1995). Treatment of transformed cultured lymphoid cells with
glucocorticoids causes G1 arrest and apoptosis, implicating G1 regulators in both
processes(King and Cidlowski, 1995, 1998). These early studies were with cultured cells,
however as mentioned below, maladaptive cell-cycle entry is also implicated in neuronal
PCD in vivo.
Author Manuscript
The necrosis pathway for cell death is characterized by cell swelling and disruptions of the
cell membrane(Proskuryakov and Gabai, 2010). The release of cytoplasm into the interstitial
space typically results in inflammation. Necrosis was traditionally thought to be an energy-
independent toxic process where the cell is passively destroyed. For example, in many cell
types moderate levels of heat or radiation stress will induce apoptosis, whereas extreme heat
or radiation will cause necrosis. However, recent studies suggest that except for under the
most extreme stress conditions, necrosis is also a regulated process that is activated by
specific pathological and physiological stimuli, and is sometimes referred to as type III
PCD, programmed necrosis, regulated necrosis, or “necroptosis”(Degterev et al., 2014).
Programmed necrosis is caspase-independent and can be further subdivided depending on
the involvement of specific factors. For example, in certain cell types stresses including
Author Manuscript
ischemia, hypoxia, and signaling through TNF-receptor family members can induce a
necroptosis pathway involving the RIP1 kinase, the JNK signaling pathway and the
mitochondria, ultimately leading to cell membrane disruption (Galluzzi et al., 2009;
Pasparakis and Vandenabeele, 2015). Several forms of programmed necrosis involve
cleavage of mitochondrial AIF by cysteine proteases such as calpain and/or cathepsin,
followed by release of the AIF from permeabilized mitochondria. The AIF then undergoes
translocation to the nucleus, and this translocation is positively regulated by cyclophilin A
and negatively regulated by Hsp70 (Delavallee et al., 2011; Gurbuxani et al., 2003; Zhu et
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 4
al., 2007). In the nucleus the AIF interacts with translocated mitochondrial EndoG to
Author Manuscript
mediate nuclear DNA fragmentation. Ongoing studies of cell death reveal that there are
multiple types of apoptosis and programmed necrosis, and that these mechanisms can
overlap depending on the cell type and the nature of the stress.
Another type of programmed cell death is autophagic cell death, sometimes called type II
PCD(Galluzzi et al., 2012; Kroemer et al., 2009). Macroautophagy is a cellular “self-eating”
process in which bulk cytoplasm and organelles become sequestered inside double-
membrane vesicles called autophagosomes. The autophagosomes in turn fuse with
lysosomes to promote degradation of the cellular materials. Autophagic cell death is
characterized by the absence of chromatin condensation and by extensive autophagic
vacuolization of the cytoplasm. Notably, macroautophagy is typically a protective and anti-
apoptotic mechanism, and therefore autophagic cell death may involve hyper-activation of a
normally beneficial process.
Author Manuscript
Mechanisms of PCD can be further divided into sub-types based on a variety of criteria,
including caspase-dependence and the degree of overlap with necrosis (Galluzzi et al.,
2012). For example, “aponecrosis” shares features of both apoptosis and necrosis, and may
represent a midpoint in a continuum between the two forms of cell death (Papucci et al.,
2004; Yakovlev and Faden, 2004). “Paraptosis” is caspase-independent and characterized by
cytoplasmic vacuolization and the requirement for gene transcription and translation.
Paraptosis can be induced by IGF-I signaling and other factors in cultured mammalian cells,
and may also occur in vivo (Sperandio et al., 2004). “Anoikis” refers to apoptotic PCD that
is induced upon detachment of the cell from the extracellular matrix (Paoli et al., 2013).
Anoikis can proceed through either the intrinsic or extrinsic apoptosis pathway depending on
the cell type, and importantly, resistance to anoikis is a hallmark of metastatic cancers
Author Manuscript
(Taddei et al., 2012; Zhong and Rescorla, 2012). The fact that most (and perhaps all) normal
cells are dependent on circulating factors and/or ECM contacts to prevent PCD may be one
mechanism that promotes multi-cellularity. Finally, “Cornification” is a specialized form of
PCD that occurs in the keratinocytes in the epidermis and that involves caspase-14(Eckhart
et al., 2013; Galluzzi et al., 2012). In cornification the dead cell material is maintained to
form the outermost layer of the skin, and is ultimately shed through desquamation.
The details of the apoptotic pathway differ between mammals and the model organisms
Drosophila and C. elegans, however the major players are largely conserved (Figure 1). In
both Drosophila and C. elegans the Bcl-2 family members regulate apoptosome formation
and caspase activation (Denton et al., 2013; Estaquier et al., 2012). In C. elegans the
apoptotic pathway was first characterized by its role in the programmed elimination of
Author Manuscript
specific somatic cells during development (Metzstein et al., 1998). The pro-apoptotic Bcl-2-
related protein EGL-1 inhibits the activity of the anti-apoptotic Bcl-2-related factor CED-9
(Figure 1). CED-9 inhibits the activity of the Apaf-1-related factor CED-4, which in turn
activates the caspase CED-3, which is related to mammalian caspase 3. EGL-1 and CED-3
are implicated in promoting mitochondrial fission and causing release of pro-apoptotic
factors AIF and EndoG from the mitochondria (Parrish et al., 2001; Wang et al., 2002).
Since then additional types of C. elegans developmental cell deaths have been identified
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 5
(Kinet and Shaham, 2014); for example, cell deaths in the female germ line and during male
Author Manuscript
tale development are EGL-1 independent(Gumienny et al., 1999; Maurer et al., 2007).
cell types, and less active than is optimal in other cell types (Gupta, 2005; Higami and
Shimokawa, 2000; Lu et al., 2012; Salminen et al., 2011; Shen and Tower, 2009; Warner,
1997).
Aging is associated with decreased PCD in several cell types. For example, in humans, aging
caused decreased expression of apoptosis genes and increased expression of senescence-
related genes in adipose mesenchymal stem cells (Alt et al., 2012). Similarly, in mouse, bone
marrow mesenchymal stem cells had decreased expression of both cell cycle and PCD genes
during aging(Wilson et al., 2010). A decrease in PCD response with age was observed in the
liver of rats challenged with the DNA-damaging agent methyl methanesulfonate (MMS)
(Suh et al., 2002), and this reduced PCD was associated with increased phosphorylation of
ERKs (extracellular signal-regulated protein kinases) in the old animal tissues (Suh, 2002).
Author Manuscript
Radiation challenge will induce apoptosis in isolated peripheral blood lymphocytes, and this
response is significantly reduced in cells isolated from aged mice (Polyak et al., 1997), as
well as cells isolated from older human patients (Camplejohn et al., 2003). Consistent with
these observations, human serum shows a reduction in markers of apoptosis during normal
aging (Kavathia et al., 2009).
In humans vascular injury can lead to neointima (scar) formation. This response is greater in
older patients due in part to the excessive proliferation and reduced apoptosis of vascular
smooth muscle cells (VSMCs), and the same result is observed in aging mice (Vazquez-
Padron et al., 2004). Interestingly, the reduced apoptosis in the VSMCs during aging may
involve reduced signaling from adjacent endothelial cells (Qian et al., 2011).
Correctly regulated PCD and phagocytosis of the resulting cell debris is required to prevent
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 6
Some cell types are characterized by increased PCD during aging. Apoptosis plays a critical
Author Manuscript
role in the development of the immune system, as well as in adult immune homeostasis.
Apoptosis functions in the selection of T-cells in the thymus, and the deletion of self-reactive
T and B cells. In addition apoptosis is required to regulate immunological memory and to
reduce the number of effector T cells after an immune response. Finally, natural killer cells
and cytotoxic T lymphocytes kill target cells by inducing apoptosis, for example, virus-
infected target cells. During aging there is a significant loss of cells in the thymus (thymus
involution) and bone marrow, associated with an age-related increase in the number of
apoptotic lymphocytes (Sainz et al., 2003). Increased apoptosis is implicated in the age-
related reduction of T-cell subtypes in the human immune system (T-cell lymphopenia),
including both CD4+ and CD8+ T-cells (Gupta, 2005). In contrast, in vitro studies report
that certain T-cell subtypes (CD8+ CD28−) resulting from repeated immune stimulation
exhibit senescence-like changes that include reduced response to stress and resistance to
Author Manuscript
apoptosis(Chou and Effros, 2013). Aging in the mammalian heart is characterized by a loss
of myocytes and hypertrophy of the remaining myocytes (Olivetti et al., 1991). Both
apoptotic and necrotic mechanisms are implicated in myocyte cell loss during normal heart
aging as well as in heart failure (Kajstura et al., 1996; Kung et al., 2011).
Apoptosis is critical in wound healing, and wound healing is generally impaired during
aging, often associated with local inflammation (Hiebert and Granville, 2012; Rai et al.,
2005). For example, mis-regulated apoptosis has been implicated in the age-associated
increase in fibrotic lung disease in two ways: increased sensitivity to apoptosis in alveolar
epithelial cells may lead to greater injury, whereas reduced sensitivity to apoptosis in
fibroblasts and myofibroblasts may promote fibrosis (Kapetanaki et al., 2013).
cell-autonomous effects result in increased susceptibility to PCD in some cell types, and
decreased PCD in other cell types. One possible interpretation of these results is that the
general decrease in PCD markers during aging may result in part from a slowing of the
normal homeostatic cell turnover rate for several adult tissues. For example, dramatically
decreased cell turnover rate with age is observed for the skin (Rinnerthaler et al., 2014), and
reduced cell turnover rates with age are also reported for the heart (Bergmann et al., 2009)
breast epithelium (Misell et al., 2005) and pituitary (Nolan et al., 1999; Richardson et al.,
2014). At the same time, alterations in systemic signaling, including inflammation and ECM
disruptions, may combine with cell-autonomous changes such as increased oxidative stress
and genomic damage to make certain cell types more sensitive to PCD, particularly when
challenged by acute or chronic stress.
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 7
and the release of the mitochondrial pro-apoptotic factors(Green and Kroemer, 2009). The
Author Manuscript
al., 1999), and this may be related to the increased cancer incidence observed in those
patients. Most cancer chemotherapies, including ionizing radiation, are thought to act by
hyper-stimulating the otherwise suppressed PCD pathways, including activating any
remaining p53 (Stegh, 2012; Zhao et al., 2012). Consistent with this idea, the efficiency of
induction of apoptosis by chemotherapy has been shown to correlate with clinical outcomes
for several cancers (Bosserman et al., 2012). However, it should be noted that other studies
suggest that, at least for certain cell types, the tumor-suppressive function of p53 is
independent of transcriptional induction of apoptosis, and instead may involve p53
regulation of DNA repair and/or metabolism(Valente et al., 2013). Finally a promising
therapy for cancer is treatment with TRAIL (tumor necrosis factor-related apoptosis-
inducing ligand). TRAIL is a naturally-occurring cytokine that binds to the DR4 and DR5
death receptors to activate the extrinsic apoptosis pathway, and it does this preferentially in
cancer cells (Refaat et al., 2014).
Author Manuscript
associated inflammation and disruptions of ECM. The resistance to apoptosis has been
examined in senescent human fibroblasts in vitro. These cells fail to reduce Bcl-2 levels
upon apoptotic stimuli including serum starvation (Wang, 1995, 1997)and oxidative stress
(Ryu et al., 2007; Yeo et al., 2000), and have reduced caspase-3 expression(Marcotte et al.,
2004) and increased expression of the anti-apoptotic factor Bcl-xL (Rochette and Brash,
2008). The resistance of senescent cells to apoptosis might also involve the down-regulation
of cell cycle factors that participate in both cell division and in cell death (King and
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 8
Cidlowski, 1995). Recently both genetic and chemical interventions have been described
Author Manuscript
that can preferentially kill senescent cells in vivo in mice, and these interventions had health
benefits for mouse models with elevated levels of senescent cells (Zhu et al., 2015). Despite
the potential negative effects of increasing the abundance of senescent cells, cellular
senescence has emerged as an important therapeutic target in cancer(Lee and Lee, 2014).
Cancer chemotherapy and ionizing radiation can cause either apoptosis or cellular
senescence depending on the cell type, and both outcomes have been shown to positively
correlate with clinical outcomes.
Rapp and Gallagher, 1996). However, PCD is increasingly implicated in several aging-
associated neurodegenerative diseases(Ghavami et al., 2014). Nervous tissue has several
properties that may contribute to a susceptibility to maladaptive PCD. For example, in the
normal adult brain, activated caspase-3 functions in the remodeling of neuronal processes
important for synaptic plasticity and certain forms of memory (Snigdha et al., 2012). The
presence of activated caspase-3 may create a risk for inappropriate PCD activation in
neurons challenged by stress. Moreover, neural tissue is rich in iron, and iron levels have
been observed to further increase with age and in neurodegenerative disease(Oshiro et al.,
2011). Iron can promote Fenton chemistry and oxidative stress, thereby favoring PCD.
Neurons appear to suppress PCD in part by using glucose metabolism to generate reducing
equivalents that prevent oxidation and activation of cytochrome c (Vaughn and Deshmukh,
2008). This may make neurons particularly susceptible to PCD upon metabolic disruption.
Author Manuscript
It is not yet clear what are the precise mechanism(s) for cell death in the aging-associated
neurodegenerative diseases. Increased or altered p53 activity is found to correlate with AD
(Lanni et al., 2012), and animal model studies implicate a PARP-dependent programmed-
Author Manuscript
necrosis pathway in PD (Lee et al., 2014). Cell cycle re-entry may be particularly important
(Folch et al., 2012). Brain tissue from neurodegenerative disease patients shows extensive
up-regulation of cell cycle proteins including CDKs. Studies of cultured neuronal cells
indicate that oxidative stress and DNA damage can induce an abortive cell cycle entry and
consequent apoptosis, suggesting that this might also occur in vivo. Taken together, these
studies suggest that only a sub-set of neurons are at risk for loss by PCD-like events during
aging.
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 9
During aging several tissues exhibit cell loss that is attributed to PCD or to PCD-like
processes. Mammals exhibit an aging-associated skeletal muscle atrophy called sarcopenia
that involves both reductions in muscle fiber size and loss of fibers(Marzetti et al., 2008).
The precise mechanisms for sarcopenia are not yet known, however studies in rodents
implicate mitochondrial malfunction and abnormal PCD signaling (Marzetti et al., 2013).
The expression of PCD markers correlates with aging and sarcopenia in specific rodent
muscle tissues. For example, aging rat gastrocnemius muscle showed increased levels of
Bax, decreased Bcl-2, activation of caspase-3 and DNA fragmentation (Song et al., 2006). In
contrast, aging rat soleus muscle showed translocation of Endonuclease G from the
mitochondria to the nucleus suggesting a caspase-independent process (Leeuwenburgh et al.,
2005). Mitochondrial mutations are implicated in sarcopenia because mtDNA deletions co-
localized with mitochondrial malfunction in skeletal muscle fibers of aged rats (Wanagat et
Author Manuscript
al., 2001), and PCD-like events were elevated in the skeletal muscle of aging mice
engineered to have greater mitochondrial mutation load (Kujoth et al., 2005). Age-related
alterations in systemic signaling are also implicated in muscle aging, based on the results of
several striking interventions. For example, fusion of young and old mouse circulatory
systems promoted the proliferative and regenerative capacity of skeletal muscle in the old
mouse (Conboy et al., 2005). Old mice exhibit a decrease in circulating levels of the growth
hormone GDF-11, and restoring GDF-11 levels in the old mouse was sufficient for much of
the benefit for muscle function (Sinha et al., 2014). Loss of myocytes is observed in the
aging heart, and studies in rodent models implicate a PCD mechanism involving increased
Bax/Bcl-2 ratio, cytochrome c release from mitochondria and Caspase-3 activation (Kwak,
2013). In mice, over-expression of the mitochondrial antioxidant enzyme MnSOD reduced
PCD in the aging heart, thereby implicating mitochondrial function and oxidative stress
Author Manuscript
(Kwak et al., 2014). In addition, mutation of mouse cathepsin K reduced AIG translocation
and PCD during aging, resulting in improved heart function and implicating both caspase-
dependent and caspase-independent mechanisms(Hua et al., 2015). Finally, increasing
GDF-11 levels in the aging mouse had a benefit for heart function, indicating a role for
altered systemic signaling in heart aging.
Several additional tissues show increased sensitivity to induction of PCD during human
aging. For example, the aging kidney has increased sensitivity to stress and PCD, and rodent
models show decreased levels of Bcl-2, and increases in Bax, cytochrome c and caspase-3
(Wang et al., 2014). Age-related hearing loss involves loss of sensory hair cells via PCD, and
mitochondrial genome mutations are implicated in this process(Fujimoto and Yamasoba,
2014). Aging-associated hair greying involves loss of melanocyte stem cells by PCD, and
Author Manuscript
decreased Bcl-2 and increased ROS are implicated in this process (Seiberg, 2013). Finally,
increased sensitivity to stress and PCD is observed in aging gastric mucosa, and rodent
studies implicate caspase-3 activation (Tarnawski et al., 2014).
In the model organism Drosophila, several studies indicate altered PCD regulation during
aging. Increased PCD-like events were observed during aging in muscle tissue and in fat
tissue, as indicated by DNA fragmentation and caspase activity (Zheng et al., 2005). In
contrast there was no evidence for a global increase in PCD in Drosophila nervous tissue
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 10
using these assays. However, a transgenic reporter for caspase-3 activity was activated
Author Manuscript
during aging in a subset of nervous tissue cell types, including the ellipsoid body that
functions in visual memory, and the antennal lobe and a sub-set of olfactory receptor
neurons involved in olfaction(Chihara et al., 2014). The olfactory receptor neurons also had
increased PARP levels and DNA fragmentation, and were sometimes lost through PCD.
These studies suggest that only a specific sub-set of neurons are at risk for PCD during aging
in Drosophila.
related gene) mutants, suggesting an aging-associated loss of normal germ-line PCD activity
(Andux and Ellis, 2008; McGee et al., 2012). Taken together the studies with the model
systems support the conclusion that aging can cause increased susceptibility to apoptosis in
some cell types, and decreased apoptosis in other cell types.
reported not to affect adult life span, suggesting that C. elegans life span is not regulated by
a canonical caspase-dependent PCD(Garigan et al., 2002), however a possible role for
caspase-independent PCD or necrosis pathways in regulating C. elegans life span cannot yet
be ruled out. Intriguingly, the canonical caspase-dependent C. elegans PCD pathway was
recently found to be required for increased life span caused by ETC mutants and retrograde
ROS signaling (Yee et al., 2014), in the absence of overt PCD. The data suggest a potentially
ancient function for the pathway in mediating retrograde mitochondrial ROS signaling and
altered nuclear gene expression independent of its function in regulating PCD.
In Drosophila, both the gut (Jiang et al., 2011; Micchelli and Perrimon, 2006; Ohlstein and
Spradling, 2006) and Malphigian tubules (equivalent to kidney nephrons) (Singh et al.,
2007)are maintained by stem cell division and cell turnover, however it has not yet been
determined if the rate of cell turnover is important for normal longevity. Over-expression of
Author Manuscript
caspase inhibitors p35 and DIAP1 in adult flies did not affect life span, suggesting that adult
fly life span is not limited by a canonical caspase-dependent PCD(Shen et al., 2009),
however a possible role for caspase-independent PCD or necrosis pathways in regulating
adult fly life span cannot yet be ruled out.
In mammals PCD is required for normal development and adult tissue homeostasis, which
complicates investigation of possible roles for PCD in mammalian life span regulation.
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 11
Drugs that inhibit PCD and necrosis show promise as potential interventions in injury and
Author Manuscript
disease (Degterev et al., 2014), however as yet there are no reports of benefits for normal
longevity.
were found to promote translation and growth and suppress longevity in response to
nutrients (Powers et al., 2006). Mitochondrial stress and retrograde signaling can also affect
the Ras/PKA pathway and increase life span, depending on the yeast genotype (Jazwinski,
2014; Kirchman et al., 1999). Aged mother cells showed characteristics of PCD including
increased markers of ROS, nuclear DNA fragmentation as indicated by TUNEL labeling,
and phosphatidylserine externalization as indicated by Annexin V staining (Laun et al.,
2001).
The other approach for studying aging in Saccharomyces cerevisiae is the chronological
aging assay. Here the yeast culture is grown until glucose is depleted, which causes the cells
to withdraw from the cell cycle (Fabrizio and Longo, 2003). The cells remain metabolically
active and the number of viable cells in the sample can be measured at various time points
Author Manuscript
by plating the cells on a more rich media and counting the number of colonies. As a function
of time the number of viable cells in the sample decreases as cells are lost to lysis. The
pathways thought to regulate life span in the chronological aging assay include Tor/Sch9 and
Ras/PKA (Wei et al., 2008). Cell death in the Saccharomyces cerevisiae chronological aging
assay has also been described as a PCD based on both morphological and genetic criteria
(Fabrizio et al., 2004; Herker et al., 2004). The morphological criteria included chromatin
condensation and fragmentation, increased markers of ROS, DNA fragmentation indicated
by TUNEL, and phosphatidylserine externalization indicated by Annexin V staining
(Carmona-Gutierrez et al., 2010a).
Several genes have been identified that function in yeast PCD similarly to PCD regulators in
mammals (Figure 1)(Carmona-Gutierrez et al., 2010a). A protease related to caspases called
a metacaspase plays a role in yeast PCD(Hill and Nystrom, 2015). Deletion of the
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 12
PCD (Buttner et al., 2007). Notably the PCD mediated by NUC1 did not require the
Author Manuscript
metacaspase YCA1. An IAP-like protein called BIR1 was identified, and over-expression of
BIR1 could both reduce PCD and extend chronological life span (Walter et al., 2006). BIR1
is in turn regulated by a yeast homolog of the mammalian serine protease HtrA2/Omi, called
NMA111(Figure 1)(Fahrenkrog, 2011; Fahrenkrog et al., 2004); however, unlike
mammalian HtrA2/Omi, which is released from the mitochondria, the yeast NMA111
functions in the nucleus. So far no interaction between BIR1 and YCA1 has been detected
(Walter et al., 2006). The specific molecular activator(s) of YCA1 during PCD remain to be
identified (Carmona-Gutierrez et al., 2010a; Carmona-Gutierrez et al., 2010b; Hill and
Nystrom, 2015), however Cytochrome c has been implicated(Silva et al., 2005).
Mitochondrial fission appears to play a role in yeast PDC similar to the situation in
mammals. The gene DNM1 promoted mitochondrial fission and PCD in response to
hydrogen peroxide, whereas the conserved mitochondrial morphology regulator FIS1
Author Manuscript
inhibited fission and YCA1-dependent cell death (Fannjiang et al., 2004). Notably deletion
of DNM1 also increased replicative life span (Scheckhuber et al., 2007). Interestingly,
human Bcl-2 could substitute for FIS1, consistent with a Bcl-2-like function for FIS1.
Finally, a BH3-only protein called YBH3 was identified (Buttner et al., 2011). Upon toxic
stress YBH3 associated with the mitochondria and disrupted mitochondrial membrane
potential. Over-expression of YBH3 sensitized cells to PCD caused by hydrogen peroxide,
however this death was not dependent upon YCA1, AIF1, NMA111 or NUC1. Deletion of
YBH3 was reported to increase both replicative and chronological life span (Buttner et al.,
2011). The studies with yeast therefore indicate multiple mechanisms for PCD involving
factors released from the mitochondria, similar to the situation in mammals.
Sexual reproduction is also associated with PCD in Saccharomyces cerevisiae. Mating type
Author Manuscript
pheromone can induce PCD in Saccharomyces cerevisiae haploid cells of the corresponding
mating type that fail to mate (Severin and Hyman, 2002); low levels of hormone promote
mating whereas high levels promote PCD. The mating type pheromone binds to its receptor
(either Ste2p or Ste3p) and activates a MAPK signaling pathway involving the Ste20p
kinase. There follows an increase in cytoplasmic Ca++ levels, increased mitochondrial
activity and ROS production, mitochondrial fission dependent upon mitochondrial protein
Ysp1p, and finally cytochrome c release and cell death (Pozniakovsky et al., 2005).
Cytochrome c release is also implicated in PCD caused by acetic acid (Ludovico et al.,
2002). PCD can also occur during meiosis of diploid yeast cells, where it is regulated by
histone H2B phosphorylation (Ahn et al., 2006; Ahn et al., 2005). It is interesting to note
that hormones involved in sexual differentiation in mammals, such as testosterone and
estrogen, are also regulators of MAPK signaling and PCD (Vasconsuelo et al., 2011), and
Author Manuscript
sexual differentiation is associated with aging in several species (Tower, 2006, 2014),
suggesting possible conservation of mechanisms.
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 13
The fungus Podospora anserina has been a pioneering model for the study of aging
(Osiewacz and Borghouts, 2000). For several genotypes the culture will grow for 3-5 weeks
and then enter senescence marked by mitochondrial DNA instabilities, increased ROS,
mitochondrial fission, accumulation of pigment and cell death (Gredilla et al., 2006;
Osiewacz et al., 2010). Genetic studies indicate that PCD factors conserved with
Saccharomyces cerevisiae and with mammals play a role in Podospora anserina aging.
Deletion of the conserved mitochondrial fission regulator PaDnm1 dramatically extended
Podospora anserina life span (Scheckhuber et al., 2007). Deletion of the metacaspase gene
PaMca1 caused increased life span, indicative a role for metacaspase-dependent PCD. In
addition, deletion of mitochondrial AIF-like factors PaAIF2 and PaAMID2 caused increased
resistance to oxidative stress and increased life span independent of metacaspase activity,
Author Manuscript
Taken together the studies in fungi suggest conservation of several aspects of PCD with
mammals, including a critical regulatory role for the mitochondria. Interestingly, fungal
PCD is regulated by sex hormones and during gametogenesis, similar to the situation in
Author Manuscript
flies, nematodes and mammals. In the fungi PCD appears to limit life span in both
replicative and chronological assays. The pro-apoptotic role of ROS, mitochondrial DNA
rearrangements and release of pro-apoptotic factors from permeabilized mitochondria
suggests that a failure in mitochondrial maintenance may contribute to the increased
sensitivity to PCD during fungal aging(Bernhardt et al., 2014), similar to the situation in
mammals.
ECM and cell-cell signals, thereby providing many potential targets for aging-associated
disruptions. Second, the normal regulation of PCD involves a delicate balance of multiple
pro-PCD and anti-PCD factors, and cells appear poised for PCD upon appropriate signaling.
This may make PCD pathways particularly vulnerable to maladaptive signals during aging.
Third, the important role of the mitochondria and ROS in PCD is likely a factor, because
mitochondria appear to be particularly susceptible to damage and malfunction during aging
across species (Bernhardt et al., 2014; Hur et al., 2014; Wallace, 2010).
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 14
Several aspects of the observed mis-regulation of PCD during aging support the idea that
Author Manuscript
and membrane permeabilization that promote PCD, leading to increased PCD in cell types
such as the muscle. Decreased mitophagy will result in longer-lived and more damage-prone
mitochondria, and decreased mitophagy is associated with aging across multiple species
(Gelino and Hansen, 2012; Liu et al., 2014; Richard et al., 2013). The mitochondrial genome
has a high mutation rate, and mitochondrial mutations are increasingly implicated in
mitochondrial malfunction during aging(Schon et al., 2012; Sevini et al., 2014). Because
mitophagy is important in clearing mutated mitochondrial genomes from the cell, the
decrease in mitophagy with age may be involved in the accumulation of mutated
mitochondrial genomes that favor mitochondrial malfunction, including maladaptive
PCD(Tower, 2014). Finally, the uni-parental inheritance of mitochondrial genomes may
create genetic conflicts (sexual antagonistic pleiotropy) that cause mitochondrial functions,
including PCD, to be particularly susceptible to aging-associated disruptions, including sex-
biased mis-regulation(Maklakov and Lummaa, 2013; Tower, 2006, 2014).
Author Manuscript
Acknowledgements
This work was supported by a grant from the Department of Health and Human Services (AG011833) and by pilot
project funds from the Southern California Environmental Health Sciences Center (5P30ES007048).
References
Ahn SH, Diaz RL, Grunstein M, Allis CD. Histone H2B deacetylation at lysine 11 is required for yeast
apoptosis induced by phosphorylation of H2B at serine 10. Mol Cell. 2006; 24:211–220. [PubMed:
17052455]
Ahn SH, Henderson KA, Keeney S, Allis CD. H2B (Ser10) phosphorylation is induced during
apoptosis and meiosis in S. cerevisiae. Cell Cycle. 2005; 4:780–783. [PubMed: 15970663]
Alt EU, Senst C, Murthy SN, Slakey DP, Dupin CL, Chaffin AE, Kadowitz PJ, Izadpanah R. Aging
Author Manuscript
alters tissue resident mesenchymal stem cell properties. Stem cell research. 2012; 8:215–225.
[PubMed: 22265741]
Andersen BB, Gundersen HJ, Pakkenberg B. Aging of the human cerebellum: a stereological study.
The Journal of comparative neurology. 2003; 466:356–365. [PubMed: 14556293]
Andux S, Ellis RE. Apoptosis maintains oocyte quality in aging Caenorhabditis elegans females. PLoS
Genet. 2008; 4:e1000295. [PubMed: 19057674]
Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabe-Heider F, Walsh S, Zupicich J, Alkass K,
Buchholz BA, Druid H, Jovinge S, Frisen J. Evidence for cardiomyocyte renewal in humans.
Science. 2009; 324:98–102. [PubMed: 19342590]
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 15
Bernhardt D, Hamann A, Osiewacz HD. The role of mitochondria in fungal aging. Current opinion in
microbiology. 2014; 22C:1–7. [PubMed: 25299751]
Author Manuscript
Kollroser M, Frohlich KU, Sigrist S, Madeo F. Endonuclease G regulates budding yeast life and
death. Mol Cell. 2007; 25:233–246. [PubMed: 17244531]
Buttner S, Ruli D, Vogtle FN, Galluzzi L, Moitzi B, Eisenberg T, Kepp O, Habernig L, Carmona-
Gutierrez D, Rockenfeller P, Laun P, Breitenbach M, Khoury C, Frohlich KU, Rechberger G,
Meisinger C, Kroemer G, Madeo F. A yeast BH3-only protein mediates the mitochondrial pathway
of apoptosis. EMBO J. 2011; 30:2779–2792. [PubMed: 21673659]
Camplejohn RS, Gilchrist R, Easton D, McKenzie-Edwards E, Barnes DM, Eccles DM, Ardern-Jones
A, Hodgson SV, Duddy PM, Eeles RA. Apoptosis, ageing and cancer susceptibility. British journal
of cancer. 2003; 88:487–490. [PubMed: 12592359]
Carmona-Gutierrez D, Bauer MA, Ring J, Knauer H, Eisenberg T, Buttner S, Ruckenstuhl C,
Reisenbichler A, Magnes C, Rechberger GN, Birner-Gruenberger R, Jungwirth H, Frohlich KU,
Sinner F, Kroemer G, Madeo F. The propeptide of yeast cathepsin D inhibits programmed
necrosis. Cell death & disease. 2011; 2:e161. [PubMed: 21593793]
Carmona-Gutierrez D, Eisenberg T, Buttner S, Meisinger C, Kroemer G, Madeo F. Apoptosis in yeast:
triggers, pathways, subroutines. Cell Death Differ. 2010a; 17:763–773. [PubMed: 20075938]
Author Manuscript
Carmona-Gutierrez D, Frohlich KU, Kroemer G, Madeo F. Metacaspases are caspases. Doubt no more.
Cell Death Differ. 2010b; 17:377–378. [PubMed: 20154701]
Chihara T, Kitabayashi A, Morimoto M, Takeuchi K, Masuyama K, Tonoki A, Davis RL, Wang JW,
Miura M. Caspase inhibition in select olfactory neurons restores innate attraction behavior in aged
Drosophila. PLoS Genet. 2014; 10:e1004437. [PubMed: 24967585]
Childs BG, Baker DJ, Kirkland JL, Campisi J, van Deursen JM. Senescence and apoptosis: dueling or
complementary cell fates? EMBO reports. Oct 13.2014 doi: 10.15252/embr.201439245.
Chimal-Monroy J, Abarca-Buis RF, Cuervo R, Diaz-Hernandez M, Bustamante M, Rios-Flores JA,
Romero-Suarez S, Farrera-Hernandez A. Molecular control of cell differentiation and programmed
cell death during digit development. IUBMB Life. 2011; 63:922–929. [PubMed: 21901820]
Chou JP, Effros RB. T cell replicative senescence in human aging. Curr Pharm Des. 2013; 19:1680–
1698. [PubMed: 23061726]
Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged
progenitor cells by exposure to a young systemic environment. Nature. 2005; 433:760–764.
Author Manuscript
[PubMed: 15716955]
Degterev A, Zhou W, Maki JL, Yuan J. Assays for necroptosis and activity of RIP kinases. Methods in
enzymology. 2014; 545:1–33. [PubMed: 25065884]
Delaney JR, Sutphin GL, Dulken B, Sim S, Kim JR, Robison B, Schleit J, Murakami CJ, Carr D, An
EH, Choi E, Chou A, Fletcher M, Jelic M, Liu B, Lockshon D, Moller RM, Pak DN, Peng Q, Peng
ZJ, Pham KM, Sage M, Solanky A, Steffen KK, Tsuchiya M, Tsuchiyama S, Johnson S, Raabe C,
Suh Y, Zhou Z, Liu X, Kennedy BK, Kaeberlein M. Sir2 deletion prevents lifespan extension in 32
long-lived mutants. Aging Cell. 2011; 10:1089–1091. [PubMed: 21902802]
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 16
21438113]
Denton D, Aung-Htut MT, Kumar S. Developmentally programmed cell death in Drosophila.
Biochimica et biophysica acta. 2013; 1833:3499–3506. [PubMed: 23810935]
Devitt A, Marshall LJ. The innate immune system and the clearance of apoptotic cells. Journal of
leukocyte biology. 2011; 90:447–457. [PubMed: 21562053]
Eckhart L, Lippens S, Tschachler E, Declercq W. Cell death by cornification. Biochimica et biophysica
acta. 2013; 1833:3471–3480. [PubMed: 23792051]
Egilmez NK, Jazwinski SM. Evidence for the involvement of a cytoplasmic factor in the aging of the
yeast Saccharomyces cerevisiae. J Bacteriol. 1989; 171:37–42. [PubMed: 2644196]
Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007; 35:495–516.
[PubMed: 17562483]
Elrod JW, Molkentin JD. Physiologic functions of cyclophilin D and the mitochondrial permeability
transition pore. Circulation journal : official journal of the Japanese Circulation Society. 2013;
77:1111–1122. [PubMed: 23538482]
Author Manuscript
Estaquier J, Vallette F, Vayssiere JL, Mignotte B. The mitochondrial pathways of apoptosis. Adv Exp
Med Biol. 2012; 942:157–183. [PubMed: 22399422]
Fabrizio P, Battistella L, Vardavas R, Gattazzo C, Liou LL, Diaspro A, Dossen JW, Gralla EB, Longo
VD. Superoxide is a mediator of an altruistic aging program in Saccharomyces cerevisiae. J Cell
Biol. 2004; 166:1055–1067. [PubMed: 15452146]
Fabrizio P, Longo VD. The chronological life span of Saccharomyces cerevisiae. Aging Cell. 2003;
2:73–81. [PubMed: 12882320]
Fahrenkrog B. Nma111p, the pro-apoptotic HtrA-like nuclear serine protease in Saccharomyces
cerevisiae: a short survey. Biochemical Society transactions. 2011; 39:1499–1501. [PubMed:
21936841]
Fahrenkrog B, Sauder U, Aebi U. The S. cerevisiae HtrA-like protein Nma111p is a nuclear serine
protease that mediates yeast apoptosis. J Cell Sci. 2004; 117:115–126. [PubMed: 14657274]
Fannjiang Y, Cheng WC, Lee SJ, Qi B, Pevsner J, McCaffery JM, Hill RB, Basanez G, Hardwick JM.
Mitochondrial fission proteins regulate programmed cell death in yeast. Genes Dev. 2004;
Author Manuscript
TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, Di Daniele N, Dixit VM, Dynlacht
BD, El-Deiry WS, Fimia GM, Flavell RA, Fulda S, Garrido C, Gougeon ML, Green DR,
Gronemeyer H, Hajnoczky G, Hardwick JM, Hengartner MO, Ichijo H, Joseph B, Jost PJ,
Kaufmann T, Kepp O, Klionsky DJ, Knight RA, Kumar S, Lemasters JJ, Levine B, Linkermann A,
Lipton SA, Lockshin RA, Lopez-Otin C, Lugli E, Madeo F, Malorni W, Marine JC, Martin SJ,
Martinou JC, Medema JP, Meier P, Melino S, Mizushima N, Moll U, Munoz-Pinedo C, Nunez G,
Oberst A, Panaretakis T, Penninger JM, Peter ME, Piacentini M, Pinton P, Prehn JH, Puthalakath
H, Rabinovich GA, Ravichandran KS, Rizzuto R, Rodrigues CM, Rubinsztein DC, Rudel T, Shi Y,
Simon HU, Stockwell BR, Szabadkai G, Tait SW, Tang HL, Tavernarakis N, Tsujimoto Y, Vanden
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 17
Berghe T, Vandenabeele P, Villunger A, Wagner EF, Walczak H, White E, Wood WG, Yuan J,
Zakeri Z, Zhivotovsky B, Melino G, Kroemer G. Essential versus accessory aspects of cell death:
Author Manuscript
recommendations of the NCCD 2015. Cell Death Differ. 2015; 22:58–73. [PubMed: 25236395]
Galluzzi L, Kepp O, Kroemer G. RIP kinases initiate programmed necrosis. Journal of molecular cell
biology. 2009; 1:8–10. [PubMed: 19679643]
Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM,
Dawson VL, El-Deiry WS, Fulda S, Gottlieb E, Green DR, Hengartner MO, Kepp O, Knight RA,
Kumar S, Lipton SA, Lu X, Madeo F, Malorni W, Mehlen P, Nunez G, Peter ME, Piacentini M,
Rubinsztein DC, Shi Y, Simon HU, Vandenabeele P, White E, Yuan J, Zhivotovsky B, Melino G,
Kroemer G. Molecular definitions of cell death subroutines: recommendations of the
Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012; 19:107–120. [PubMed:
21760595]
Garigan D, Hsu AL, Fraser AG, Kamath RS, Ahringer J, Kenyon C. Genetic analysis of tissue aging in
Caenorhabditis elegans: a role for heat-shock factor and bacterial proliferation. Genetics. 2002;
161:1101–1112. [PubMed: 12136014]
Gelino S, Hansen M. Autophagy - An Emerging Anti-Aging Mechanism. Journal of clinical &
Author Manuscript
Gumienny TL, Lambie E, Hartwieg E, Horvitz HR, Hengartner MO. Genetic control of programmed
cell death in the Caenorhabditis elegans hermaphrodite germline. Development. 1999; 126:1011–
1022. [PubMed: 9927601]
Gunther C, Neumann H, Neurath MF, Becker C. Apoptosis, necrosis and necroptosis: cell death
regulation in the intestinal epithelium. Gut. 2013; 62:1062–1071. [PubMed: 22689519]
Gupta S. Molecular mechanisms of apoptosis in the cells of the immune system in human aging.
Immunological reviews. 2005; 205:114–129. [PubMed: 15882349]
Gupta S, Agrawal A, Agrawal S, Su H, Gollapudi S. A paradox of immunodeficiency and
inflammation in human aging: lessons learned from apoptosis. Immunity & ageing : I & A. 2006;
3:5. [PubMed: 16712718]
Gurbuxani S, Schmitt E, Cande C, Parcellier A, Hammann A, Daugas E, Kouranti I, Spahr C, Pance A,
Kroemer G, Garrido C. Heat shock protein 70 binding inhibits the nuclear import of apoptosis-
inducing factor. Oncogene. 2003; 22:6669–6678. [PubMed: 14555980]
Hankins HM, Baldridge RD, Xu P, Graham TR. Role of flippases, scramblases and transfer proteins in
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 18
Hiebert PR, Granville DJ. Granzyme B in injury, inflammation, and repair. Trends Mol Med. 2012;
18:732–741. [PubMed: 23099058]
Author Manuscript
Higami Y, Shimokawa I. Apoptosis in the aging process. Cell and tissue research. 2000; 301:125–132.
[PubMed: 10928285]
Hill SM, Nystrom T. The dual role of a yeast metacaspase: What doesn't kill you makes you stronger.
Bioessays. Feb 13.2015 doi: 10.1002/bies.201400208.
Hroudova J, Singh N, Fisar Z. Mitochondrial dysfunctions in neurodegenerative diseases: relevance to
Alzheimer's disease. BioMed research international. May 12.2014 2014 :175062. doi:
10.1155/2014/175062. [PubMed: 24900954]
Hua Y, Robinson TJ, Cao Y, Shi GP, Ren J, Nair S. Cathepsin K knockout alleviates aging-induced
cardiac dysfunction. Aging Cell. Feb 18.2015 doi: 10.1111/acel.12276.
Hur JH, Stork DA, Walker DW. Complex-I-ty in aging. Journal of bioenergetics and biomembranes.
2014; 46:329–335. [PubMed: 24961226]
Jazwinski SM. The retrograde response: a conserved compensatory reaction to damage from within
and from without. Progress in molecular biology and translational science. 2014; 127:133–154.
[PubMed: 25149216]
Author Manuscript
Jego G, Hazoume A, Seigneuric R, Garrido C. Targeting heat shock proteins in cancer. Cancer letters.
2013; 332:275–285. [PubMed: 21078542]
Jiang H, Grenley MO, Bravo MJ, Blumhagen RZ, Edgar BA. EGFR/Ras/MAPK signaling mediates
adult midgut epithelial homeostasis and regeneration in Drosophila. Cell Stem Cell. 2011; 8:84–
95. [PubMed: 21167805]
Kajstura J, Cheng W, Sarangarajan R, Li P, Li B, Nitahara JA, Chapnick S, Reiss K, Olivetti G,
Anversa P. Necrotic and apoptotic myocyte cell death in the aging heart of Fischer 344 rats. The
American journal of physiology. 1996; 271:H1215–1228. [PubMed: 8853362]
Kapetanaki MG, Mora AL, Rojas M. Influence of age on wound healing and fibrosis. J Pathol. 2013;
229:310–322. [PubMed: 23124998]
Kavathia N, Jain A, Walston J, Beamer BA, Fedarko NS. Serum markers of apoptosis decrease with
age and cancer stage. Aging (Albany NY). 2009; 1:652–663. [PubMed: 20157546]
Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging
implications in tissue kinetics. British journal of cancer. 1972; 26:239–257. [PubMed: 4561027]
Author Manuscript
Kile BT. The role of apoptosis in megakaryocytes and platelets. British journal of haematology. 2014;
165:217–226. [PubMed: 24467740]
Kinet MJ, Shaham S. Noncanonical cell death in the nematode Caenorhabditis elegans. Methods in
enzymology. 2014; 545:157–180. [PubMed: 25065890]
King KL, Cidlowski JA. Cell cycle and apoptosis: common pathways to life and death. J Cell
Biochem. 1995; 58:175–180. [PubMed: 7673325]
King KL, Cidlowski JA. Cell cycle regulation and apoptosis. Annu Rev Physiol. 1998; 60:601–617.
[PubMed: 9558478]
Kirchman PA, Kim S, Lai CY, Jazwinski SM. Interorganelle signaling is a determinant of longevity in
Saccharomyces cerevisiae. Genetics. 1999; 152:179–190. [PubMed: 10224252]
Kirkland JL, Tchkonia T. Clinical strategies and animal models for developing senolytic agents. Exp
Gerontol. Oct 28.2014 pii: S0531-5565(14)00291-5. doi: 10.1016/j.exger.2014.10.012.
Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV,
El-Deiry WS, Golstein P, Green DR, Hengartner M, Knight RA, Kumar S, Lipton SA, Malorni W,
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 19
Kwak HB. Effects of aging and exercise training on apoptosis in the heart. Journal of exercise
rehabilitation. 2013; 9:212–219. [PubMed: 24278863]
Author Manuscript
Kwak HB, Lee Y, Kim JH, Van Remmen H, Richardson AG, Lawler JM. MnSOD Overexpression
Reduces Fibrosis and Pro-Apoptotic Signaling in the Aging Mouse Heart. J Gerontol A Biol Sci
Med Sci. Jul 11.2014 pii: glu090. doi 10.1093/gerona/glu090.
Lanni C, Racchi M, Memo M, Govoni S, Uberti D. p53 at the crossroads between cancer and
neurodegeneration. Free Radic Biol Med. 2012; 52:1727–1733. [PubMed: 22387179]
Laun P, Pichova A, Madeo F, Fuchs J, Ellinger A, Kohlwein S, Dawes I, Frohlich KU, Breitenbach M.
Aged mother cells of Saccharomyces cerevisiae show markers of oxidative stress and apoptosis.
Mol Microbiol. 2001; 39:1166–1173. [PubMed: 11251834]
Lee M, Lee JS. Exploiting tumor cell senescence in anticancer therapy. BMB reports. 2014; 47:51–59.
[PubMed: 24411464]
Lee Y, Kang HC, Lee BD, Lee YI, Kim YP, Shin JH. Poly (ADP-ribose) in the pathogenesis of
Parkinson's disease. BMB reports. 2014; 47:424–432. [PubMed: 24874851]
Leeuwenburgh C, Gurley CM, Strotman BA, Dupont-Versteegden EE. Age-related differences in
apoptosis with disuse atrophy in soleus muscle. American journal of physiology. Regulatory,
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 20
Metzstein MM, Stanfield GM, Horvitz HR. Genetics of programmed cell death in C. elegans: past,
present and future. Trends Genet. 1998; 14:410–416. [PubMed: 9820030]
Author Manuscript
Micchelli CA, Perrimon N. Evidence that stem cells reside in the adult Drosophila midgut epithelium.
Nature. 2006; 439:475–479. [PubMed: 16340959]
Misell LM, Hwang ES, Au A, Esserman L, Hellerstein MK. Development of a novel method for
measuring in vivo breast epithelial cell proliferation in humans. Breast cancer research and
treatment. 2005; 89:257–264. [PubMed: 15754124]
Nixon RA, Yang DS. Autophagy and neuronal cell death in neurological disorders. Cold Spring Harb
Perspect Biol 4. Oct 1.2012 4(10) pii: a008839. doi: 10.1101/cshperspect.a008839.
Nolan LA, Lunness HR, Lightman SL, Levy A. The effects of age and spontaneous adenoma
formation on trophic activity in the rat pituitary gland: a comparison with trophic activity in the
human pituitary and in human pituitary adenomas. Journal of neuroendocrinology. 1999; 11:393–
401. [PubMed: 10320567]
Ohlstein B, Spradling A. The adult Drosophila posterior midgut is maintained by pluripotent stem
cells. Nature. 2006; 439:470–474. [PubMed: 16340960]
Olivetti G, Melissari M, Capasso JM, Anversa P. Cardiomyopathy of the aging human heart. Myocyte
Author Manuscript
loss and reactive cellular hypertrophy. Circ Res. 1991; 68:1560–1568. [PubMed: 2036710]
Oppenheim RW. Cell death during development of the nervous system. Annual review of
neuroscience. 1991; 14:453–501.
Oshiro S, Morioka MS, Kikuchi M. Dysregulation of iron metabolism in Alzheimer's disease,
Parkinson's disease, and amyotrophic lateral sclerosis. Advances in pharmacological sciences.
2011; 2011:378278. [PubMed: 22013437]
Osiewacz HD, Borghouts C. Cellular copper homeostasis, mitochondrial DNA instabilities, and
lifespan control in the filamentous fungus Podospora anserina. Exp Gerontol. 2000; 35:677–686.
[PubMed: 11053657]
Osiewacz HD, Brust D, Hamann A, Kunstmann B, Luce K, Muller-Ohldach M, Scheckhuber CQ,
Servos J, Strobel I. Mitochondrial pathways governing stress resistance, life, and death in the
fungal aging model Podospora anserina. Ann N Y Acad Sci. 2010; 1197:54–66. [PubMed:
20536834]
Otera H, Ishihara N, Mihara K. New insights into the function and regulation of mitochondrial fission.
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 21
Qian D, Wu X, Jiang H, Gao P, Kuang C, Wang K, Huang L. Aging reduces susceptibility of vascular
smooth muscle cells to H(2)O(2)-induced apoptosis through the down-regulation of Jagged1
Author Manuscript
apoptotic protein BCL-xL. Mech Ageing Dev. 2008; 129:207–214. [PubMed: 18262222]
Ryu SJ, Oh YS, Park SC. Failure of stress-induced downregulation of Bcl-2 contributes to apoptosis
resistance in senescent human diploid fibroblasts. Cell Death Differ. 2007; 14:1020–1028.
[PubMed: 17290286]
Sainz RM, Mayo JC, Reiter RJ, Tan DX, Rodriguez C. Apoptosis in primary lymphoid organs with
aging. Microscopy research and technique. 2003; 62:524–539. [PubMed: 14635146]
Salminen A, Ojala J, Kaarniranta K. Apoptosis and aging: increased resistance to apoptosis enhances
the aging process. Cell Mol Life Sci. 2011; 68:1021–1031. [PubMed: 21116678]
Salvioli S, Monti D, Lanzarini C, Conte M, Pirazzini C, Bacalini MG, Garagnani P, Giuliani C,
Fontanesi E, Ostan R, Bucci L, Sevini F, Yani SL, Barbieri A, Lomartire L, Borelli V, Vianello D,
Bellavista E, Martucci M, Cevenini E, Pini E, Scurti M, Biondi F, Santoro A, Capri M,
Franceschi C. Immune system, cell senescence, aging and longevity--inflamm-aging reappraised.
Curr Pharm Des. 2013; 19:1675–1679. [PubMed: 23589904]
Savage VM, Allen AP, Brown JH, Gillooly JF, Herman AB, Woodruff WH, West GB. Scaling of
number, size, and metabolic rate of cells with body size in mammals. Proc Natl Acad Sci U S A.
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 22
Severin FF, Hyman AA. Pheromone induces programmed cell death in S. cerevisiae. Curr Biol. 2002;
12:R233–235. [PubMed: 11937036]
Author Manuscript
[PubMed: 15195070]
Spillare EA, Robles AI, Wang XW, Shen JC, Yu CE, Schellenberg GD, Harris CC. p53-mediated
apoptosis is attenuated in Werner syndrome cells. Genes Dev. 1999; 13:1355–1360. [PubMed:
10364153]
Stegh AH. Targeting the p53 signaling pathway in cancer therapy - the promises, challenges and perils.
Expert opinion on therapeutic targets. 2012; 16:67–83. [PubMed: 22239435]
Suh Y. Cell signaling in aging and apoptosis. Mech Ageing Dev. 2002; 123:881–890. [PubMed:
12044936]
Suh Y, Lee KA, Kim WH, Han BG, Vijg J, Park SC. Aging alters the apoptotic response to genotoxic
stress. Nat Med. 2002; 8:3–4. [PubMed: 11786880]
Taddei ML, Giannoni E, Fiaschi T, Chiarugi P. Anoikis: an emerging hallmark in health and diseases. J
Pathol. 2012; 226:380–393. [PubMed: 21953325]
Tarnawski AS, Ahluwalia A, Jones MK. Increased susceptibility of aging gastric mucosa to injury: the
mechanisms and clinical implications. World journal of gastroenterology : WJG. 2014; 20:4467–
4482. [PubMed: 24782600]
Author Manuscript
Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the cellular level. Nat Rev Mol
Cell Biol. 2008; 9:231–241. [PubMed: 18073771]
Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent
secretory phenotype: therapeutic opportunities. J Clin Invest. 2013; 123:966–972. [PubMed:
23454759]
Tower J. Sex-specific regulation of aging and apoptosis. Mech Ageing Dev. 2006; 127:705–718.
[PubMed: 16764907]
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 23
Tower J. Mitochondrial maintenance failure in aging and role of sexual dimorphism. Archives of
biochemistry and biophysics. Oct 25.2014 pii: S0003-9861 (14)00375-0. doi: 10.1016/j.abb.
Author Manuscript
2014.10.008.
Valente LJ, Gray DH, Michalak EM, Pinon-Hofbauer J, Egle A, Scott CL, Janic A, Strasser A. p53
efficiently suppresses tumor development in the complete absence of its cell-cycle inhibitory and
proapoptotic effectors p21, Puma, and Noxa. Cell reports. 2013; 3:1339–1345. [PubMed:
23665218]
Vasconsuelo A, Pronsato L, Ronda AC, Boland R, Milanesi L. Role of 17beta-estradiol and
testosterone in apoptosis. Steroids. 2011; 76:1223–1231. [PubMed: 21855557]
Vaughn AE, Deshmukh M. Glucose metabolism inhibits apoptosis in neurons and cancer cells by
redox inactivation of cytochrome c. Nat Cell Biol. 2008; 10:1477–1483. [PubMed: 19029908]
Vazquez A, Bond EE, Levine AJ, Bond GL. The genetics of the p53 pathway, apoptosis and cancer
therapy. Nature reviews. Drug discovery. 2008; 7:979–987. [PubMed: 19043449]
Vazquez-Padron RI, Lasko D, Li S, Louis L, Pestana IA, Pang M, Liotta C, Fornoni A, Aitouche A,
Pham SM. Aging exacerbates neointimal formation, and increases proliferation and reduces
susceptibility to apoptosis of vascular smooth muscle cells in mice. Journal of vascular surgery.
Author Manuscript
orthologue regulates apoptosis in yeast. J Cell Biol. 2004; 166:969–974. [PubMed: 15381687]
Yakovlev AG, Faden AI. Mechanisms of neural cell death: implications for development of
neuroprotective treatment strategies. NeuroRx : the journal of the American Society for
Experimental NeuroTherapeutics. 2004; 1:5–16. [PubMed: 15717003]
Yee C, Yang W, Hekimi S. The intrinsic apoptosis pathway mediates the pro-longevity response to
mitochondrial ROS in C. elegans. Cell. 2014; 157:897–909. [PubMed: 24813612]
Yeo EJ, Hwang YC, Kang CM, Choy HE, Park SC. Reduction of UV-induced cell death in the human
senescent fibroblasts. Mol Cells. 2000; 10:415–422. [PubMed: 10987139]
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 24
Zheng J, Edelman SW, Tharmarajah G, Walker DW, Pletcher SD, Seroude L. Differential patterns of
apoptosis in response to aging in Drosophila. Proc Natl Acad Sci U S A. 2005; 102:12083–
12088. [PubMed: 16099837]
Zhong X, Rescorla FJ. Cell surface adhesion molecules and adhesion-initiated signaling:
understanding of anoikis resistance mechanisms and therapeutic opportunities. Cellular
signalling. 2012; 24:393–401. [PubMed: 22024283]
Zhu C, Wang X, Deinum J, Huang Z, Gao J, Modjtahedi N, Neagu MR, Nilsson M, Eriksson PS,
Hagberg H, Luban J, Kroemer G, Blomgren K. Cyclophilin A participates in the nuclear
translocation of apoptosis-inducing factor in neurons after cerebral hypoxia-ischemia. J Exp
Med. 2007; 204:1741–1748. [PubMed: 17635954]
Zhu Y, Tchkonia T, Pirtskhalava T, Gower A, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Borden G,
Lenburg M, O'Hara SP, LaRusso NF, Miller JD, Roos CM, Verzosa GC, LeBrasseur NK, Wren
JD, Farr JN, Khosla S, Stout MB, McGowan SJ, Fuhrmann-Stroissnigg H, Gurkar AU, Zhao J,
Colangelo D, Dorronsoro A, Ling YY, Barghouthy AS, Navarro DC, Sano T, Robbins PD,
Author Manuscript
Niedernhofer LJ, Kirkland JL. The Achilles' Heel of Senescent Cells: From Transcriptome to
Senolytic Drugs. Aging Cell. Mar 9.2015 doi: 10.1111/acel.12344.
Zuckerman V, Wolyniec K, Sionov RV, Haupt S, Haupt Y. Tumour suppression by p53: the importance
of apoptosis and cellular senescence. J Pathol. 2009; 219:3–15. [PubMed: 19562738]
Author Manuscript
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.
Tower Page 25
Author Manuscript
Author Manuscript
Author Manuscript
Figure 1.
Cross-species comparison of PCD factors implicated in aging. The factors and regulatory
relationships that appear similar between mammals, C. elegans and S. cerevisiae are
emphasized; additional regulatory factors exist for each species but are not indicated for sake
of clarity. For mammals the canonical intrinsic caspase-dependent apoptotic pathway is
outlined. AIF and EndoG are caspase-independent and translocate to the nucleus where they
mediate DNA fragmentation and chromatin condensation. Please see text for details.
Author Manuscript
Ageing Res Rev. Author manuscript; available in PMC 2016 September 01.