Protocols For Macroalgae Research
Protocols For Macroalgae Research
Research
Protocols for Macroalgae
Research
Edited by
Bénédicte Charrier
Thomas Wichard
C.R.K. Reddy
Central cover photo courtesy of (c) Station Biologique de Roscoff, Yann Fontana.
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742
This book contains information obtained from authentic and highly regarded sources. Reasonable
efforts have been made to publish reliable data and information, but the author and publisher cannot
assume responsibility for the validity of all materials or the consequences of their use. The authors and
publishers have attempted to trace the copyright holders of all material reproduced in this publication
and apologize to copyright holders if permission to publish in this form has not been obtained. If any
copyright material has not been acknowledged please write and let us know so we may rectify in any
future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced,
transmitted, or utilized in any form by any electronic, mechanical, or other means, now known or
hereafter invented, including photocopying, microfilming, and recording, or in any information
storage or retrieval system, without written permission from the publishers.
For permission to photocopy or use material electronically from this work, please access www.
copyright.com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC),
222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that
provides licenses and registration for a variety of users. For organizations that have been granted a
photocopy license by the CCC, a separate system of payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are
used only for identification and explanation without intent to infringe.
v
vi Contents
ix
x Preface
Bénédicte Charrier
Roscoff, France
Thomas Wichard
Jena, Germany
C.R.K. Reddy
Bhavnagar, India
Editors
Dr. Bénédicte Charrier is a senior scientist from Centre National de la
Recherche Scientifique (CNRS) working at Station Biologique de Roscoff,
Roscoff, France. She is a specialist in macroalgae morphogenesis and
development, with an expertise on the filamentous alga Ectocarpus silicu-
losus. She is currently the Chair of the European network Phycomorph, sup-
ported by the Cooperation in Science & Technology (COST) Association
(2015–2019), which aims to coordinate research in growth, reproduction,
and morphogenesis of macroalgae in Europe and in association with
Asian and North American laboratories.
xiii
Contributors
Tânia Aires* and
Centro de Ciencias do Mar
Centro de Investigación Marina
(CCMAR)
Quintay (CIMARQ), Facultad de
CIMAR, Universidade do Algarve
Ecología y Recursos Naturales
Faro, Portugal
Universidad Andres Bello
Quintay, Chile
G. Alongi
Department of Biological, Inka Bartsch*
Geological and Environmental Alfred-Wegener-Institute
Sciences Helmholtz Center for Polar and
University of Catania Marine Research
Catania, Italy Bremerhaven, Germany
xv
xvi Contributors
Elizabeth Garrido-Ramírez
Centro de Investigación para la Paulien Harmsen
Sustentabilidad, Facultad de Wageningen Food & Biobased
Ecología y Recursos Naturales Research
Universidad Andres Bello Wageningen, the Netherlands
Santiago, Chile
Sandra Heinrich*
Angelika Graiff* Biozentrum Klein Flottbek
Applied Ecology and Phycology, University of Hamburg
Institute of Biological Sciences Hamburg, Germany
University of Rostock
Rostock, Germany Cécile Hervé*
Marine Biology
Agnès Groisillier* Station Biologique de Roscoff
Algal Biology and Interactions Roscoff, France
with the Environment
Station Biologique de Roscoff
Roscoff, France Takashi Hirayama
Institute of Plant Science and
Britta Grote Resources
Alfred-Wegener Institute Okayama University
Helmholtz Centre for Polar and Kurashiki, Japan
Marine Research (AWI)
Bremerhaven, Germany Xiaoru Hou*
Center of Bioresource and
Vishal Gupta Biorefinery
Biological Oceanography Division Danish Technological Institute
CSIR–National Institute of Taastrup, Denmark
Oceanography
Goa, India
Contributors xix
Thomas Torode
Centre for Plant Sciences, Faculty
of Biological Sciences
University of Leeds
Leeds, United Kingdom
and
The Sainsbury Laboratory
University of Cambridge
Cambridge, United Kingdom
* Corresponding author.
section one
Cultivating and preserving
seaweeds
chapter one
Seaweed in high-energy
environments
Protocol to move Saccharina
cultivation offshore
Bela H. Buck and Britta Grote
Contents
1.1 Introduction to offshore aquaculture..................................................... 4
1.2 State of the art of offshore seaweed cultivation.................................... 5
1.3 Land-based preparation and site selection ........................................... 8
1.3.1 Site selection .................................................................................. 9
1.3.1.1 Site selection: Advance information
of the local area ashore and at sea ............................. 10
1.3.1.2 Site selection: Site-specific, oceanographic,
and water quality parameter ...................................... 11
1.3.1.3 Site selection: Economic/technical/expansion
feasibility ....................................................................... 12
1.3.2 Equipment and system design .................................................. 13
1.3.2.1 System design for the offshore seaweed farm ......... 17
1.3.3 Seeding procedure ...................................................................... 18
1.4 Transfer at sea .......................................................................................... 20
1.4.1 Deployment of the offshore farm ............................................. 20
1.4.1.1 Deployment of the farm .............................................. 21
1.4.2 Operation, maintenance, and harvest...................................... 24
1.4.2.1 Operation and maintenance at sea ............................ 24
1.4.2.2 Harvest .......................................................................... 25
1.4.3 Problems and failures ................................................................ 26
1.4.3.1 Prevention of mistakes ................................................ 27
1.5 Multi-use of offshore installations ....................................................... 29
References.......................................................................................................... 32
3
4 Protocols for Macroalgae Research
(a) (b)
Figure 1.1 Alfred Wegener Institute Helmholtz Centre for Polar and Marine
Research (AWI) in Bremerhaven (Germany). The image shows the main build-
ing with two research vessels in front. (a) RV Heincke vessel was used to carry
the equipment for the offshore farm and for deploying the moorings, and
(b) RV Uthörn vessel was used to install the complete harness, including system
design, buoyancy, seeded Laminarian ropes, and for O&M and harvest. (Courtesy
of Müller-Elsner and AWI.)
and Buchholz 1996; Buck and Buchholz 2004a, 2005). The technology used
in earlier studies was then revised with a focus on the easy deployment,
maintenance, harvesting, strength and connectedness to the foundations of
offshore wind turbines (Lüning and Buchholz 1996, Buck 2001; Buck 2004;
Buck and Buchholz 2004a, b). For this purpose, the first co-use studies for
the combination of aquaculture and offshore wind farms emerged (Buck
2004). One result of these studies was the patented ring construction for the
offshore cultivation of seaweed on an industrial scale (Buck and Buchholz
2004a, b).
Similar to the research in Germany, the offshore seaweed cultiva-
tion is discussed in co-use with existing offshore structures in Norway
(Skjermo et al. 2014). The main reason to relocate aquaculture offshore
is the negative impact of nutrient-rich effluents from the Norwegian
salmon farms on marine ecosystems in coastal areas. In 2011, the project
SWEEDTECH investigated the development of an economic system for
commercial-scale offshore seaweed cultivation, including the develop-
ment of seeding strategies of carrier material, design, and improvement
for a structural rig, and of innovative deployment and harvesting methods
(SES 2015a, b). As a result of this project, a new seaweed cultivation
Chapter one: Seaweed in high-energy environments 7
2000 until today (Buck et al. 2008; Buck et al. 2017a, b). Several types of
seaweed cultivation in the offshore realm were tested, such as longline
or ring constructions (Buck and Buchholz 2004a). The protocol briefly
describes the land-based preparation, including a list of equipment
needed, the seeding procedure, and the preparations to move offshore.
However, as this chapter is focusing on the offshore cultivation of seaweed,
for example, S. latissima, we do not go into detail regarding the complete
life cycle and reproduction of this species and will describe the cultiva-
tion mainly about the offshore aspect, including the technical design. For
detailed descriptions of the life cycle of S. latissima, the seedling protocol
and hybridization of kelp also refer Chapter 2 by Forbord et al. (2017) and
Chapter 3 by Bartsch within this compilation.
the early 2000 (Buck 2004). As a result, a ring design was developed that
resisted all sea conditions, whereas at the same time producing approxi-
mately one ton of seaweed biomass (wet weight) on roughly 20 m2 (Buck
and Buchholz 2004a,b). Since then, several system designs were developed
offshore from different institutions and withstood storm conditions.
In the following section, we summarize the major design criteria when
moving from nearshore sheltered conditions in offshore environments:
Third stage:
Figure 1.2 Site-selection process for a seaweed farm, including typical seaweed
site-selection criteria. First stage is the start of the selection process and will end
through the second stage at the end of the third stage. At the end of each stage, several
sites are identified but will be reduced to the specification process (grey arrows).
Parameters with an exclamation mark need special attention when moving off the
coast into a hostile environment.
• It could be relevant for the permit, and any other emerging issues
related to regulations and jurisdictions. It could support or hin-
der the issue of a lease and can be influenced if there are already
reserved areas, which, in turn, would have the marginal potential for
stakeholder conflicts.
• It could also be important for potential cofinance, any other finan-
cial support, and taxation.
• The condition of the seafloor (soft bottom, hard substrate, and depth)
will reduce the number of selected sites, and therefore ease the site
selection process.
• Preapproved areas for aquaculture would support the selection of
good sites.
• This could influence the growth, morphology, and health, and the
biofouling on the seaweed (nutrient concentration, light, water qual-
ity, etc., on short-term and an annual level). Most of the conditions
should be stable year round.
• The offshore conditions (wave/swell, current velocity, wind/fetch,
etc.), and the depth/topography, will affect the technology needed,
as the construction should, of course, withstand all potential
worst-case conditions while also having a long, expected lifetime.
• It is urgent to have a land-based facility close to the shore/harbor/
pier to allow quick and logistically easy handling.
The success of the planning procedure of the enterprise and the site selec-
tion will depend to a large extent on a thorough site survey and proper
assessment about the project development.
Chapter one: Seaweed in high-energy environments 13
(a) Ball-like floats for Marker/corner buoy (b) Torpedo/corner buoy Pencil-like floats
buoyancy (small) (large) (large) for buoyancy
Longline/ (small)
backbone
50−150 m
30 m
Anchor Connecting
Ball-like Pencil-like rope rings Subanchor Main anchor
float float
(f ) (g)
Figure 1.3 (a–g): System design and the concept of seaweed cultivation devices.
(a) Longline (backbone) system with floating buoyancy and the seaweed culture
unit hanging perpendicular in the water column; (b) large grid design with float-
ing buoyancy and rectangular culture units; (c) connection devices with (C 1)
rings as coupling center piece, connection of floats to the backbone within the
strands of the rope (C 2), or by using metal tiles (C 3); (d) ladder construction
with culture units attached to the multiple backbone of the system; (e) unit at one
end of a backbone with different anchors, holding devices (chains, ropes), and
flotation; (f) different buoy shapes and dipping depths when riding the swell;
and (g) backbone with floating and submerged flotation and the unusable segment.
(Modified after Buck 2007 and Buck & Buchholz 2004.)
14 Protocols for Macroalgae Research
Central buoy
AWI Central
guide ring Carrier
ropes
Thimble Upper
crow’s
foot
Metal
cuff Culture
(c) line
Lower
crow’s
foot
Warble
Central
(d)
steel
cable
(a)
Figure 1.4 (a–d): Offshore-ring device for the cultivation of seaweed. (a) Ring
construction for the culture of Laminarian species at offshore locations, includ-
ing major elements magnified in; (b) metal cuffs, to which the upper and lower
crow’s feet, the ring tube, and the carrier ropes are attached; (c) the central guide
ring with attached carrier ropes and culture lines; and (d) transition between the
central steel cable of the mooring and that of the lower crow’s foot. (Modified after
Buck and Buchholz 2004a.)
(a) (b)
(c) (d)
(e) (f )
Figure 1.5 (a–f): Different development steps of the offshore-ring device. (a) Shows
the center piece in the first stage, which was because of the severe conditions in
the open ocean exchanged by the central guide ring; (c) connection of the carrier
ropes to the ring and the modified coupling devices (d); (e) small ring device with
2.5–3.0 m in diameter and the version with 5 m in diameter during the harvest (f).
(Modified after Buck et al. 2017b.)
16 Protocols for Macroalgae Research
(a)
(c) (b)
(d) (e)
Offshore
Streamlined,
flad and narrow
Undulated,
ruffled and wide
Row of tanks with
automated seeding drums
(a) (b) Sheltered
Shaft
Power
reel
(d)
Seeding drums
with ropes
Motor
(c) (e)
(f ) (g) (h)
Figure 1.7 (a–h): Preparation of seeded ropes for the transfer to the offshore farm.
(a) Seaweed production site with greenhouse and seaweed production tanks at the
AWI in List (Sylt, North-Germany); (b) adaptation of seaweed blades (Saccharina
latissima) originating from offshore and sheltered environments; (c–e) tanks with
seeding drums and its motorization; (f–h) seeding rope preparation from empty
curtain devices to young sporophytes ready for transfer at sea. Red arrows display
the process from prepared empty curtain-like ropes (f) to seeded ropes coiled on
drums (c), through young sporophytes (g) to storage for the transfer at sea (h).
(Modified after Buck & Buchholz 2005; Buck et al. 2017a.)
20 Protocols for Macroalgae Research
(b)
(a)
(c) (d)
Figure 1.8 (a–d): Deployment at sea under harsh conditions. (a) Short time
slots are not rare and should be used if necessary to deploy heavy equipment
(e.g., concrete blocks); (b) the vessel needs to maneuver on the spot; (c) equipment
can be extremely heavy and needs the entire crew to handle, also at night times;
(d) typical harsh conditions at the seaweed farm site at Roter Sand, 17 nautical
miles off the coast of the city of Bremerhaven (Germany).
22 Protocols for Macroalgae Research
(a) (b)
(c)
(d) (e)
(f)
(g) (h)
(i) (j)
Figure 1.9 (a–j): Deployment of the seaweed farm at the offshore site. (a)–(c)
concrete anchor clocks (2 tons) lashed on board of the service vessel RV Heincke
and deployment of the concrete blocks at the farm site; (d) floating-ring device
(first generation) after deployment; (e) coupling of various ring devices before
towing them to the farm site; (f) mooring line length adjusting and preparation
depending on different depths; (g) torpedo-like sensor for towing tests to examine
force loads resulting from and on farm devices; (h) deployment of farm designs
in a connected mode; (i) O&M vessel Aade at the farm site; (j) longlines after
deployment. (Modified after Buck et al. 2017a,b.)
(a) (b)
(c)
Figure 1.10 (a–c): Land-based work previously conducted before the system will
be transferred at sea. (a) Preparation and arrangement of swivels, thimbles, and
shackles with the Laminarian seeding line; (b) heat preparation of rope ends
by using an electric heat cutter to avoid unraveling and opening of rope ends;
(c) image shows the size of chains, shackles, swivels, and thimbles to connect the
buoyancy to the backbone.
Mooring
• Care and consideration should be taken for the moorings pro-
cedure to deploy the backbone and the entire seaweed harness.
• Topography conditions and sediment type is an important infor-
mation to ease the mooring procedure.
• The mooring device is only effective if it remains in its posi-
tion under the farm load at all times unless the farm should be
retrieved for any reason.
Diving at the farm site
• Diving in the open ocean is another challenge and needs expe-
rienced divers with the right certificate while also following all
potential safety regulations.
• However, diving still needs to be done as the farmer from time
to time needs to know what is happening with the system under
24 Protocols for Macroalgae Research
the surface: Is the mooring still in place, are the ropes, chains
connected, how is the condition of the connecting material (swiv-
els, thimbles, shackles, etc.)?
1.4.2.2 Harvest
Identification of market size
• To find the right time of harvest, a certain length of the cultivated
plants should be gained. Market size depends on species, seed-
ing density, time of the year, and site conditions (nutrients, physi-
cal stress, etc.). For example, S. latissima grown in the North Sea
(East Atlantic) normally reach a market size of approximately
200 cm between May–June (Figure 1.11).
• If the harvest will be conducted too early, it is important to plan
the further process. As open ocean locations could be subject to
swift weather condition change, it might be necessary to harvest
the crop, even if the plant lamina did not reach the planned mar-
ket size. Here, it is essential to check the local weather forecast.
• Further, if the harvest will be planned too late, an increased tip
loss could result as fouling organisms settle on the blades and
Between
180–240 cm
(a) (b)
In the following section, some of the problems and failures are listed,
which should be taken into account before a seaweed enterprise would
be installed in the open ocean. Offshore aquaculture constructions were
engineered to withstand tremendous forces for many years, but can be
damaged or destroyed during heavy storms. Staying ahead of the O&M
required on the ring device proved problematic during the project, and
it was a constant challenge for personnel to maintain the integrity of the
installation and mooring. Entanglement, damage, and loss occurred dur-
ing continuing storm and heavy seas conditions and even exacerbated in
the next round of damage if repairs could not be organized quickly between
weather events (Figure 1.13). Other problems accumulated in designing
incorrect dimensions in size, connectedness, attachment, coupling, and
space (Figure 1.13), and resulted in further entanglement, increased loads,
and, corresponding to the service load, in too much biomass per size of
construction. Escaping the wave and current energy at the water surface
into a submerged mode is critical, as the seaweed need a certain amount of
irradiance to allow photosynthesis. Learning from these miscalculations
and setbacks, the following issues have to be taken into account.
(d) (e) (f )
Figure 1.13 (a–f): Failures and miscalculations from first offshore seaweed farms
at Helgoland and at the offshore light house Roter Sand (a,b) Moreover, displays
submerged buoys in full and collapsed shape; (c) shows a ravel of buoys, ropes,
seeding lines, and connecting pieces after a heavy storm event; (d) shows a bro-
ken buoy rope leading to the loss of equipment; (e) is an image of an unstable
ring device (First generation in 1994) with a too large biomass for harvest; and
(f) shows the entanglement of little ring devices resulting from a wrong mooring
and harness calculation. (Modified after Kaiser and Chambers 2017; Buck and
Langan 2017b.)
Planning
• Ocean conditions and weather determine everything offshore,
so plans must be made and adjusted accordingly. Nothing can be
forced if conditions do not allow it as the ocean will always win
in that situation.
• Redundancy and backup systems where applicable, especially
with regards to moorings, which are arguably the most critical
components of the farm system.
• Prepare as much equipment as possible while the system is
onshore. It becomes exponentially harder to work on ropes,
chains, anchors, backbones, shackles, etc., the moment it enters
the water.
Chapter one: Seaweed in high-energy environments 29
Offshore ring
Pylon
Mooring
1, 5–5 m
1, 5–5 m
Buoyancy
Offshore ring
Longline
Macroalgae
Blue mussels Concrete
Mooring block
(a) (b)
Figure 1.14 (a–b): First drawings to show the Multi-use concept combining aqua-
culture with wind farms. (a) Top a birds-eye view of longline constructions. Special
pylon anchorages ensure access and maintenance of the wind energy plants by ship;
down side view of a submerged mussel and seaweed longline culture. The wind
energy plant pylon is used as the anchorage for one end of the long line. The line
can reach a length of 100–300 m. Longer lines may be mounted to the next pylon;
(b) top A bird’s-eye view of the described ring construction. The combined rings are
fixed around the pylon and a pylon with a single offshore ring construction using
two anchorages; down side view of a pylon with offshore ring construction used for
cultivation of algae and mussels. An anchor stone is placed at the outermost point
of the ring construction; (Continued)
30 Protocols for Macroalgae Research
Rotating drum
Buoyancy SOSSEC
Longline
Macroalgae
Oyster
trays
Oyster cage Concrete
block
Blue mussels
(c) (d)
Figure 1.14 (Continued) (c–d): First drawings to show the Multi-use concept com-
bining aquaculture with wind farms. (c) side view of a turbine–oyster culture
combination (oyster trays fixed to a longline and a rotating oyster cage/drum);
and (d) SOSSEC design (Submersible Offshore Shellfish and Seaweed Cage),
which is in a submerged mode and can be lifted to the surface for harvest during
culture. (Buck 2000, 2004; Buck and Langan 2017.)
(Holm et al. 2017). These claims can not only lead to user conflicts but
also to smart combinations of multiple uses of marine space with eco-
nomic and ecological benefits (Michler-Cieluch et al. 2009; Krause et al.
2011; Van Hoof et al. 2014). Such multi-use concepts are not new ideas as
diverse marine activities already coexisted for decades, such as fishing
and shipping, nature conservation, and a mussel fishery, and artificial
reefs and angling.
Resulting from the Blue Growth strategy of the European
Commission, the importance of the perspective of offshore aquaculture
is increasing. Moving aquaculture offshore has the potential for further
growth of the industry in many European offshore areas (Troell et al.
2009; Rosenthal et al. 2012a, b). In recent years, many studies investigat-
ing the co-use of offshore structures and areas have been conducted or
are still ongoing (i.e., MERMAID, COEXIST, TROPOS, ICES WGAQUA,
and MUSES).
The North Sea is, for example, a highly used marine space with
fisheries, tourism, shipping, oil and gas extraction, cables and
Chapter one: Seaweed in high-energy environments 31
References
ACE 2015. ACE expands with seaweed site. Accessed February 20, 2017, at http://
aceaqua.no/en/2015/09/ace-expands-with-seaweed-site
Barrington, K., Chopin, T., and Robinson, S. 2009. Integrated multi-trophic aqua-
culture (IMTA) in marine temperate waters. In: D. Soto (Ed.), Integrated
Mariculture: A Global Review. FAO Fisheries and Aquaculture Technical Paper
No. 529, Rome, Italy: FAO, pp. 7–46.
Bartsch, I. 2017. Derivation of clonal stock cultures and hybridisation of kelps: Tool
for strain preservation and breeding programs. In: B. Charrier, T. Wichard,
and C.R.K. Reddy (Eds.), Protocols for Macroalgae Research, Chapter 3. Boca
Raton, FL: CRC Press, Taylor & Francis Group.
Benassai, G., Mariani, P., Stenberg, C., and Christoffersen, M. 2014. A sustainabil-
ity index of potential co-location of offshore wind farms and open water
aquaculture. Ocean Coast. Manag. 95: 213–218.
Benetti, D., Benetti, G.I., Rivera, J.A., and O’Hanlon, B. 2010. Site selection criteria
for open ocean aquaculture. Mar. Tech. Soc. J. 44(3): 22–35.
Broch, O.J., Ellingsen, I.H., Forbord, S., Wang, X., Volent, Z., Alver, M.O., Handå, A.
et al. 2013. Modelling the cultivation and bioremediation potential of the
kelp Saccharina latissima in close proximity to an exposed salmon farm in
Norway. Aquac. Environ. Interact. 4: 186–206.
Buchholz, C.M., Krause, G., and Buck, B.H. 2012. Seaweed and man. In: C. Wiencke
and C. K. Bischof (Eds.), Seaweed Biology, Ecological Studies. Berlin, Germany:
Springer-Verlag, pp. 471–493.
Buck, B.H. 2000. Open ocean aquaculture und offshore-windparks: Eine mach-
barkeitsstudie über die multifunktionale nutzung von offshore-windparks
und offshore-marikultur im raum nordsee. Reports on Polar and Marine
Research, 412. Alfred Wegener Institute for Polar and Marine Research,
Bremerhaven, Germany.
Buck, B.H. 2001. Combined utilisation of wind farming and mariculture in the
North Sea. Biennial Report of Science Highlights at the Alfred Wegener
Institute Helmholtz Centre for Polar and Marine Research, Bremerhaven,
Germany, pp. 33–39.
Buck, B.H. 2004. Farming in a high energy environment: Potentials and con-
straints of sustainable offshore aquaculture in the German bight (North
Sea), PhD Thesis, Alfred Wegener Institute for Polar and Marine Research
Bremerhaven, 214 p., Bremen, Germany: University of Bremen.
Buck, B.H. 2007. Experimental trials on the feasibility of offshore seed produc-
tion of the mussel Mytilus edulis in the German bight: Installation, tech-
nical requirements and environmental conditions. Helgol. Mar. Res. 61:
87–101.
Buck, B.H. and Buchholz, C.M. 2004a. The offshore-ring: A new system design for
the open ocean aquaculture of macroalgae. J. Appl. Phycol. 16: 355–368.
Buck, B.H. and Buchholz, C.M. 2004b. Support device for the cultivation of macro
organisms in marine waters. Patent: PCT/DE2005/000234.
Buck, B.H. and Buchholz, C.M. 2005. Response of offshore cultivated Laminaria
saccharina to hydrodynamic forcing in the North Sea. Aquaculture 250:
674–691.
Chapter one: Seaweed in high-energy environments 33
Buck, B.H. and Krause, G. 2012. Integration of aquaculture and renewable energy
systems. In: R.A. Meyers (Ed.), Encyclopaedia of Sustainability Science and
Technology Vol. 1. Berlin, Germany: Springer Science+Business Media LLC.
Chapter No. 180, pp. 511–533.
Buck, B.H. and Langan, R. (Eds.) 2017a. Aquaculture Perspective of Multi-use Sites in
the Open Ocean: The Untapped Potential for Marine Resources in the Anthropocene.
Springer, 404 p.
Buck, B.H. and Langan, R. 2017b. Epilogue—Pathways towards sustainable ocean
food production. In: B.H. Buck and R. Langan (Eds.), Aquaculture Perspective
of Multi-Use Sites in the Open Ocean: The Untapped Potential for Marine Resources
in the Anthropocene. Cham, Switzerland: Springer International Publishing
AG, pp. 394–404.
Buck, B.H., Ebeling, M., and Griffin, R. 2011. Seaweed aquaculture as a co-use
in an offshore wind farm—Feasibilities and constraints of economic via-
bility for cape wind (Massachusetts). AWI-Report – Project No. USA 11/
A01—12 p.
Buck, B.H., Krause, G., and Rosenthal, H. 2004. Extensive open ocean aquacul-
ture development within wind farms in Germany: The prospect of off-
shore co-management and legal constraints. Ocean Coast. Manag. 47(3–4):
95–122.
Buck, B.H., Krause, G., Michler-Cieluch, T., Brenner, M., Buchholz, C.M., Busch,
J.A., Fisch, R., Geisen, M., and Zielinski, O. 2008. Meeting the quest for spa-
tial efficiency: Progress and prospects of extensive aquaculture within off-
shore wind farms. Helgol. Mar. Res. 62: 269–281.
Buck, B.H., Krause, G., Pogoda, P., Grote, B., Wever, L., Goseberg, N., Schupp,
M.F., Mochtak, A., and Czybulka, D. 2017b. Case study German bight:
Pioneer projects of aquaculture-wind farm multi-uses. In: B.H. Buck and
R. Langan (Eds.), Aquaculture Perspective of Multi-use Sites in the Open Ocean:
The Untapped Potential for Marine Resources in the Anthropocene. Cham,
Switzerland: Springer International Publishing AG, pp. 253–354.
Buck, B.H., Nevejan, N., Wille, M., Chambers, M., and Chopin, T. 2017a. Offshore
and multi-use aquaculture with extractive species: Seaweeds and bivalves.
In: B.H. Buck and R. Langan (Eds.), Aquaculture Perspective of Multi-Use
Sites in the Open Ocean: The Untapped Potential for Marine Resources in the
Anthropocene. Cham, Switzerland: Springer International Publishing AG,
pp. 23–69.
Chopin, T. 2014. Seaweeds: Top mariculture crop, ecosystem service provider.
GAA 17: 54–56.
Clements, J. and Chopin, T. 2016. Ocean acidification and marine aquaculture in
North America: Potential impacts and mitigation strategies. Rev. Aquacult.
1–16. doi: 10.1111/raq.12140.
Corey, P., Kim, J.K., Garbary, D.J., Prithiviraj, B., and Duston, J. 2012. Bioremediation
potential of Chondrus crispus (Basin Head) and Palmaria palmata: Effect
of temperature and high nitrate on nutrient removal. J. Appl. Phycol. 24:
441–448.
Costanza, R. and Folke, C. 1997. Valuing ecosystem services with efficiency, fair-
ness and sustainability as goals. In: G. Daily (Ed.), Nature’s Services: Societal
Dependence on Natural Ecosystems. Washington, DC: Island Press, pp. 49–70.
34 Protocols for Macroalgae Research
Lagerveld, S., Röckmann, C., and Scholl, M. (Eds.) 2014. Combining offshore
wind energy and large-scale mussel farming: Background and technical,
ecological and economic considerations. IMARES report number C056/14
http://www.maritimecampus.nl/sites/default/files/C056%2014%20Report-
Blauwdruk-_SLMS-lcs.pdf
Langan, R. 2009. Opportunities and challenges for offshore farming. Chapter 29
In: G. Burnell and G. Allen (Eds.), New Technologies in Aquaculture: Improving
Production Efficiency, Quality and Environmental Management. Cambridge,
UK: Woodhead Publishing Limited.
Lüning, K. and Buchholz, C. 1996. Massenkultur mariner makroalgen bei helgo-
land zur gewinnung von phycokolloiden und zur verwendung als biosorp-
tionsmittel. Helgoland, Germany: BAH Report, 75 p.
Mann, K.H. 1972. Ecological energetics of the sea-weed zone in a marine bay on
the Atlantic coast of Canada. II. Productivity of the seaweeds. Mar. Biol. 14:
199–209.
Marinho, G.S., Holdt, S.L., Birkeland, M.J., and Angelidaki, I. 2015. Commercial
cultivation and bioremediation potential of sugar kelp, Saccharina latissima,
in Danish waters. J. Appl. Phycol. doi: 10.1007/s10811-014-0519-8.
McHugh, D.J. 2003. A guide to the seaweed industry. FAO Fisheries Technical
Papers T441.
McElwee, J. 1997. Practical experience in off-shore cage rearing, the good, the
bad and the ugly. In: C. Hesley (Ed.), Open Ocean Aquaculture: Chartering the
Future of Ocean Farming. Proceedings of an International Conference, April 23–25,
1997, Maui, Hawaii: UNIHI-Seagrant-CP-98-08, Maui, University of Hawaii
Sea Grant College Program, pp. 89–105.
Michler-Cieluch, T., Krause, G., and Buck, B.H. 2009. Reflections on integrating
operation and maintenance activities of offshore wind farms and maricul-
ture. Ocean Coast. Manag. 52(1): 57–68.
Naylor, R.L., Goldburg, R.J., Primavera, J.H., Kautsky, N., Beveridge, M.C., Clay, J.,
Folke, C., Lubchenco, J., Mooney, H., and Troell, M. 2000. Effect of aquacul-
ture on world fish supplies. Nature 405(6790): 1017–1024.
Neori, A., Chopin, T., Troell, M., Buschmann, A.H., Kraemer, G., Halling, C.,
Shpigel, M., and Yarish, C. 2004. Integrated aquaculture: Rationale, evolu-
tion and state of the art emphasizing seaweed biofiltration in modern aqua-
culture. Aquaculture 231: 361–391.
Neushul, M. and Harger, B.W.W. 1985. Studies of biomass yield from a near-shore
macroalgal test farm. J. Solar Energy Eng. 107: 93–96.
Neushul, M., Benson, J., Harger, B.W.W., and Charters, A.C. 1992. Macroalgal
farming in the sea: Water motion and nitrate uptake. J. Appl. Phycol. 4:
255–265.
Perez, P., Kaas, R., Campello, F., Arbault, S., and Barbaroux, O. 1992. La Culture des
Algues Marines Dans le Monde. Nantes, France: IFREMER.
Poelman, M. and Bolman, B. 2010. Task 1.3 COEXIST: Inventory of possible com-
binations of different forms of aquaculture and/or fisheries and other
activities. Accessed March 24, 2017, at http://www.coexistproject.eu/
images/COEXIST/deliverables/WP1/COEXIST_245178_D1.3_FINAL.pdf
Rosenthal, H., Costa-Pierce, B.A., Krause, G., and Buck, B.H. 2012a. Bremerhaven
Declaration on the Future of Global Open Ocean Aquaculture, Part I: Preamble
and Recommendations. Bremerhaven, Germany: The Bremerhaven Economic
Development Company.
36 Protocols for Macroalgae Research
Rosenthal, H., Costa-Pierce, B.A., Krause, G., and Buck, B.H. 2012b. Bremerhaven
Declaration on the Future of Global Open Ocean Aquaculture—Part II:
Recommendations on Subject Areas and Justifications. Aquaculture Forum on
Open Ocean Aquaculture Development—From Visions to Reality: The Future
of Offshore Farming. Bremerhaven, Germany: The Bremerhaven Economic
Development Company.
Ryan, J. 2005. Offshore aquaculture—Do we need it, and why is it taking so long?
International Salmon Farmers Association (Ireland). Expert workshop on
sustainable aquaculture, DG JRC European Commission, pp. 17–18. January
2005, Seville, Spain: Institute for Prospective Technological Studies.
SES. 2015a. Patented Seaweed Carrier system by SES, Patent No. EP 09 836 439.1.
SES—Seaweed Energy Solutions, Trondheim, Oslo, Norway.
SES. 2015b. Seaweed Carrier system for the cultivation of macroalgae in offshore
environments patented by SES—Seaweed Energy Solutions, Trondheim,
Norway.
Skjermo, J., Aasen, I.M., Arff, J., Broch, O.J., Carvajal, A., Christie, H., Forbord, S.
et al. 2014. A new Norwegian bioeconomy based on cultivation and pro-
cessing of seaweed: Opportunities and R&D needs. SINTEF Fisheries and
Aquaculture, Norway, March 6, 2014, 46 p.
Starchild, A. 1980. Seaward Ho!—The ocean frontier. In: Office of Technology
Assessment (Ed.), Energy from open ocean kelp farms (pp. 83–98). Honolulu,
Hawaii: University Press of the Pacific.
Stelzenmüller, V., Schulze, T., Gimpel, A., Bartelings, H., Bello, E., Bergh, O.,
Bolman, B. et al. 2013. Guidance on a better integration of aquaculture, fish-
eries, and other activities in the coastal zone: From tools to practical exam-
ples. Coexist project—Interaction in coastal waters.
Stickney, R.R. and McVey, J. 2002. Responsible Aquaculture. Wallingford,
Oxfordshire: CABI Press.
Stuiver, M., Gerritsen, A.L., Fontein, R.J., and Agricola, H.J. 2012. Multifunctionele
platforms: Perspectief voor de toekomst? Aquacultuur 27(5): 6–12
Troell, M., Joyce, A., Chopin, T., Neori, A., Buschmann, A.H., and Fang, J.G. 2009.
Ecological engineering in aquaculture: Potential for integrated multi-trophic
aquaculture (IMTA) in marine offshore systems. Aquaculture 297: 1–9.
Van Hoof, L.J.W., Steenbergen, J., Bolman, B.C., Rockmann, C., Kraan, M.L., Piet,
G.J., Jak, R.G. et al. 2014. Zee op Zicht: Inzicht; een zoektocht naar een
integraal afwegingskader voor het gebruik van de zee. IMARES Report.
Accessed March 24, 2017, at http://www.wageningenur.nl/nl/Publicatie-
details.htm?publicationId=publication-way-343835313839
chapter two
Contents
2.1 Introduction ............................................................................................. 38
2.1.1 The life cycle of laminariales .................................................... 39
2.2 State of the art .......................................................................................... 41
2.3 Materials................................................................................................... 41
2.3.1 Collection of sporophytes and induction of sori .................... 41
2.3.2 Sori disinfection, dehydration, spore release,
and spore counting ..................................................................... 42
2.3.3 Starting gametophyte cultures ................................................. 42
2.3.4 Maintenance of gametophyte cultures .................................... 42
2.3.5 Fertility test .................................................................................. 43
2.3.6 Seeding ......................................................................................... 43
2.4 Experimental procedures ...................................................................... 43
2.4.1 Seed supply and sporulation..................................................... 43
2.4.1.1 Collection of sporophytes and induction
of sorus.....................................................................43
2.4.1.2 Sorus disinfection ........................................................ 45
2.4.1.3 Dehydration .................................................................. 45
2.4.1.4 Spore release ................................................................. 45
2.4.1.5 Spore density ................................................................ 46
2.4.2 Gametophyte cultivation ........................................................... 46
2.4.2.1 Starting-up gametophyte cultures............................. 46
2.4.2.2 Maintenance of gametophyte cultures ..................... 48
2.4.2.3 Contamination control ................................................ 49
2.4.2.4 Fertility test ................................................................... 49
2.4.3 Seeding ......................................................................................... 49
2.4.3.1 Seeding with spores..................................................... 50
2.4.3.2 Seeding with gametophytes ....................................... 50
2.4.3.3 Hatchery systems ......................................................... 50
37
38 Protocols for Macroalgae Research
2.1 Introduction
The sugar kelp Saccharina latissima (Linnaeus) as discussed by Lane,
Mayes, Druehl, and Saunders (synonym: Laminaria saccharina [Linnaeus]
Lamouroux) is a cold-water species distributed circumpolar in the north-
ern hemisphere (Bolton et al. 1983) where it occurs from the intertidal
down to the bottom of the photic zone. It is present on both sides of the
Atlantic from Nova Scotia to Europe and in the Pacific along the North
American coast and some areas outside Japan and in the Arctic Russia
(Wilce 1965; van den Hoek and Donze 1967; Druehl 1970; Druehl and
Kaneko 1973; Sjøtun 1985). Temperature, exposure, and competition are
the limiting factors of the propagation of the species, and the southern limit
is highly determined by temperature (Lüning 1990). Their natural growth
sites are clear and turbid coastal waters (Borum et al. 2002), and water
temperatures between 10°C and 15°C have been found to give optimum
growth (Fortes and Lüning 1980; Bolton and Lüning 1982). S. latissima is
one of the fastest growing species of kelp in European waters with annual
production capacities estimated at 75–170 tons wet weight per hectare
at sea (Broch et al. 2013). Cultivation of S. latissima is characterized by
fast growth, but with low predictability of production volumes and bio-
mass quality due to seasonal-, regional-, and site-dependent growth and
composition and different cultivation strategies. In cool-temperate North
Atlantic waters, S. latissima shows a seasonal- and depth-dependent
growth pattern with faster growth close to the surface in autumn and
winter and fast growth also at deeper depths in the photic zone in spring
and early summer. From late spring to late summer, heavy fouling and
necrosis of the distal end of the blade occurs. Harvesting should therefore
take place in early summer to avoid destruction of the crop and loss of
biomass (Handå et al. 2013; Førde et al. 2016). Europe’s contribution to
global macroalgae cultivation is neglectable (FAO 2016). However, new
consumer trends, market demands, and opportunities for multiple use
of macroalgae such as food, bioactive components for functional food
and feed ingredients, fertilizers, and biofuels (Shahidi 2009; Holdt and
Chapter two: Cultivation protocol for Saccharina latissima 39
Kraan 2011; Rajapakse and Kim 2011; Fleurence et al. 2012; Hafting et al.
2012; Tabarsa et al. 2012; Wargacki et al. 2012; Fernand et al. 2016) have
strengthened the motivation for industrial macroalgae cultivation in
Europe. Industrial, cost-effective cultivation requires novel technology of
the whole production line targeting mechanization and automation of the
seedling processes, deployment at sea and the harvesting operation, but
the necessary equipment for industrial-scale production does not exist.
Cultivation equipment must be developed for different environmental
conditions, and logistic challenges during deployment and harvesting
must be solved. An in-depth understanding of the life cycles and the
environmental and biological requirements of macroalgae is a prerequi-
site to establish protocols for all phases of the cultivation. In the current
chapter, we present a protocol for year-round production of S. latissima
seedlings that is applicable for small-scale laboratory studies and that can
be upscaled for mass production.
2. Vegetative phase
3. Reproductive phase
Female gametophyte -induced by optimal
Zoospores
Sori conditions
Sporangia Antheridia
Oogonia
Spermatozoid
Egg
Gametophytic
haploid generation
Sporophytic
diploid generation
Developing sporophyte
Mature sporophyte
Figure 2.1 Schematic presentation of the life cycle of S. latissima. The sporophyte
produces unilocular sporangia with meiospores that are released into surround-
ing waters when mature. These develop into male and female gametophytes.
When exposed to blue light, the male gametophytes produce antheridia with
spermatozoids, and the female gametophytes produce oogonia with one egg.
Fertilization leads to development of small sporophytes that grow into the full-
sized sporophytes. (Courtesy of Sanna Matsson.)
and Neushul 1978; Lüning 1980). Once the light and temperature condi-
tions are suitable, the development of reproductive structures is triggered
(Lüning 1980). Female gametophytes produce an oogonium, which when
fertile produces one nonmotile spherical gamete (oosphere/egg). The
male gametophyte develops spermatocysts that produce spermatozoids
with two flagella. A signal chemical attracts the spermatozoids, which
swim to the oogonium and fertilize the egg (Müller et al. 1979; Müller
1989). The fertilized gametes develop into a microscopic sporophyte that
grows into the full-sized sporophyte. The haploid gametophyte stage can
be cultivated under controlled environmental conditions in a laboratory.
Gametophytes are dark tolerant and can survive in the darkness for up
Chapter two: Cultivation protocol for Saccharina latissima 41
to 18 months (Druehl and Boal 1981; tom Dieck 1993). In weak red light,
gametophytes have vegetative growth and will increase in size, without
producing gametes or spermatocysts. Fertilization can be induced at any
moment by modifying the light regime and expose the gametophytes to
blue light. Studies have shown that the ability to produce gametes is still
good after 30 years of cultivation under red-light conditions in many kelp
species (Druehl et al. 2005).
2.3 Materials
2.3.1 Collection of sporophytes and induction of sori
• Boxes or plastic bags
• Coolers (if outdoor or transport temperature is above 15°C)
• Scissors for trimming the sporophytes
• Temperature-regulated room holding 10°C
• Tanks with nutrient-rich seawater and fluorescents lamps for storage
and conditioning of sporophytes
• Time switch
• Pressurized air or electrical aquarium pump for aeration
42 Protocols for Macroalgae Research
2.3.6 Seeding
• Temperature-regulated room with white-light holding 10°C
• Filtered and UV-treated nutrient-rich seawater
• Seeding substrate (string, rope, and sheet)
• Container
• Spray bottle
Figure 2.2 The dark area on the thallus of S. latissima is called sorus and contains
millions of zoospores.
season, this can take from 4 to 12 weeks but normally from 6 to 8 weeks
(Forbord et al. 2012; see Table 2.1 for key parameters).
Tanks of 200–300 L are sufficient for 20–30 sporophytes (Note 2).
Tanks are illuminated 8 h per day with fluorescent lamps providing a
light intensity of around 70–100 µmol photons m−2 s−1 at the water surface.
Chapter two: Cultivation protocol for Saccharina latissima 45
The seawater source should ensure water with a high and stable level of
nutrients and temperatures not exceeding 10°C. Water intake below 50 m
depth may be necessary. The seawater is filtered before use. Aeration is
used to keep the sporophytes suspended and circulating in the water
column so that all individuals are equally illuminated.
2.4.1.3 Dehydration
Disinfected sori are dried with paper towel to remove the surface water
without leaving the surface withered. Dried sori are stored in a plastic
bag/box with lid for 18–48 h at 4°C to dehydrate.
Other counting chambers can be used, for example, Sedgewick Rafter and
Burker’s chamber.
Parafilm. Culture flasks of 250–500 mL are used in the initial stage, and
as the biomass increases, the culture is either split into two or the total
volume of the culture is scaled up, as shown in Figure 2.4a. After mini-
mum four weeks, the growth medium can be renewed the first time, and
to ensure a minimal evaporation rate, the cultures are gently aerated.
The water level is marked on the culture vessel at inoculation so that
evaporation can be monitored and distilled water added in case of heavy
evaporation.
48 Protocols for Macroalgae Research
(a) (b)
Figure 2.4 (a) Gametophyte cultures after two months of inoculation and
(b) gametophytes grown under red-light condition for 32 days (20× magnification).
9. Label the flasks with origin and date and mark the water level.
10. Place the flasks back in the red-light culture room/climate cabinet.
11. Allow gentle aeration so that the gametophytes stay suspended.
2.4.3 Seeding
Different kind of substrates can be used for seeding (strings, ropes, and sheets)
with different materials (nylon, polypropylene, polyester, and polyester silk),
and the ropes and strings can be both twisted and braided. This protocol is
adapted to cultivation on strings (1–2 mm) and ropes (5–10 mm) (Note 8).
50 Protocols for Macroalgae Research
(a) (b)
Figure 2.5 (a) Seedling cultivation systems with cylinders and (b) seedling culti-
vation systems with tanks.
(a) (b)
Figure 2.6 (a) Four-week-old juvenile seedlings grown on a thin string (1, 2 mm
twisted nylon) and (b) spools with seedlings ready for deployment at sea after
six weeks in the hatchery.
if the conditions are optimal, an egg can be seen as early as on day 8 after
seeding. The egg looks similar to the primary cell but is distinguished by
its larger size and the empty, transparent membrane of the primary cell left
behind it. The empty membrane has the same size and shape as the egg cell
and is the best indication of the cell being an egg and not a primary cell.
Fertilized eggs will develop rapidly into juvenile sporophytes. The first cell
divisions are usually horizontal, and at the size of approximately five cells,
the cells start to divide vertically. Figure 2.7 shows the development from
spores to seedlings under optimal lab conditions, but one can clearly see
the development of contaminating diatoms as the sori have not been disin-
fected prior to spore release. In commercial production, the development
might take slightly more time as larger cultivation systems normally give
less degree of control and evenness in the system.
2.4.4 Deployment
2.4.4.1 Transport
Seedlings must be protected from dehydration and extreme temperatures
during transport. Spools and ropes are therefore wrapped in a thin layer
of plastic to avoid evaporation of surface water and stored in coolers if the
ambient temperature is below 0°C or above 15°C. Transportation of seed-
lings over a period up to 12 h have shown good results.
Figure 2.7 Development of spores to sporophytes under optimal lab conditions, but without any sorus disinfection before spore
release. Pictures are taken regularly over 21 days (day 3, 6, 8, 11 [picture 1–4, upper row], 14, 16, 18, and 21 [picture 4–8 lower row]
post seeding). Pictures 1–2 show primary cells; in picture three eggs are clearly visible and pictures 4–8 show the development of
juvenile sporophytes and contaminating diatoms.
Protocols for Macroalgae Research
Chapter two: Cultivation protocol for Saccharina latissima 55
(a) (b)
(c)
Figure 2.8 (a) Twisting machine with thick rope in the center and seeded thin
strings on spools, (b) thick rope with the thin seedling string twisted onto it,
and (c) S. latissima cultivated three months in the sea, from mid-February to
mid-May.
56 Protocols for Macroalgae Research
S. latissima can be deployed from early autumn to late winter before the main
growth season in spring. Figure 2.8c shows S. latissima after three months of
cultivation in the sea. High and predictable productivity and reduced bio-
fouling during sea cultivation claim for novel production technology com-
bined with seasonal and regional cultivation strategies.
2.5 Notes
Note 1: Conditioning tanks are preferably kept separate from the hatch-
ery to avoid transfer of contaminating organisms from sporophytes
to gametophyte cultures and seedlings.
Note 2: The size and shape of the conditioning tanks are of minor
importance for the sori induction.
Note 3: Always use gloves and protecting glasses when working with
NaOCl or other hazardous chemicals and work with good ventila-
tion (fume cupboard or outside).
Note 4: The spore release peaks during the first 35–40 min after ini-
tiation (Arbona and Molla 2006) but can be continued for at least
90 min if spore density is scarce after the first 40 min.
Note 5: Spores are counted in the squares, and a minimum of 4 squares
should be counted if the density is very high, and all 10 squares
should be counted if the density is lower.
Note 6: Red light is acquired either by LED lights (620–750 nm) or by
fluorescent tubes covered in red cellophane or red plexiglass.
Note 7: All equipment is cleaned with hot water and detergent and auto-
claved/sterilized between each culture maintenance.
Note 8: Make sure that the substrate for seeding is hydrophilic (water
loving) so that the spore solution/gametophytes is/are absorbed and
not repelled by the substrate.
Note 9: If you have a low amount of spore solution available after the
spore release or in the case of a very low-spore density, you can col-
lect the spore solution that runs off from the spraying and reuse it or
consider bathing the substrate instead of spraying.
Note 10: Microscope slides are seeded and incubated together with the
spools/ropes to check the development using a light microscope
during the incubation phase.
Note 11: The hatchery normally has activity between August and
February, so in the months between production, the water and cool-
ing systems should be turned off to let the hatchery dry out. Disinfect
or wash all equipment such as ropes, water hoses, flow meters, and
so on, and chlorinate the pipe system one week in advance before
you start the production.
Note 12: Treatment with GeO2 may have a negative effect on the juvenile
sporophytes, causing deformation and reduced growth.
Chapter two: Cultivation protocol for Saccharina latissima 57
References
Arbona J.F and Molla M. 2006. Aquaculture explained: Cultivation of the brown
seaweed Alaria esculenta. In: Watson L (Ed.), Dublin: Bord Iascaigh Mhara,
50 p.
Bolton, J.J., Germann, I., Lüning, K. 1983. Hybridization between Atlantic and
Pacific representatives of the Simplices section of Laminaria (Phaeophyta).
Phycologia 22:133–140.
Bolton, J.J. and Lüning, K. 1982. Optimal growth and maximal survival tem-
peratures of Atlantic Laminaria species (Phaeophyta) in culture. Mar Biol
66:89–94.
Borum, J., Pedersen, M., Krause-Jensen, D., Christensen, P., Nielsen, K. 2002.
Biomass, photosynthesis and growth of Laminaria saccharina in a high-arctic
fjord, NE Greenland. Mar Biol 141:11–19.
Broch, O.J., Ellingsen, I., Forbord, S., Wang, X., Volent, Z., Alver, M.O., Handå, A.
et al. 2013. Modelling the cultivation and bioremediation potential of the
kelp Saccharina latissima in close proximity to an exposed salmon farm in
Norway. Aquac Environ Interact 4:187–206.
Druehl, L.D. 1970. The pattern of Laminariales distribution in the northwest
Pacific. Phycologia 9(3/4):237–247.
Druehl, L.D. and Boal, R. 1981. Manipulation of the laminarialean life-cycle and its
consequences for kombu mariculture. Proc Internat Seaweed Symp 6:161–170.
Druehl, L.D., Collins, J.D., Lane, C.E., Saunders, G.W. 2005. An evaluation of
methods used to assess intergeneric hybdridization in kelp using Pacific
Laminariales (Phaeophyceae). J Appl Phycol 41:250–262.
Druehl, L.D. and Kaneko, T. 1973. On the Laminaria saccharina from Hokkaido. Bot
Mag Tokyo 86:323–326.
Edwards, M. and Watson, L. 2011. Aquaculture Explained Cultivating Laminaria
Digitata. Irish Sea Fisheries Board, Dunlaoghaire, Ireland.
FAO. 2016. The state of world fisheries and aquaculture 2016. Contributing to Food
Security and Nutrition for All. Food and Agricultural Organization of the
United Nations, Rome, Italy, 200 pp.
Fernand, F., Israel, A., Skjermo, J., Wichard, T., Timmermann, K.R., Golberg, A.
2016. Offshore macroalgae biomass for bioenergy production: Environmental
aspects, technological achievements and challenges. Renew Sus Energ Rev
doi: 10.1016/j.rser.2016.10.046.
Fleurence, J., Morancais, M., Dumay, J., Decottignies, P., Turpin, V., Munier, M.,
Garcia-Bueno, N., Jaouen, P. 2012. What are the prospects for using seaweed
in human nutrition and for marine animals raised through aquaculture?
Trends Food Sci Technol 27:57–61.
Forbord, S., Skjermo, J., Arff, J., Handå, A., Reitan, K.I., Bjerregaard, R., Lüning, K.
2012. Development of Saccharina latissima (Phaeophyceae) kelp hatcheries
with year-round production of zoospores and juvenile sporophytes on
culture ropes for kelp aquaculture. J Appl Phycol 24:393–399.
Førde, H., Forbord, S., Handå, A., Fossberg, J., Arff, J., Johnsen, G., Reitan, K.I. 2016.
Development of bryozoan fouling on cultivated kelp (Saccharina latissima) in
Norway. J Appl Phycol 28:1225–1234.
Fortes, M.D. and Lüning, K. 1980. Growth rates of North Sea macroalgae in rela-
tion to temperature, PAR and photoperiod. Helgoländer Meeresuntersuchungen
34:15–29.
58 Protocols for Macroalgae Research
Hafting, J.T., Critchley, A.T., Cornish, M.L., Hubley, S.A., Archibald, A.F. 2012.
On-land cultivation of functional seaweed products for human usage. J Appl
Phycol 24:385–392.
Handå, A., Forbord, S., Wang, X., Broch, O.J., Dahle, S.W., Størseth, T.S., Reitan, K.I.,
Olsen, Y., Skjermo, J. 2013. Seasonal- and depth-dependent growth of culti-
vated kelp (Saccharina latissima) in close proximity to salmon (Salmo salar)
aquaculture in Norway. Aquaculture 414–415:191–201.
Harrison, P.J. and Berges, J.A. 2005. Marine culture media. In: Algal Culturing
Techniques R. Andersen (Ed.), pp. 21–33. Academic Press, New York.
Holdt, S. and Kraan, S. 2011. Bioactive compounds in seaweed: Functional food
applications and legislation. J Appl Phycol 23:543–597.
Hollenberg, G.J. 1939. Culture studies of marine algae. I. Eisenia arborea. Amer J Bot
26:34–41.
Joska, M. and Bolton, J. 1987. In situ measurement of zoospore release and sea-
sonality of reproduction in Ecklonia maxima (Alariaceae, Laminariales). Brit
Phycol J 22:209–214.
Kain, J.M. 1975. The biology of Laminaria Hyperborea VII. Reproduction of the spo-
rophyte. J Mar Biol Assoc U.K. 55:567–582.
Kain, J.M. 1979. A view of the genus Laminaria. Oceanogr Mar Biol Ann Rev
17:101–161.
Lüning, K. 1980. Critical levels of light and temperature regulating gametogenesis
of three Laminaria species. J Phycol 16:1–15.
Lüning, K. 1990. Seaweeds. Their Environment, Biogeography, and Ecophysiology.
Wiley, New York.
Lüning, K. and Dring, M.J. 1975. Reproduction, growth and photosynthesis of
gametophytes of Laminaria saccharina grown in blue and red light. Mar Biol
29:195–200.
Lüning, K. and Neushul, M. 1978. Light and temperature demands for growth
and reproduction of laminarian gametophytes in southern and central
California. Mar Biol 45:297–309.
Müller, D.G. 1989. Sexuality and sexual attraction. In: Algae as Experimental Systems,
A.W. Coleman, L.J. Goff, and J.R. Stein-Taylor (Eds.), pp. 201–213. A.R. Liss,
New York.
Müller, D.G., Gassmann, G., Lüning, K. 1979. Isolation of a spermatozoid—
Releasing and attracting substance from female gametophytes of Laminaria
digitata. Nature 279:430–431.
Pang, S. and Lüning, K. 2004. Breaking seasonal limitation: Year-round sporo-
genesis in the brown alga Laminaria saccharina by blocking the transport of
putative sporulation inhibitors. Aquaculture 240:531–541.
Rajapakse, N. and Kim, S.K. 2011. Nutritional and digestive health benefits of sea-
weed. Adv Food Nutr Res 64:17–28.
Redmond, S., Green, L., Yarish, C., Kim, J., Neefus, C. 2014. New England Seaweed
Culture Handbook-Nursery Systems. Connecticut Sea Grant CTSG-14-01. 92 p.
http://nsgl.gso.uri.edu/nhu/nhuh14001.pdf.
Rød, K.K. 2012. Sori disinfection in cultivation of Saccharina latissima—Evaluation
of chemical treatments against diatom contamination. Master thesis,
Norwegian University of Science and Technology, Trondheim, Norway.
Sauvageau, C. 1916. Sur les gametophytes de deux Laminaires. C R Acad Sci
56:1–240.
Chapter two: Cultivation protocol for Saccharina latissima 59
Contents
3.1 Introduction ............................................................................................. 62
3.2 State of the art .......................................................................................... 63
3.3 Materials................................................................................................... 64
3.3.1 Facilities and equipment for collection of sporogenous
material and release of spores................................................... 64
3.3.1.1 In the field ..................................................................... 64
3.3.1.2 In the laboratory ........................................................... 65
3.3.2 Facilities and equipment list for isolation and
propagation of clonal cultures .................................................. 65
3.3.3 Facilities and equipment list for performing
laboratory-scale hybridization programs ................................ 66
3.4 Experimental procedures ...................................................................... 66
3.4.1 Collection and preparation of fertile sporophytes ................. 66
3.4.2 Induced spore release ................................................................. 66
3.4.3 Conditions to keep gametophytes vegetative ......................... 69
3.4.4 Isolation of clonal gametophyte cultures ................................ 70
3.4.5 Propagation of clonal cultures .................................................. 71
3.4.6 Hybridization of clonal gametophyte cultures ...................... 72
3.5 Notes ......................................................................................................... 75
Acknowledgment ............................................................................................. 76
References.......................................................................................................... 76
61
62 Protocols for Macroalgae Research
3.1 Introduction
The term kelp is the common name of all species of the marine brown algal
order Laminariales, but as common names often do not have a precise
definition, in the literature, it is partially also used for other big brown
algae of the order Fucales or Desmarestiales, all of which form coastal
underwater forests. Here, we only deal with kelps in the restricted defini-
tion, that is, the Laminariales. Kelps thrive along rocky shores of polar to
cold-temperate waters worldwide, or more seldom in cooler deep-waters
of warm-temperate to tropical sites, such as the Mediterranean Sea or the
Galapagos islands (Lüning 1990; Bolton 2010). Besides their considerable
ecological role as ecosystem engineering species, a few taxa, especially
of the genera Laminaria, Saccharina, Undaria, and Macrocystis, are of major
economic importance, either being harvested in the field or cultivated,
the latter particularly in Asia and in Chile (FAO 2016). Interest in kelps
and kelp cultivation as a resource for food products, alginates, and biofuel
production among others is growing steadily and thus strain preserva-
tion, in combination with genetic engineering of strains or classical breed-
ing programs, may become more and more important in the future. This
is especially significant under the threat of global warming where wide
stretches of coasts currently harboring seaweed aquacultures will become
tropical to subtropical (Bartsch et al. 2012) and may require warm-tolerant
cultivars.
One prerequisite for the easy selection and maintenance of strains is
the life cycle of Laminariales. Kelps are diploid macroscopic algae, which
consist of a lamina (blade), stipe (stem), and haptera (holdfast), and are
mostly perennial. When they become fertile, groups of sporangia (sorus)
develop either on the main blade or in some species on smaller special
blades, the so-called sporophylls. During spore formation, meiosis takes
place resulting in mobile isomorph haploid male and female spores of
approximately 4–8 µm length. After spore germination, single-celled
gametophytes may either directly enter reproduction (gametogenesis) or
initiate vegetative growth depending on environmental conditions. The
latter process is used to derive clonal stock cultures for multiuse purposes.
For a scheme of the life cycle and further details, see Chapter 2 by Forbord
et al. (2017) and Bartsch et al. (2008). The capacity of microscopic clonal
gametophytes to grow and preserve their ability for sexual reproduction
even after >30 years of maintenance (Druehl et al. 2005; Bartsch, pers.
comm.) is a prerequisite for the long-term preservation of clonal male and
female gametophytes.
The current chapter elucidates kelp cultivation aspects that have not
been addressed in Chapter 2. The skilled researcher or application man-
ager will be able to combine both protocols for specific needs. General
seed supply and sporulation protocols described in Chapter 2 are a good
Chapter three: Derivation of clonal stock cultures 63
3.3 Materials
In general, all listed materials should be clean or where possible sterile.
Figure 3.1 Sporophylls of Alaria esculenta showing different stages of sorus rip-
ening. Scale bar = 1 cm.
The smaller the piece of sorus is, the higher the probability that it is not
contaminated by diatoms or other organisms.
1. Cut out sorus; small pieces of ripe sori are sufficient for the purpose.
2. Clean sorus with tissue paper wetted with distilled water and wipe
dry. Chemical disinfection is not needed.
3. Avoid material that is infected by endophytes or epiphytes
(Figure 3.2) or shows signs of biofouling.
4. Store the sorus in a humid chamber (e.g., by addition of wet paper
towels) overnight at low temperatures (e.g., 4°C in the refrigerator).
Adapt size of the chamber to the size of the sorus. In our case, a Petri
dish is sufficient. Take care that the sorus does not come into contact
with freshwater (Note 3).
5. Rehydrate sorus piece in a clean Petri dish with sterile seawater
(10°C–15°C) the next day.
6. Subject to light and relatively warm but nonlethal temperatures
(~15°C). Spores are usually released within 10–30 min.
7. After spore release, remove the sorus and discard.
68 Protocols for Macroalgae Research
The shorter the release period, the less chance there is for any unwanted
contamination. In contrast to the protocol by Forbord et al. 2017 (Chapter 2),
a mass release of spores is not essential.
During expeditions/field—work without direct laboratory access, the
following protocol may be useful to collect spores:
50 μm
Figure 3.3 Germinated kelp spores in a density that should ideally be less than
shown for isolation purposes.
also induce gametogenesis (Motomura and Sakai 1981; Lewis et al. 2013).
Thus, the nutrient medium is ideally iron-free and, during the first four
weeks, should not be exchanged at all. Later on, medium change is suffi-
cient once per month. As long-term exposition to GeO2 may also be harm-
ful to kelps, exchange the initial seawater after approximately four weeks,
and only add P and N according to the protocol of Zhang et al. (2008) or
≤ ½ PES. The application of relatively high temperatures is another pos-
sibility to inhibit gametogenesis. Kelp gametophytes normally do not
become fertile if exposed to temperatures ~4°C lower than their upper
survival limit (e.g., tom Dieck and Oliveira 1993).
• Draw out glass Pasteur pipettes to get a narrower mouth or just melt
the tip of the glass pipette to smooth the outer margin of the mouth
or take a microcapillary tube. Avoid forceps for isolation purposes.
Even fine forceps are not suitable as the gametophytes easily stick to
them.
• Identify well-separated male and female gametophytes.
• Select one single gametophyte with your pipette.
• Transfer the droplet of water with the single gametophyte into a new
sterile Petri dish.
• Scrutinize this droplet under the (inverted) microscope to verify
that you selected a single gametophyte and verify the sex.
• Fill up with sterile ½ PES (ideally iron free) or sterile seawater just
enriched with N and P (Zhang et al. 2008).
• Seal with Parafilm, label, and replace into conditions allowing for
vegetative growth.
• Repeat this process with the opposite sex so that you generate at
least one clonal pair of gametophytes per parental sporophyte.
(a) (b)
(c) (d)
(e)
Figure 3.4 Vegetative clonal gametophytes and their propagation. (a) Red light
grown gametophytes, (b) vegetative gametophyte cushions, (c) sterile mortar with
small vegetative gametophyte cushions, (d) carefully fragmented gametophytes,
and (e) fragmented gametophytes filled up with seawater for sowing.
72 Protocols for Macroalgae Research
Figure 3.5 Saccharina latissima: Microscopic sporophytes (with rhizoids) and pre-
sumable parthenosporophytes (deformed sporophytes without rhizoids). Scale
bar = 50 µm.
74 Protocols for Macroalgae Research
3.5 Notes
Note 1: Advantages and disadvantages of clonal gametophytes:
Clonal gametophytes are rather easy to obtain and to maintain for
requirements of basic research and commercial applications. The
advantages of clonal gametophytes are that (1) they can be kept
for decades, (2) probably remain genetically relatively stable (final
evidence is still missing), and (3) sporophytes can be initiated at
any time of the year and can thus uncouple seeding from the sea-
sonal availability of spores. The disadvantages of clonal gameto-
phytes are that (1) much time is needed for their establishment, that
(2) they have to be protected from fatal contamination or disease,
and that (3) care has to be taken that clones stay vegetative but
grow sufficiently.
Note 2: Fertility of sporophytes: The season of sporophyte fertility is
different from species-to-species (see, e.g., Bartsch et al. 2008). Most
species are fertile during autumn to winter, some over summer, and
in some species/regions, fertile material may be found year-round
but with different intensities. In instances where environmental
76 Protocols for Macroalgae Research
conditions have been stressful, it may occur that sori looking per-
fect do not release viable spores, and germination capacity is low.
This has been shown for L. digitata in summer (Bartsch et al. 2013).
Note 3: Spore release methods: Each laboratory has its own methods
to release spores. Some protocols keep cleaned sori for 2 h in the
air, whereas others keep them for two days in the refrigerator with-
out the wet chamber. The disadvantage of the forced spore release
procedure is that not only ripe but also unripe sporangia burst and
release healthy motile and unripe spores with no swimming activ-
ity. The motility status of spores should thus be checked under the
microscope after release. In some cases, the tissue may release con-
siderable amounts of mucus (alginate) that can be harmful to the
spores and may require immediate attention (e.g., filtration through
net-gauze or dilution with sterile seawater or the transfer of the
spore solution into new tubes/Petri dishes).
Acknowledgment
I thank Klaus Lüning who initiated my interest in kelps many years ago,
provided the initial training in the described methods, and fostered my
early career. I thank Bertrand Jacquemin for introducing me to the spore
capture method in the Falcon tubes without induced spore release. I thank
A. Wagner and D. Liesner for checking of inconsistencies and J. Bartsch
for polishing the English of the manuscript.
References
Bartsch, I., J. Vogt, C. Pehlke, and D. Hanelt. 2013. Prevailing sea surface tem-
peratures inhibit summer reproduction of the kelp Laminaria digitata at
Helgoland (North sea). J. Phycol. 49:1061–1073.
Bartsch, I., C. Wiencke, K. Bischof et al. 2008. The genus Laminaria sensu lato:
Recent insight and development. Eur. J. Phycol. 43:1–86.
Bartsch, I., C. Wiencke, and T. Laepple. 2012. Global seaweed biogeography under
a changing climate: The prospected effects of temperature. In Seaweed
Biology. Novel Insights into Ecophysiology, Ecology and Utilization, Wiencke, C.
and Bischof, K. (Eds.), Springer, Heidelberg, Germany, pp. 383–406.
Bolton, J.J. 2010. The biogeography of kelps (Laminariales, Phaeophyceae):
A global analysis with new insights from recent advances in molecular
phylogenetics. Helgol. Mar. Res. 64:263–279.
Bolton, J.J., I. Germann, and K. Lüning. 1983. Hybridization between Atlantic and
Pacific representatives of the Simplices section of Laminaria (Phaeophyta).
Phycologia 22:133–140.
Buchholz, C. and K. Lüning. 1999. Isolated, distal blade discs of the brown alga
Laminaria digitata form sorus, but not discs near to the meristematic transi-
tion zone. J. Appl. Phycol. 11:579–584.
Chapter three: Derivation of clonal stock cultures 77
Druehl, L.D., J.D. Collins, C.E. Lane, and G.W. Saunders. 2005. An evaluation of
methods used to assess intergeneric hybridization in kelp using Pacific
Laminariales (Phaeophyceae). J. Phycol. 41:250–262.
Fang, T.C., J.H. Tai, Y.L. Ou, C.C. Tui, and T.C. Chen. 1978. Some genetic observa-
tions on the monoploid breeding of Laminaria japonica. Sci. Sin. 21:401–408.
Food and Agriculture Organization of the United Nations. 2016. Fisheries and
Aquaculture Information and Statistics Services. Retrieved from http://www.
fao.org/figis/ (accessed May 3, 2016).
Forbord, S., K.B. Steinhovden, K.K. Rød, A. Handå, and J. Skjermo. 2017. Cultivation
protocol for Saccharina latissima. In Protocols for Macroalgae Research, B.
Charrier, T. Wichard, and C.R.K. Reddy (Eds.), Chapter 2. Boca Raton, FL:
CRC Press, Taylor & Francis Group.
Funano, T. 1980. Crossing experiments between several species of Laminaria in
Hokkaido. Hokusuishi-Geppo 37:181–207 (in Japanese).
Hwang, E.K., Y.G. Gong, and C.S. Park. 2012. Cultivation of a hybrid of free-living
gametophytes between Undariopsis peterseniana and Undaria pinnatifida:
Morphological aspects and cultivation period. J. Appl. Phycol. 24:401–408.
Kraan, S., A.V. Tramullas, and M.D. Guiry. 2000. The edible brown seaweed Alaria
esculenta (Phaeophyceae, Laminariales): Hybridization, growth and genetic
comparisons of six Irish populations. J. Appl. Phycol. 12:577–583.
Lewis, R.J. and M. Neushul. 1994. Northern and southern hemisphere hybrids of
Macrocystis (Phaeophyceae). J. Phycol. 30:346–353.
Lewis, R.J. and M. Neushul. 1995. Intergeneric hybridization among five genera
of the family Lessoniaceae (Phaeophyceae) and evidence for polyploidy in a
fertile Pelagophycus × Macrocystis hybrid. J. Phycol. 31:1012–1017.
Lewis, R.J., M. Green, and M.E. Azfal. 2013. Effects of chelated iron on oogenesis and
vegetative growth of kelp gametophytes (Phaeophyceae). Phycol. Res. 61:46–51.
Lewis, R.J., B. Jiang, M. Neushul, and X.G. Fei. 1993. Haploid parthenogenetic spo-
rophytes of Laminaria japonica (Phaeophyceae). J. Phycol. 29:363–369.
Li, X., Y. Cong, G. Yang et al. 2007. Trait evaluation and trial cultivation of Dongfang
No. 2, the hybrid of a male gametophyte clone of Laminaria longissima
(Laminariales, Phaeophyta) and a female one of L. japonica. J. Appl. Phycol.
19:139–151.
Li, X., Z. Zhang, S. Qu et al. 2016. Breeding of an intraspecific kelp hybrid
Dongfang no. 6 (Saccharina japonica, Phaeophyceae, Laminariales) for suit-
able processing products and evaluation of its culture performance. J. Appl.
Phycol. 28:439–447.
Loureiro, R., C.M.M. Gacon, and C. Rébours. 2015. Seaweed cultivation: Potential
and challenges of crop domestication at an unprecedented pace. New
Phytologist 206:489–492.
Lüning, K. 1981. Egg release in gametophytes of Laminaria saccharina: Induction by
darkness and inhibition by blue light and UV. Br. Phycol. J. 16:579–593.
Lüning, K. 1990. Seaweeds: Their Environment, Biogeography, and Ecophysiology. New
York: Wiley.
Lüning, K. and M.J. Dring. 1975. Reproduction, growth and photosynthesis of
gametophytes of Laminaria saccharina grown in blue and red light. Mar. Biol.
29:195–200.
Motomura, T. and Y. Sakai. 1981. Effect of chelated iron in culture media on oogen-
esis in Laminaria angustata. Bull. Japan Soc. Sci. Fish. 47:1535–1540.
78 Protocols for Macroalgae Research
Cryopreservation of macroalgae
John G. Day
Contents
4.1 Introduction.............................................................................................. 79
4.2 State of the art .......................................................................................... 82
4.3 Materials ................................................................................................... 86
4.3.1 Materials required for both methods ....................................... 86
4.3.2 Materials for conventional colligative cryopreservation ....... 86
4.3.3 Materials required for a vitrification-based approach........... 87
4.4 Experimental procedures ....................................................................... 87
4.4.1 Method for conventional colligative cryopreservation.......... 87
4.4.2 Method for a vitrification-based approach .............................. 88
4.5 Notes.......................................................................................................... 90
Acknowledgment ............................................................................................. 91
References.......................................................................................................... 92
4.1 Introduction
Cryopreservation is most usually considered to be the storage of living
cells at ultra-low cryogenic temperatures, most commonly in liquid, or
vapor phase, liquid nitrogen (–196°C), and it has achieved a status of rou-
tine and confident application for many organisms (Fuller et al. 2004).
However, in the vast majority of cases, purely plunging samples straight
into liquid nitrogen will result in total disruption of cellular and intra-
cellular architecture and function. Therefore, methodologies have been
developed, which reduce or prevent cryoinjury; these can be categorized
under two main approaches: (1) traditional controlled cooling-rate freez-
ing and (2) vitrification (Figure 4.1).
In the first approach, cells are cryoprotected using colligative, pen-
etrating cryoprotectants, often described as cryoprotective additives,
which lower the freezing point and ameliorate the damaging effects of
excessive solute concentration (Mazur, 2004). These are commonly used
in association with osmotic additives, which are sometimes applied in
a growth phase, before cryopreservation, in which the freezable water
content is reduced (Withers, 1975). Carefully regulated slow cooling is
79
80 Protocols for Macroalgae Research
Conventional Vitrification
slow cooling approaches
Ice nucleation
Cryogenic
storage Storage Storage
(−196°C)
(a) (b)
Table 4.1 (Continued) Reports of successful cryopreservation of macroalgae with storage at −196°C
Alga Procedure Storage duration Reported viabilitya Reference
L. longissima 37%–60%
Kjellmaniella crassifolia 8%–67%
Ecklonia stolonifera 28%–60%
E. kurome 1%–42%
Undaria pinnatifida 1%–60%
Gametophytic cells of Eisenia Conventional two-step 200 days 27% Male Kono et al. 1998
bicyclis cooling + CPA 31% female
Gametophytes of Undaria Conventional two-step Renard et al. 1992
pinnatifida cooling + CPA
Sporelings and apical Conventional two-step >1 h 0%–93% van der Meer and
segments of mature thalli of cooling + CPA Simpson, 1984
the marine red alga Gracilaria
tikvahiae
Gracilaria foliifera Thallus 36%
Sporelings 43%
Devaleraea ramentaceae Thallus 100%
Sporelings 100%
Palmaria palmata Thallus 100%
Sporelings 100%
Chondrus crispus Thallus 70%
Sporelings 86%
Ulva lactuca Thallus 100%
(Continued)
Protocols for Macroalgae Research
Chapter four:
Table 4.1 (Continued) Reports of successful cryopreservation of macroalgae with storage at −196°C
Alga Procedure Storage duration Reported viabilitya Reference
Enteromorpha intestinalis Conventional two-step >1 week 100% Fleck, 1998
thallus cooling + CPA
Gametophytic thalli of One-step plunge in >80% Choi et al. 2013
Porphyra yezoensis cryostraw + CPA
Protoplasts of Porphyra Conventional vitrification 2 days 67% Liu et al. 2004
yezoensis with a range of
vitrification solutions
Undaria pinnatifida Encapsulation 31% Male Wang et al. 2011
gametophytes dehydration with PVS2, 26% female
Cryopreservation of macroalgae
vitrification
Gametophytes of Laminaria Encapsulation 25%–75% Vigneron et al. 1997
digitata dehydration in sucrose
followed by slow cooling
then plunge
a In many cases, these data are on the basis of positive assessment using a vital and/or a mortal stain to differentiate between live and dead cells
rather than the capacity of individual spores, cells, or thalli to regrow. The available data are from samples assessed at least 24 h after thawing/
rewarming, as materials immediately post-thaw will inevitably provide an overestimate of viability levels.
85
86 Protocols for Macroalgae Research
4.3 Materials
4.3.1 Materials required for both methods
• Incubator/growth cabinet, a class I biological safety cabinet/laminar
flow cabinet
• Appropriate cryosafety personal protective equipment, including
goggles/face shield/visor, gloves (rated for liquid nitrogen emer-
sion), and long cryo apron
• Disposable pipettes/disposable plastic Pasteur pipettes (PastettesTM)
• 2 mL cryogenic tubes
• A small (~1 L) dewar
• A heated water bath
• Storage cryostat/refrigerator with an appropriate inventory
system
• Long forceps
• Culture medium: Modified Provasoli, f/2, or another appropriate
medium
• 70% (v/v) ethanol
• Liquid nitrogen
(+40°C) to rewarm for 2–3 min (do not completely cover the cryovials
in water). Carefully, wipe the cap and tube with a tissue containing
70% ethanol to ensure sterile conditions.
14. Transfer the (now very sticky) beads from the cryovial (with a small
spoon spatula) to 5 mL of culture medium for 1 h (for bead pre-
swelling and rehydration), and then transfer each bead (10 beads
per cryovial) into one square of the (5 × 5 squares) Petri dish (Biddy
Sterilin, or equivalent), with each square containing 1 mL of fresh
medium. Alternatively, place beads in a small Petri dish containing
fresh sterile medium. Then cover in aluminum foil and relabel with
strain designation and date (Note 9).
15. Incubate under the standard culturing temperature for the cryo-
preserved organism; after 24 h, partially remove the foil, and
after another 24–96 h, remove all the foil coverings (Notes 10 and 14).
16. After one week, using standard aseptic techniques, replace the
medium (Note 11).
17. After an appropriate period (2–8 weeks, depending of the strain),
a normal culture should be obtained. This may be maintained by
routine serial transfer, or employed for experimental use.
4.5 Notes
Note 1: Alternatively, for many materials, a passive freezer unit such as
Mr FrostyTM may be used. These units have usually been designed to
have a nonlinear cooling rate of ~1°C min−1 when placed in a −80°C
freezer. Follow the manufacturer’s instructions provided as these
may vary between units.
Note 2: Exchange culture medium once a week prior to cryopreserva-
tion, to maximize culture quality and minimize the bacterial num-
bers in nonaxenic strains. This is to prevent overgrowth of the alga
by bacteria stimulated by nutrient released on cutting the algal thalli.
All algae interact both positively and negatively with their associated
bacterial flora. For some taxa, if the associated flora are removed,
altered, or diminished, growth and phenotypic abnormalities may
be observed (Wichard and Oertel, 2010; Spoerner et al. 2012).
Note 3: At the CCAP, 15 vials are normally filled and processed (see
Note 6).
Note 4: In general, mechanical seeding of ice has not been used for
algal cryopreservation, but if it is an option in the system employed,
empirical experimentation may be used to ascertain if it results in
higher post-thaw viability.
Note 5: It is essential that the cryovials are not allowed to warm up
before plunging into liquid nitrogen.
Chapter four: Cryopreservation of macroalgae 91
Acknowledgment
The author acknowledges National Capability funding from the UK
Natural Environment Research Council for the CCAP. In addition, he
would like to thank Roland Fleck (King’s College London) for provid-
ing the images used in Figure 4.2 and Rachel Saxon (SAMS) for technical
assistance in creating the figure.
92 Protocols for Macroalgae Research
References
Arbault, S. and Delanoue, G. 1994. Cryopreservation of Porphyra linearis
Conchocelis phase. Cryptogamie Algologie 15:65–72.
Arbault, S., Renard, P., Perez, R., and Kaas, R. 1990. Cryopreservation trials on
gametophytes of the food alga Undaria pinnatifida (Laminariales). Aquatic
Living Resources 3:207–215.
Benson, E. E. 2004. Cryoconserving algal and plant diversity: Historical perspec-
tives and future challenges. In Life in the Frozen State, B. Fuller, N. Lane, and
E. E. Benson (Eds.), pp. 299–328. Boca Raton, FL: CRC Press.
Bodas, K., Brennig, C., Diller, K. R., and Brand, J. J. 1995. Cryopreservation of blue-
green and eukaryotic algae in the culture collection at the University of
Texas at Austin. CryoLetters 16:267–274.
Choi, Y. H., Nam, T. J., and Kuwano, K. J. 2013. Cryopreservation of gametophytic
thalli of Porphyra yezoensis (Rhodophyceae) by one-step fast cooling. Journal
of Applied Phycology 25:531–546.
Day, J. G. 1998. Cryo-conservation of microalgae and cyanobacteria. CryoLetters
Supplement 1:7–14.
Day, J. G., Benson, E. E., Harding, K., Knowles, B., Idowu, M., Bremner, D.,
Santos, F. et al. 2005. Cryopreservation and conservation of microalgae: The
development of a pan-european scientific and biotechnological resource
(the COBRA project). CryoLetters 26:231–238.
Day, J. G. and Brand, J. J. 2005. Cryopreservation methods for maintaining cultures.
In Algal Culturing Techniques, R. A. Andersen (Ed.), pp. 165–187. New York:
Academic Press.
Dumet, D. J., Block, W., Worland, R., Reed, B. M., and Benson, E. E. 2000. Profiling
cryopreservation protocols for Ribes ciliatum using differential scanning
calorimetry. CryoLetters 21:367–378.
Elster, J., Lukavský, J., Harding, K., Benson, E. E., and Day, J. G. 2008. Deployment
of the encapsulation/dehydration protocol to cryopreserve polar micro-
algae held at the Czech Republic academy of sciences institute of botany.
CryoLetters 29:27–28.
Engelmann, F. and Takagi, T. 2000. Cryopreservation of Tropical Plant Germplasm;
Current Research Progress and Application. JIRCAS International Agriculture
Series No. 8. Rome, Italy: IPGRI.
Fabre, J. and Dereuddre, J. 1990. Encapsulation-dehydration: A new approach to
cryopreservation of potato shoot-tips. CryoLetters 11:413–426.
Fleck, R. A. 1998. The Assessment of Cell Damage and Recovery in Cryopreserved
Freshwater Protists. PhD thesis. Dundee, Scotland: University of the Abertay
Dundee.
Fuller, B., Lane, N., and Benson, E. E. 2004. Life in the Frozen State. Boca Raton, FL:
CRC Press.
Harding, K., Day, J. G., Lorenz, M., Timmerman, H., Friedl, T., Bremner, D. H., and
Benson, E. E. 2004. Introducing the concept and application of vitrification
for the cryo-conservation of algae- a mini-review. Nova Hedwigia 79:207–226.
Harding, K., Müller, J., Lorenz, M., Timmerman, H., Friedl, T., Day, J. G., and
Benson, E. E. 2008. Deployment of the encapsulation/dehydration proto-
col to cryopreserve microalgae held at SammLung von Algenkulturen,
Universität Göttingen Germany. CryoLetters 29:15–20.
Chapter four: Cryopreservation of macroalgae 93
Harding, K., Müller, J., Timmermann, H., Lorenz, M., Day, J. G., and Friedl, T. 2010.
The use of amplified fragment length polymorphism (AFLP) markers to
verify the identity and genetic stability of euglenoids cryopreserved using
encapsulation dehydration cryoprotective strategies. CryoLetters 31:460–472.
Heesch, S., Day, J. G., Yamagishi, T., Kawai, H., and Küpper, F. C. 2012.
Cryopreservation of the model alga Ectocarpus (Phaeophyceae). CryoLetters
33:327–336.
Kono, S., Kuwano, K., and Saga, N. 1998. Cryopreservation of Eisenia bicyclis
(Laminariales, Phaeophyta) in liquid nitrogen. Journal of Marine Biotechnology
6:220–223.
Kuwano, K., Argua, Y., and Saga, N. 1993. Cryopreservation of the conchocelis of
the marine alga Porphyra yezoensis Ueda (Rhodophyta) in liquid nitrogen.
Plant Science 94:215–225.
Kuwano, K., Kono, S., Jo, Y. H., Shin, J. A., and Saga, N. 2004. Cryopreservation
of the gametophytic cells of Laminariales (Phaeophyta) in liquid nitrogen.
Journal of Phycology 40:606–610.
Lalrinsanga, P. L., Deshmukhe, G., Chakraborty, S. K., Dwivedi, A., Barman, N.,
and Kumar, N. 2009. Preliminary studies on cryopreservation of vegetative
thalli of some economically important seaweeds of India. Journal of Applied
Aquaculture 21:250–262.
Lee, Y. N. and Nam, K. W. 2016. Cryopreservation of gametophytic thalli of Ulva
prolifera (Ulvales, Chlorophyta) from Korea. Journal of Applied Phycology
28:1207–1213.
Liu, H. Q., Yu, W. G., Dai, J. X., Gong, Q. H., Yang, K. F., and Lu, X. Z. 2004.
Cryopreservation of protoplasts of the alga Porphyra yezoensis by vitrifica-
tion. Plant Science 166:97–102.
Lukešová, A., Hrouzek, P., Harding, K., Benson, E. E., and Day, J. G. 2008.
Deployment of the encapsulation/dehydration protocol to cryopreserve
diverse microalgae held at the Institute of Soil Biology, Academy of Sciences
of the Czech Republic. CryoLetters 29:21–26.
Mazur, P. 2004. Principles of cryobiology. In Life in the Frozen State, B. Fuller,
N. Lane, and E. E. Benson (Eds.), pp. 3–66. Boca Raton, FL: CRC Press.
Morris, G. J. 1976. The Cryopreservation of Chlorella 1. Interactions of cooling, pro-
tective additive and warming rate. Archives of Microbiology 107:57–62.
Renard, P., Arbault, S., Kaas., R., and Perez, R. 1992. A method for the cryo-
preservation of the gametophytes of the food alga Undaria pinnatifida
(Laminariales). Comptes Rendus De I Academie des Science Serie III Life
Science 315:445–451.
Sakai, A. 2004. Plant cryopreservation. In Life in the Frozen State, B. Fuller, N. Lane,
and E. E. Benson (Eds.), pp. 329–346. Boca Raton, FL: CRC Press.
Spoerner, M., Wichard, T., Bachhuber, T., Stratmann, J., and Oertel, W. 2012.
Growth and thallus morphogenesis of Ulva mutabilis (Chlorophyta) depends
on a combination of two bacterial species excreting regulatory factors.
Journal of Phycology 48:1433–1447.
Taylor, R. and Fletcher, R. L. 1998. Cryopreservation of eukaryotic algae—A
review of methodologies. Journal of Applied Phycology 10:481–501.
Taylor, R. and Fletcher, R. L. 1999. A simple method for the freeze-preservation of
zoospores of the green macroalga Enteromorpha intestinalis. Journal of Applied
Phycology 11:257–262.
94 Protocols for Macroalgae Research
Contents
5.1 Introduction ............................................................................................. 95
5.2 State of the art .......................................................................................... 97
5.3 Materials................................................................................................. 100
5.4 Experimental procedures .................................................................... 100
5.4.1 Sampling .................................................................................... 100
5.4.2 DNA extraction ......................................................................... 101
5.4.3 16S rRNA amplification............................................................ 102
5.4.4 16S rRNA amplicon sequencing data analysis ..................... 103
5.4.5 Functional prediction based on 16S rRNA gene
sequencing ................................................................................. 105
5.5 Notes ....................................................................................................... 108
Acknowledgments ..........................................................................................110
References.........................................................................................................110
5.1 Introduction
Evidence shows that marine macroalgae (seaweeds) rely on their associated
microbiome for basic functions, such as growth, development, nutrient sup-
ply, protection, and, in some cases, adaptation and acclimation to environ-
mental stresses (Singh and Reddy 2014; Wichard 2015; Dittami et al. 2016).
The term holobiont was first coined for corals because of their dependency
on associated microbes for survival (Rosenberg et al. 2007), but empirical
evidence broadened this term to other organisms, such as seaweeds (Barott
et al. 2011). It is now widely recognized that it is essential to consider the
entire holobiont (i.e., the host and its associated microbiota) to understand
how seaweeds adapt and evolve (Singh and Reddy 2014; Dittami et al. 2016).
95
96 Protocols for Macroalgae Research
is now being considered in evolutionary studies, and this has also raised
interest from the seaweed industry (Charrier et al. 2017). The way sea-
weed–bacterial associations are established and how microbial recruit-
ment is initiated is still an ongoing discussion. The most accepted theory
relies on a combination of stochastic processes and niche selection consis-
tent with the competitive lottery model as proposed by Burke et al. (2011).
In Burke’s study, bacterial community taxonomic structure was different
between samples and over time but functionally equivalent. Thus, it is
proposed that a set of functional characteristics is required to allow colo-
nization of a certain niche (in this case, the seaweed itself and its own
niche) and that determines which bacteria are recruited from the available
guild. But the specific phylogenetic shifts directly related to environmen-
tal stresses remain to be explained; experimental studies are needed to
fully understand the recruitment process.
Besides their invaluable ecological role as habitat-structuring spe-
cies, seaweeds are rich in biologically active compounds, which led to
their exploitation in several economical areas, including their use as food
resource (Gupta and Abu-Ghannam 2011), source of medicinal and phar-
maceutical components (Smit 2004), and antifouling properties (Qian
et al. 2010). Seaweed-associated microbiome studies can improve seaweed
aquaculture by increasing productivity and fitness (Singh and Reddy
2016) and by preventing diseases, just as manipulation of bacteria associ-
ated with human gut and crops can improve human health and agricul-
tural yield, respectively.
The growing need to understand these vital interactions and the role
played by bacterial communities on seaweed evolution, adaptation, and
development has promoted development of laboratory and informatics
techniques. In the past decades, next-generation sequencing (NGS) and
powerful bioinformatic tools allowed one to unveil the previously hidden
world of seaweeds.
Bacteria diversity
on
Metagenomics
cti
ra
Direct Pyrosequencing, Illumina
t
ex
sequencing MiSeq/HiSeq
A
DN
RNA extraction
Metatranscriptomics
HiSeq
Protein extraction
Bacteria potentiality
To ex
Metaproteomics
m
ta tra
l c ac
Holobiont IEF, SDS-page,
o ti
Bacteria functionality
un
mass spectrometry
po on
(seaweed+associated microbiome)
ds
Metabolomics
NMR,
mass spectrometry
activity
Bacteria
Chapter five: Unraveling seaweeds bacteriomes
Figure 5.1 Schematic diagram representing the different metaomics techniques and respective outputs, used to uncover seaweed
microbiome identity/diversity, potentiality, functionality, and activity. 16S amplicon sequencing, metagenomics, and metatranscrip-
tomics analyses can be achieved using 454 Roche GS FLX+ and Illumina (Mi-Seq or Hi-Seq) sequencing methods. Subsequently,
software programs such as QIIME and Mothur can be used for bacterial community diversity studies, whereas MG-RAST, IMG/M,
METAREP, CAMERA, and MEGAN4 can be used for metagenomes’ functional annotation. Metaproteomic data can be analyzed by
isoelectric focusing (IEF), sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS–PAGE), matrix-assisted laser desorption/
ionization time-of-flight mass spectrometry (MALDI–TOF–MS), and liquid chromatography–mass spectrometry (LC–MS). Bacterial
activity (metabolome) can be assessed by nuclear magnetic resonance spectroscopy and diverse mass spectrometry methods. Here
we describe the sequencing of the 16S amplicon used to determine the diversity of the microbial communities and the identity of
99
their community members. Image and legend are adapted from Singh and Reddy (2016).
100 Protocols for Macroalgae Research
5.3 Materials
• Latex or nitrile gloves
• Scalpel and blades or scissors
• 96% (v/v) ethanol
• Paper tissue
• Permanent marker
• 2 mL tubes (or cryotubes in case you use liquid nitrogen)
• Xpedition™ Lysis/Stabilization Solution (Zymo Research) or liquid
nitrogen
• Storage box for 2 mL tubes
• DNA decontaminating solution (e.g., 10% [v/v] bleach)
• Nitrile gloves
• Micropipettes (different volumes) and their respective pipette tips
with filters
• 96% (v/v) ethanol
• Beaker
• Tweezers
• Scalpel and blades
• Bunsen burner
• Bead-beater
• Ice bath
• Microcentrifuge
• Vortex
• UV light (optional, but recommended)
• Water bath or heating block (Note 1)
Before starting the extraction, make sure that the workspace is clean
by disinfecting all surfaces with bleach or other DNA decontaminating
solution (consider placing pipettes, scalpel, and tweezers under UV light
before the procedure). Prepare a beaker with ethanol in which your han-
dling material (tweezers and scalpel) will be placed. Use gloves during
the entire process.
Sample preparation should be carried out right before extraction,
and transfer of samples into the MO BIO bead-beating tubes should
be performed close to a flame. If samples stored in Xpedition™ Lysis/
Stabilization solution are used, vortex the sampling tubes and transfer
a small piece of tissue into the MO BIO bead-beating tube, followed by
100 µL of the stabilization solution. Samples stored in liquid nitrogen ide-
ally should not be thawed before the tissue transference. Disinfect the
scalpel and tweezer using ethanol and flame in between transfers to avoid
contamination from one sample to another.
After sample preparation, start with step 3 of the MO BIO PowerSoil®
DNA Isolation Kit’s Experienced User Protocol (Note 7).
Qiime
PICRUSt
Metagenomes prediction
Predict_metagenomes.py
Alpha and beta diversity,
rarefaction, taxa abundance,
heatmap, and so on Functional characterization
Categorize_by_function.py
Other software
(e.g., PAST, PRIMER-
E+PERMANOVA, R
[Vegan, Phyloseq])
Figure 5.2 Schematic diagram representing the initial and main steps (along with
the respective scripts) to analyze 16S amplicon sequencing data in QIIME (gray
shaded area) and subsequent functional profiles prediction using PICRUSt (dark-
gray shaded area). Downstream statistical and diversity analysis, and respective
visualization using QIIME, are represented by gray-shaded areas. Other sug-
gested software (with QIIME/PICRUSt generated OTU/KEGG pathways table)
are represented by the white-shaded area.
Once data and metadata are prepared, the following steps can be followed
(also illustrated in Figure 5.2):
In every step, arguments and parameters can be changed to fit the data
and methodologies of choice (for details, see individual QIIME scripts
here: http://QIIME.org/scripts/index.html).
The analysis process described in this point is illustrated and summa-
rized in Figure 5.2. As an example of a possible output from the process
explained above, Figure 5.3 depicts part of the results’ analysis of a study
on bacterial community associated with two different red seaweed spe-
cies (based on 16S rRNA amplicon sequencing).
0.2
Group I
Group III
A. taxiformis Islands
p = 0.001
−0.2
p = 0.002
Group II
A. taxiformis Mainland
−0.4
−0.2 0 0.2 0.4
CAP1
Once the necessary files are in the right format and the adequate
database is used (Note 13), PICRUSt predicts the functional profile of the
seaweed bacterial community. The main steps (scripts) to obtain a pre-
dicted metagenome table are the following:
0.52
0.50
Proportion of sequences assigned to
0.48
Nitrogen Metabolism (%)
0.46
0.44
0.42
0.40
0.38
0.36
0.34
Phyllosphere Rhizoids
(fronds and stolon)
5.5 Notes
Note 1: For the DNA extraction step, the listed materials only refer
to those not provided in the MO BIO’s PowerSoil™ DNA Isolation
Kit.
Note 2: This protocol comprehends total bacterial community (both
endo- and epiphytic bacteria). If one is interested in just sampling the
bacterial biofilm, cotton swabs should be rubbed on the tissue sur-
face and stored as described earlier. For sampling endophytic bacte-
ria, please consider the protocol provided by, for example, Delbridge
et al. (2004), Hollants et al. (2010), Aires et al. (2012).
Note 3: Sampling strategy and labeling will depend on the researcher’s
question/hypothesis. This protocol is designed to sample the entire
individual by considering tissue specificity.
Note 4: Field sampling tends to be messy and makes the required asep-
tic conditions challenging. Organizing all the storage material in
advance facilitates the process and avoids undesirable contamina-
tions. Example: Sampling tubes can be labeled in advance according
to sampling expectations, samples to be preserved in the stabili-
zation solution should be placed in a plastic box and kept vertical
to avoid spills and, when possible, tubes should be isolated using
Parafilm tape.
Note 5: In step 4, if Zymo solution is being used as a preservation
method, it might interact with solution C1 and form a white precipi-
tate. The precipitate should be removed by placing the tubes at 65°C
for 5 min.
Note 6: Immediately place the tubes in the bead-beater for, at least, two
min (the time might depend on the tissue type and facility of disrup-
tion) and resume to step 6 of the experienced user protocol.
Note 7: Extraction process should be performed as quickly as possible
in a sterile environment. To do so, prepare and label all the required
tubes beforehand, keep all the materials you need close by, and keep
the Bunsen burner flame on during the entire extraction. To avoid
undesirable spills and subsequent contamination, in step 15 of the
experienced user protocol for PowerSoil® DNA Isolation Kit, load
600 µL of the supernatant mixed with the C4 buffer onto the spin
filter (instead of the recommended 675 µL). More than three loads
might be needed. Always make a blank extraction to evaluate pos-
sible contamination.
Note 8: PCR specifications should be optimized in each lab according to
the reagents, primers, and the thermocycler machine used. However,
the entire PCR process should always be carried out under sterile
Chapter five: Unraveling seaweeds bacteriomes 109
Acknowledgments
We thank the Phycomorph COST action (FA1406) and acknowledge
the financial support of FCT (SFRH/BPD/107878/2015 (AE), SFRH/
BPD/116774/2016 (TA) and GENEKELP).
References
Aires, T., Marbà, N., Serrao, E. A., Duarte, C. M., Arnaud-Haond, S. 2012. Selective
elimination of chloroplastidial DNA for metagenomics of bacteria associ-
ated with the green alga Caulerpa taxifolia (Bryopsidophyceae). J Phycol 48 (2):
483–490.
Aires, T., Moalic, Y., Serrao, E. A., Arnaud-Haond, S. 2015. Hologenome theory
supported by co-occurrence networks of species-specific bacterial commu-
nities in siphonous algae (Caulerpa). FEMS Microbiol Ecol 91: 1–14.
Aires, T., Serrão, E. A., Engelen, A. H. 2016. Host and environmental specificity
in bacterial communities associated to two highly invasive marine species
(Genus Asparagopsis). Front Microbiol 7: 559.
Allen-Vercoe, E. 2013. Bringing the gut microbiota into focus through microbial
culture: Recent progress and future perspective. Curr Opin Microbiol 16 (5):
625–629.
Apprill, A., Mcnally, S., Parsons, R., Weber, L. 2015. Minor revision to V4 region
SSU rRNA 806R gene primer greatly increases detection of SAR11 bacterio-
plankton. Aquat Microb Ecol 75 (2): 129–137.
Barott, K. L., Rodriguez-Brito, B., Janouškovec, J. et al. 2011. Microbial diversity
associated with four functional groups of benthic reef algae and the reef-
building coral Montastrea annularis. Environ Microbiol 13 (5): 1192–1204.
Bengtsson, M. M., Sjøtun, K., Øvreås, L. 2010. Seasonal dynamics of bacterial bio-
films on the Kelp Laminaria hyperborea. Aquat Microb Ecol 60 (1): 71–83.
Bodenhausen, N., Horton, M. W., Bergelson, J. 2013. Bacterial communities asso-
ciated with the leaves and the roots of Arabidopsis thaliana. PLoS One 8 (2):
e56329.
Bolinches, J., Lemos, M. L., Barja, J. L. 1988. Population dynamics of heterotrophic
bacterial communities associated with Fucus vesiculosus and Ulva rigida in an
estuary. Microb Ecol 15 (3): 345–357.
Burke, C., Kjelleberg, S., Thomas T. 2009. Selective extraction of bacterial DNA
from the surfaces of macroalgae. Appl Environ Microbiol 75 (1): 252–256.
Burke, C., Steinberg, P., Rusch, D., Kjelleberg, S., Thomas, T. 2011. Bacterial com-
munity assembly based on functional genes rather than species. PNAS
108 (34): 14288–14293.
Chapter five: Unraveling seaweeds bacteriomes 111
Caporaso, J. G., Kuczynski, J., Stombaugh, J. et al. 2010. Correspondence QIIME
allows analysis of high-throughput community sequencing data intensity nor-
malization improves color calling in SOLiD sequencing. Nature 7 (5): 335–336.
Charrier, B., Coates, J. C., Stavridou, I. 2017. Surfing amongst oil-tankers:
Connecting emerging research fields to the current international landscape.
Trends Plant Sci 22 (1): 1–3.
Chisholm, J. R. M., Dauga, C., Ageron, E., Grimont, P. A. D., Jaubert, J. M. 1996.
Roots’ in mixotrophic algae. Nature 381: 382.
Cundell, A. M., Sleeter, T. D., Mitchell, R. 1977. Microbial populations associated
with the surface of the brown alga Ascophyllum nodosum. Microb Ecol 4 (1):
81–91.
Delbridge, L., Coulburn, J., Fagerberg, W., Tisa, L. S. 2004. Community profiles of
bacterial endosymbionts in four species of Caulerpa. Symbiosis 37: 335–344.
Dittami, S. M., Duboscq-Bidot, L., Perennou, M. et al. 2016. Host–microbe interac-
tions as a driver of acclimation to salinity gradients in brown algal cultures.
ISME J 10: 51–63.
Egan, S., Harder, T., Burke, C., Steinberg, P., Kjelleberg, S., Thomas, T. 2013. The
seaweed holobiont: Understanding seaweed-bacteria interactions. FEMS
Microbiol Rev 37 (3): 462–476.
Eigemann, F., Hilt, S., Salka, I., Grossart, H. P. 2013. Bacterial community composi-
tion associated with freshwater algae: Species specificity versus dependency
on environmental conditions and source community. FEMS Microbiol Ecol
83 (3): 650–663.
Gupta, S., Abu-Ghannam, N. 2011. Bioactive potential and possible health effects
of edible brown seaweeds. Trends Food Sci Technol 22 (6): 315–326.
Hanshew, A. S., Mason, C. J., Raffa, K. F., Currie, C. R. 2013. Minimization of chlo-
roplast contamination in 16S rRNA gene pyrosequencing of insect herbivore
bacterial communities. J Microbiol Methods 95 (2): 149–155.
Hollants, J., Decleyre, H., Leliaert, F., De Clerck, O., Willems, A. 2011. Life without
a cell membrane: Challenging the specificity of bacterial endophytes within
Bryopsis (Bryopsidales, Chlorophyta). BMC Microbiol 11: 255.
Hollants, J., Leliaert, F., De Clerck, O., Willems, A. 2010. How endo- is endo-? Surface
sterilization of delicate samples: A Bryopsis (Bryopsidales, Chlorophyta) case
study. Symbiosis 51: 131–138.
Hollants, J., Leliaert, F., Verbruggen, H., Willems, A., De Clerck, O. 2013. Permanent
residents or temporary lodgers: Characterizing intracellular bacterial com-
munities in the siphonous green alga Bryopsis. Proc Biol Sci 280: 20122659.
Lachnit, T., Blümel, M., Imhoff, J. F., Wahl, M. 2009. Specific epibacterial commu-
nities on macroalgae: Phylogeny matters more than habitat. Aquat Biol 5:
181–186.
Lachnit, T., Meske, D., Wahl, M., Harder, T., Schmitz, R. 2011. Epibacterial commu-
nity patterns on marine macroalgae are host-specific but temporally vari-
able. Environ Microbiol 13: 655–665.
Lane, D. J. 1991. 16S/23S rRNA sequencing. In Nucleic Acid Techniques in Bacterial
Systematic, E. Stackebrandt and M. Goodfellow (Eds.), pp. 115–175. New York:
John Wiley & Sons.
Langille, M. G. I., Zaneveld, J., Caporaso, J. G. et al. 2013. Predictive functional
profiling of microbial communities using 16S rRNA marker gene sequences.
Nature Biotechnol 31 (9): 814–821.
112 Protocols for Macroalgae Research
Lundberg, D. S., Yourstone, S., Mieczkowski, P., Jones, C. D., Dangl, J. L. 2013.
Practical innovations for high-throughput amplicon sequencing. Nat Meth
10 (10): 999–1002.
Mancuso, F. P., D’Hondt, S., Willems, A., Airoldi, L., De Clerck, O. 2016. Diversity
and temporal dynamics of the epiphytic bacterial communities associated
with the canopy-forming seaweed Cystoseira compressa (Esper) Gerloff and
Nizamuddin. Front Microbiol 7: 476.
Marzinelli, E. M., Campbell, A. H., Valdes, E. Z. et al. 2015. Continental-scale vari-
ation in seaweed host-associated bacterial communities is a function of host
condition, not geography. Environ Microbiol 17(10): 4078–4088.
Matsuo, Y., Imagawa, H., Nishizawa, M., Shizuri, Y. 2005. Isolation of an algal
morphogenesis inducer from a marine bacterium. Science 307: 1598.
Meusnier, I., Olsen, J. L., Stam, W. T., Destombe, C., Valero, M. 2001. Phylogenetic
analyses of Caulerpa taxifolia (Chlorophyta) and of its associated bacterial
microflora provide clues to the origin of the Mediterranean introduction.
Mol Ecol 10: 931–946.
Nylund, G. M., Persson, F., Lindegarth, M., Cervin, G., Hermansson, M., Pavia,
H. 2010. The red alga Bonnemaisonia asparagoides regulates epiphytic bacte-
rial abundance and community composition by chemical defence. FEMS
Microbiol Ecol 71 (1): 84–93.
Parks, D. H., Tyson, G. W., Hugenholtz, P., Beiko, R. G. 2014. STAMP: Statistical anal-
ysis of taxonomic and functional profiles. Bioinformatics 30 (21): 3123–3124.
Qian, P., Xu, Y., Fusetani, N. 2010. Natural products as antifouling compounds:
Recent progress and future perspectives. Biofouling 26 (2): 223–234.
Riesenfeld, C. S., Schloss, P. D., Handelsman, J. 2004. Metagenomics: Genomic
analysis of microbial communities. Annu Rev Genet 38: 525–552.
Rosenberg, E., Koren, O., Reshef, L., Efrony, R., Zilber-Rosenberg, I. 2007. The
role of microorganisms in coral health, disease and evolution. Nature Rev
Microbiol 5: 355–362.
Schäfer, H., Muyzer, G. 2001. Denaturing gradient gel electrophoresis in marine
microbial ecology. Method Microbiol 30: 425–468.
Schloss, P. D., Westcott, S. L., Ryabin, T. et al. 2009. Introducing mothur: Open-
source, platform-independent, community-supported software for describ-
ing and comparing microbial communities. Appl Environ Microbiol 75 (23):
7537–7541.
Singh, R. P., Reddy, C. R. K. 2014. Seaweed-microbial interactions: Key functions of
seaweed-associated bacteria. FEMS Microbiol Ecol 88: 213–230.
Singh, R. P., Reddy, C. R. K. 2016. Unraveling the functions of the macroalgal
microbiome. Front Microbiol 6: 1–8.
Smit, A. J. 2004. Medicinal and pharmaceutical uses of seaweed natural products:
A Review. J Appl Phycol 16 (4): 245–262.
Spoerner, M., Wichard, T., Bachhuber, T., Stratmann, J., Oertel, W. 2012. Growth
and thallus morphogenesis of Ulva mutabilis (Chlorophyta) depends on a
combination of two bacterial species excreting regulatory factors. J Phycol
48 (6): 1433–1447.
Chapter five: Unraveling seaweeds bacteriomes 113
Stratil, S. B., Neulinger, S. C., Knecht, H., Friedrichs, A. K., Wahl, M. 2013.
Temperature-driven shifts in the epibiotic bacterial community composi-
tion of the brown macroalga Fucus vesiculosus. Microbiologyopen 2: 338–349.
Tujula, N. A., Crocetti, G. R., Burke, C., Thomas, T., Holmström, C., Kjelleberg, S.
2010. Variability and abundance of the epiphytic bacterial community asso-
ciated with a green marine Ulvacean alga. ISME 4 (2): 301–311.
Vieira, C., Engelen, A. H., Guentas, L. et al. 2016. Species specificity of bacteria
associated to the brown seaweeds Lobophora (Dictyotales, Phaeophyceae)
and their potential for induction of rapid coral bleaching in Acropora
muricata. Front Microbiol 7: 316.
Wang, Y., Qian, P. Y. 2009. Conservative fragments in bacterial 16S rRNA genes
and primer design for 16S ribosomal DNA amplicons in metagenomic stud-
ies. PLoS One 4 (10): e7401.
Weiss, A., Costa, R., Wichard, T. 2017. Morphogenesis of Ulva mutabilis
(Chlorophyta) induced by Maribacter species (Bacteroidetes, Flavobacteriaceae).
Botanica Marina 60 (2): 197–206.
Whatley, J. M., John, P., Whatley F. R. 1979. From extracellular to intracellular: The
establishment of mitochondria and chloroplasts. Proc Biol Sci 204: 165–187.
Wichard, T. 2015. Exploring bacteria-induced growth and morphogenesis in the
green macroalga order Ulvales (Chlorophyta). Front Plant Sci 6: 86.
chapter six
Contents
6.1 Introduction ............................................................................................115
6.2 State of the art .........................................................................................116
6.3 Materials..................................................................................................118
6.3.1 Trace metal cleaning ..................................................................118
6.3.2 Algal material for spore release and toxicity test..................118
6.4 Experimental procedures .....................................................................119
6.4.1 Trace metal clean techniques ...................................................119
6.4.2 Algal material ............................................................................ 120
6.4.3 Toxicity test ................................................................................ 120
6.4.4 End points .................................................................................. 122
6.4.5 Adaptation of the protocol to microalgae and adult
macroalgal specimens .............................................................. 124
6.5 Notes ....................................................................................................... 124
References........................................................................................................ 125
6.1 Introduction
Marine ecotoxicology is a branch of toxicology dealing with the study
of the toxic effects, caused by natural or synthetic pollutants, with fun-
damental focus on the members of the marine ecosystems, for example,
flora and fauna. The objective of ecotoxicological studies is to determine
the impacts of anthropogenic toxic waste discharged to marine environ-
ments (Chapman 1995). The main tool used in ecotoxicological research
is a short-term assay to measure the toxicity of a pollutant (e.g., heavy
metals) on different physiological parameters of a key species (Burridge
and Bidwell 2002). Empirical data can be used to estimate the long-term
115
116 Protocols for Macroalgae Research
6.3 Materials
6.3.1 Trace metal cleaning
• Distilled and deionized water (~18 MΩ cm): Several liters are required
for trace metal cleaning.
• Nonabrasive soap, detergent, or cleaning powder.
• Acid solution: Prepare 10% 2 M HCl in deionized water.
• 5–10 L basin for soaking labwares.
• 500 mL polyethylene plastic bottles.
• Polystyrene tissue culture vessels: Six-well plate (Costar 3516,
Corning Inc.).
• All labwares for handling algal materials, spore release, and ecotox-
icity test: For example, beakers, graduated cylinder, screw-capped
tubes, trays, and containers, among others.
• Laminar flow hood.
• Laboratory gown.
• Hand gloves.
(a) (b)
Figure 6.1 Reproductive tissue aka sorus from (a) sporophylls of Alaria esculenta
and (b) lamina of Saccharina latissima.
(a) (b)
70
y = 0.2946x + 3.209
60
Germination inhibition (%)
R2 = 0.8814
50
40
30
20
10
0
0 50 100 150 200
Dissolved metal concentration (μg L−1)
Figure 6.4 Relationship between dissolved heavy metal concentration and ger-
mination rate expressed as percentage. Linear regression was used to obtain
dose–response relationship. The calculated half maximal effective concentration
EC50 is 159 µg L−1 dissolved metal.
124 Protocols for Macroalgae Research
6.5 Notes
Note 1: Ideally, it is recommended to use different individuals as rep-
licates to measure individual variability to a given stress factor.
However, in the case of extensive experimental variables and levels
Chapter six: Heavy metal ecotoxicity on the early life history stages 125
References
Abalde, J., Cid, A., Reiriz, S., Torres, E., and Herrero, C. 1995. Response of the
marine microalga Dunaliella tertiolecta (Chlorophyceae) to copper toxicity in
short time experiments. Bull. Environ. Contam. Toxicol. 54:317–324.
Abràmoff, M.D., Magalhães, P.J., and Ram, S.J. 2004. Image processing with image.
J. Biophotonics Int. 11:36–42.
Andersen, R.A. and Kawashi, M. 2005. Traditional microalgae isolation tech-
niques. In Andersen, R. A. (Ed.) Algal Culturing Techniques. 1st ed. Elsevier
Academic Press, Burlington, NJ, pp. 83–100.
Anderson, B.S., Hunt, J.W., Turpen, S.L., Coulon, A.R., and Martin, M. 1990.
Copper toxicity to microscopic stages of giant kelp Macrocystis pyrifera:
Interpopulation comparisons and temporal variability. Mar. Ecol. Prog. Ser.
68:147–156.
Baumann, H.A., Morrison, L., and Stengel, D.B. 2009. Metal accumulation and
toxicity measured by PAM-Chlorophyll fluorescence in seven species of
marine macroalgae. Ecotoxicol. Environ. Saf. 72:1063–1075.
126 Protocols for Macroalgae Research
Burridge, T.R. and Bidwell, J.R. 2002. Review of the potential use of brown algal
ecotoxicological assays in monitoring effluent discharge and pollution in
southern Australia. Mar. Pollut. Bull. 45:140–147.
Chapman, P.M. 1995. Ecotoxicology and pollution—Key issues. Mar. Pollut. Bull.
31:167–177.
Contreras, L., Medina, M.H., Andrade, S., Oppliger, L.V., and Correa, J.A. 2007.
Effects of copper on early developmental stages of Lessonia nigrescens Bory
(Phaeophyceae). Environ. Pollut. 145:75–83.
Eklund, B.T. and Kautsky, L. 2003. Review on toxicity testing with marine mac-
roalgae and the need for method standardization—Exemplified with copper
and phenol. Mar. Pollut. Bull. 46:171–181.
Garman, G.D., Pillai, M.C., and Cherr, G.N. 1994. Inhibition of cellular events dur-
ing early algal gametophyte development: Effects of select metals and an
aqueous petroleum waste. Aquat. Toxicol. 28:127–144.
Guillard, R.R.L. and Sieracki, M.S. 2005. Counting cells in cultures with the
light microscope. In Andersen, R. A. (Ed.) Algal Culturing Techniques. 1st ed.
Elsevier Academic Press, Burlington, NJ, pp. 239–252.
Haglund, K., Björklund, M., Gunnare, S., Sandberg, A., Olander, U., and Pedersén,
M. 1996. New method for toxicity assessment in marine and brackish
environments using the macroalga Gracilaria tenuistipitata (Gracilariales,
Rhodophyta). Hydrobiologia. 326/327:317–325.
Leal, P.P., Hurd, C.L., Fernández, P.A., and Roleda, M.Y. 2017. Ocean acidification
and kelp development: reduced pH has no negative effects on meiospore
germination and gametophyte development of Macrocystis pyrifera and
Undaria pinnatifida. J. Phycol. 53:557–566.
Leal, P.P., Hurd, C.L., and Roleda, M.Y. 2014. Meiospores produced in sori of non-
sporophyllous laminae of Macrocystis pyrifera (Laminariales, Phaeophyceae)
may enhance reproductive output. J. Phycol. 50:400–405.
Leal, P.P., Hurd, C.L., Sander, S.G., Armstrong, E.A., and Roleda, M.Y. 2016a.
Copper ecotoxicology of marine algae: A methodological appraisal. Chem.
Ecol. 32:786–800.
Leal, P.P., Hurd, C.L., Sander, S.G., Kortner, B., and Roleda, M.Y. 2016b. Exposure
to chronic and high dissolved copper concentrations impede meiospore
development of the kelps Macrocystis pyrifera and Undaria pinnatifida
(Ochrophyta). Phycologia. 55:12–20.
Levy, J.L., Stauber, J.L., and Jolley, D.F. 2007. Sensitivity of marine microalgae to
copper: The effect of biotic factors on copper adsorption and toxicity. Sci.
Total Environ. 387:141–154.
Magnusson, M., Heimann, K., and Negri, A.P. 2008. Comparative effects of herbi-
cides on photosynthesis and growth of tropical estuarine microalgae. Mar.
Pollut. Bull. 56:1545–1552.
Maienthal, E. and Becker, D. 1976. A survey of current literature on sampling,
sample handling, and long term storage for environmental materials. Natl.
Bur. Stand. (U.S.), Tech. Note 929:40 p.
Richter, R. 2003. Clean Chemistry Techniques for the Modern Laboratory. First.
Milestone Press, Shelton, CT. 96 p.
Roleda, M.Y., Campana, G.L., Wiencke, C., Hanelt, D., Quartino, M.L., and Wulff,
A. 2009. Sensitivity of Antarctic Urospora penicilliformis (Ulotrichales,
Chlorophyta) to ultraviolet radiation is life-stage dependent. J. Phycol.
45:600–609.
Chapter six: Heavy metal ecotoxicity on the early life history stages 127
Roleda, M.Y., Morris, J.N., McGraw, C.M., and Hurd, C.L. 2012. Ocean acidification
and seaweed reproduction: Increased CO2 ameliorates the negative effect of
lowered pH on meiospore germination in the giant kelp Macrocystis pyrifera
(Laminariales, Phaeophyceae). Glob. Chang. Biol. 18:854–864.
Roleda, M.Y., Wiencke, C., Hanelt, D., and Bischof, K. 2007. Sensitivity of the early
life stages of macroalgae from the northern hemisphere to ultraviolet radia-
tion. Photochem. Photobiol. 83:851–862.
Stauber, J.L. 1995. Toxicity testing using marine and freshwater unicellular algae.
Australas. J. Ecotoxicol. 1:15–24.
van Dam, R.A., Harford, A.J., Houston, M.A., Hogan, A.C., and Negri, A.P. 2008.
Tropical marine toxicity testing in Australia: A review and recommenda-
tions. Australas. J. Ecotoxicol. 14:55–88.
Yong, Y.S., Yong, W.T.L., and Anton, A. 2013. Analysis of formulae for determina-
tion of seaweed growth rate. J. Appl. Phycol. 25:1831–1834.
chapter seven
Contents
7.1 Introduction ........................................................................................... 129
7.2 State of the art ........................................................................................ 130
7.3 Materials................................................................................................. 131
7.3.1 Reagents ..................................................................................... 131
7.3.2 Equipment .................................................................................. 131
7.3.3 Reagent setup ............................................................................ 132
7.4 Experimental procedures .................................................................... 132
7.5 Notes ....................................................................................................... 135
Acknowledgments ......................................................................................... 136
References........................................................................................................ 136
7.1 Introduction
Protoplasts, the plant somatic cells devoid of cell wall, offer a unique
experimental material for cellular biology and biotechnology (Davey et al.
2005). The most remarkable achievement of protoplast research has been
the development of various agricultural crop varieties with improved
agronomic traits by somatic hybridization (Bajaj 2004). Protoplasts have
also been extensively employed in genetic transformation studies because
of their amenability for genetic manipulation techniques (Eeckhaut et al.
2013). The recent developments in modern biotechnology techniques have
further broadened the scope of application of protoplast research in fur-
ther understanding the genetic regulations involved in dedifferentiation
(Xiao et al. 2012), organelle distribution and epigenetic changes (Jiang et al.
129
130 Protocols for Macroalgae Research
2013), signal transduction (Costa et al. 2012), transient gene expression
system (Miao and Jiang 2007; Yoo et al. 2007; Zhang et al. 2011; Pitzschke
and Persak 2012; Thévenin et al. 2012), interaction between transcription
factors and genes (Li et al. 2011; Wehner et al. 2011; Huha et al. 2013;
Ge et al. 2013), cell-wall biosynthesis (Zhao et al. 2008; Bhargava et al.
2013), gene deletion, and mutation library creation (Ge et al. 2013). Recent
interest on cell-specific expression profiling has further provided impetus
to the protoplast research (Robert et al. 2007; Tohge et al. 2011). In addition,
plant protoplasts have also been employed as simple, fast, reliable, and
versatile one-hybrid like screening system to investigate the interaction
studies between transcription factors and their associated genes offering
advantages over the yeast two-hybrid system (Thévenin et al. 2012).
The protoplast isolation from terrestrial plants was first initiated by
Von Hannstein in 1880 by using the mechanical method and was subse-
quently refined by Plowe in 1931 (Cocking 1961). Cocking in 1961 showed
the first successful isolation of protoplasts from tomato seedling root tips
using cellulase extracted from fungus Myrothecium verrucariae (Cocking
1961). The successful regeneration of plant protoplasts with the enzymatic
treatment of tissue was first published by Takebe et al. (1971) in tobacco,
almost a decade later from the first report of protoplasts isolation. As com-
pared with higher plants, marine macroalgal protoplast research is in a
nascent stage and dates back to 1970.
and top shell) guts (Reddy et al. 2008). In addition to these complexities,
most of the enzyme formulations previously employed for isolation of
protoplasts from Ulvophycean species were made in seawater (Saga 1984;
Saga and Kudo 1989; Uppalapati and Fujita 2002). The high-ionic potential
of the medium was reported to interfere with studies on electrofusion,
and other investigation on ion channels. Consequently, enzyme formula-
tions made in distilled water supplemented with a higher concentration of
osmolyte, for example, mannitol (1 M), have been preferred (Zhang 1983;
Fujita and Migita 1985). However, the viability (30%) and regeneration
abilities (50% of viable protoplasts) of such isolated protoplasts have been
found to be markedly low as compared with those obtained with seawater-
based enzyme mixture. The high-osmotic medium has been reported to
affect the regeneration rate and subsequent morphogenesis of cultured
protoplasts (Saga et al. 1986). The use of higher concentrations of osmo-
lytes (sugar alcohols) is prone to cause microbial contamination imposing a
gradual dilution of it in protoplast culture medium to improve and induce
morphogenesis. Therefore, the development of a facile, reliable, and uni-
fied protocol for routine protoplast preparation with reproducible yield is
of paramount importance to circumvent the inherent problems associated
with the use of previously reported enzyme mixtures regarding variabil-
ity in yield, regeneration rate, and developmental morphogenesis.
7.3 Materials
7.3.1 Reagents
• Sodium chloride (NaCl).
• 2-(N-Morpholino) ethanesulfonic acid.
• Dextran sulfate sodium salt.
• Mannitol.
• Cellulase Onozuka R-10 (Yakult Co. Ltd., Tokyo, Japan).
• Macerozyme R-10 (Yakult Co. Ltd., Tokyo, Japan).
• Deionized water (Millipore water).
7.3.2 Equipment
• Weighing balance (analytical type).
• Weighing boats.
• Magnetic stirrer with stirring rod and bar.
• pH meter.
• Refrigerated centrifuge.
• Centrifuge tubes (polypropylene).
• Tube racks.
• Cryovials.
132 Protocols for Macroalgae Research
The schematic description of the protocol is shown in Figure 7.1. The iso-
lated protoplasts following this protocol from different species of genus
Ulva were spherical in shape, ranging from 20 to 32 µm in size and con-
tained a cup-shaped parietal chloroplast. The resultant protoplast yields,
and regeneration rate are summarized in Table 7.1. Regeneration rate of
protoplasts was calculated on the basis of the number of cells dividing
and regenerating into germlings. Most of the protoplasts under opti-
mal culture conditions (temperature 25°C and photon flux intensity of
15 µM m−2 s−1) regenerated cell wall after 72 h and subsequently initiated
cell division. The dedifferentiation of protoplast to develop germlings is
shown in Figure 7.2. Cell-wall development was confirmed as the emission
of blue fluorescence after staining with calcofluor white MR2 (Fluorescent
brightener 28, Sigma Aldrich, USA) (Figure 7.2).
Algal Thalli
is
Debr l
Enzyme Seawater va
remo
ion mix removal
b at
cu
In
Filtra
ion t
Centrifuge Protoplasts
Figure 7.1 Schematic diagram of the protocol developed for rapid isolation of
protoplasts from Ulvophycean species.
134 Protocols for Macroalgae Research
Table 7.1 Protoplasts yields and regeneration rates obtained from different
Ulvophycean species following the described protocol
Protoplasts yield
Species (×106 cells/g fresh wt.) Regeneration rate (%)
Ulva conglobata 85.2 ± 2.9 92.3 ± 6.8
U. fasciata 81.4 ± 2.1 90.7 ± 7.3
U. lactuca (wild) 74.5 ± 1.6 89.4 ± 5.6
U. lactuca (sterile mutant) 76.6 ± 1.2 91.2 ± 4.5
U. pertusa (sterile mutant) 74.4 ± 1.9 92.3 ± 6.2
U. reticulata 88.6 ± 2.3 90.3 ± 6.7
U. rigida 79.6 ± 1.1 89.5 ± 2.4
Enteromorpha tubulosa 89.7 ± 1.9 91.4 ± 4.7
E. flexuosa 95.4 ± 2.2 91.4 ± 4.8
E. compressa 82.5 ± 1.5 90.5 ± 5.9
Monostroma oxyspermum 102.8 ± 1.8 90.5 ± 6.5
40 μm 40 μm 50 μm
(a) (b) (c)
50 μm 100 μm 500 μm
(d) (e) (f )
1 mm 60 μm
7.5 Notes
Note 1: While preparing and handling the enzyme mix, it is advised
to maintain the temperature at 4°C. Exposing the enzyme mix at
room temperature for long durations is detrimental to its activity
and results in poor or no yields of protoplasts.
Note 2: Store enzyme mix at −20°C.
Note 3: The exponentially growing algal tissue is required to obtain
good yields of protoplasts (Table 7.2).
Note 4: Protoplasts are highly fragile because of the lack of rigid cell
wall; therefore, they advised to handle very carefully. Shake or acci-
dental jerks may lead to the rupture of released protoplast.
Note 5: The supernatant after step 12 can be collected and stored at
−20°C for further reuse as enzyme mix for protoplast preparation.
This can be continued for five times.
Acknowledgments
The financial support received from the FP7 program of European
Commission under grant agreement no. 241383 is gratefully acknowl-
edged. VG thanks, CSIR-NIO, Goa for infrastructural support in writing
this protocol.
References
Bajaj, Y.P.S. 2004. Biotechnology in agriculture and forestry. In Somatic Hybridization
in Crop Improvement I, vol. 27. Springer-Verlag, Berlin, Germany.
Bhargava, A., Ahad, A., Wang, S., Mansfield, S.D., Haughn, G.W., Douglas, C.J.,
and Ellis, B.E. 2013. The interacting MYB75 and KNAT7 transcription fac-
tors modulate secondary cell wall deposition both in stems and seed coat in
Arabidopsis. Planta 237: 1199–1211.
Cocking, E.C. 1961. Properties of isolated plant protoplasts. Nature 191: 780–782.
Costa, A. et al. 2012. The Arabidopsis central vacuole as an expression system
for intracellular transporters: Functional characterization of the Cl−/H+
exchanger CLC-7. J. Physiol. 590: 3421–3430.
Davey, M.R., Anthony, P., Power, J.B., and Lowe, K.C. 2005. Plant protoplasts: Status
and biotechnological perspectives. Biotechnol. Adv. 23: 131–171.
Eeckhaut, T., Lakshmanan, P.S., Deryckere, D., van Bockstaele, E., and van
Huylenbroeck, J. 2013. Progress in plant protoplast research. Planta 238:
991–1003.
Enomoto, K. and Hirose, H. 1972. Culture studies on artificially induced apla-
nospores in the marine alga Boergesenia forbesii (Harvey) Feldman
(Chlorophyceae, Siphonocladales). Phycologia 11: 119–122.
Fujita, Y. and Migita, S. 1985. Isolation and culture of protoplasts from seaweeds.
Bull. Fac. Fish. Nagasaki Univ. 57: 39–45.
Ge, C.Y., Duan, Y.B., Zhou, M.G., and Chen, C.J. 2013. A protoplast transformation
system for gene deletion and complementation in Sclerotinia sclerotiorum.
J. Phytopathol. 161: 800–806.
Huha, S.U., Kimb, M.J., and Paeka, K.H. 2013. Arabidopsis Pumilio protein APUM5
suppresses Cucumber mosaic virus infection via direct binding of viral
RNAs. Proc. Natl. Acad. Sci. USA. 110: 779–784.
Jiang, F., Zhu, J., and Liu, H.L. 2013. Protoplasts: A useful research system for plant
cell biology, especially dedifferentiation. Protoplasma 250: 1231–1238.
Kobayashi, K. 1975. Growth of extra cellular protoplast of Bryopsis maxima in an
agar medium. Bull. Tokyo Gakugei Univ. Ser. 27: 1–5.
Krishna Kumar, G.R., Addepalli, M.K., and Reddy, C.R.K. 1999. Regeneration of
the thallus of Monostroma oxyspermum (Chlorophyta) from protoplasts in
axenic culture. Phycologia 38: 503–507.
Li, E., Wang, S., Liu, Y., Chen, J.G., and Douglas, C.J. 2011. Ovate family protein4
(OFP4) interaction with KNAT7 regulates secondary cell wall formation in
Arabidopsis thaliana. Plant J. 67: 328–341.
Miao, Y. and Jiang, L. 2007. Transient expression of fluorescent fusion proteins in
protoplasts of suspension cultured cells. Nat. Protoc. 2: 2348–2353.
Millner, P.A., Callow, M.E., and Evans, L.V. 1979. Preparation of protoplasts from
the green alga Enteromorpha intestinalis (L.). Planta 147: 174–177.
Chapter seven: A simple protocol for protoplasts production 137
Zhang, Y. et al. 2011. A highly efficient rice green tissue protoplast system for
transient gene expression and studying light/chloroplast-related processes.
Plant Methods 7: 30.
Zhao, M., Morohashi, K., Hatlestad, G., Grotewold, E., and Lloyd, A. 2008. The
TTG1-bHLH-MYB complex controls trichome cell fate and patterning
through direct targeting of regulatory loci. Development 135: 1991–1999.
chapter eight
Contents
8.1 Introduction ........................................................................................... 140
8.2 State of the art .........................................................................................141
8.3 Materials................................................................................................. 142
8.3.1 Ulva culture................................................................................ 142
8.3.2 Culturing of U. mutabilis on a large scale .............................. 142
8.3.3 Harvesting of Ulva biomass for the extraction and the
bioassay-guided fractionation of inhibitors .......................... 143
8.3.4 Induction of gametogenesis/sporogenesis and
bioassay-guided fractionation of the inhibitors ................... 143
8.3.5 Sporulation inhibitor 1: Extraction ......................................... 144
8.3.5.1 Sporulation inhibitor 1: Size-exclusion
chromatography for cleanup .................................... 144
8.3.6 Sporulation inhibitor 2: Extraction and bioassay-guided
fractionation............................................................................... 144
8.3.7 Swarming Inhibitor: Extraction and bioassay-guided
fractionation............................................................................... 145
8.4 Experimental procedures .................................................................... 145
8.4.1 Cultivation of U. mutabilis under standardized
conditions ................................................................................145
8.4.2 Induction of the gametogenesis/zoo sporogenesis and
preparation of the bioassays .................................................... 147
8.4.3 Sporulation inhibitor 1 ............................................................. 149
8.4.3.1 Extraction of the sporulation inhibitor 1 from
thallus .......................................................................... 149
8.4.3.2 Extraction of the sporulation inhibitor 1 from
(axenic) growth medium ........................................... 150
139
140 Protocols for Macroalgae Research
8.1 Introduction
Ulva species (Chlorophyta), also known as sea lettuce, are the most abun-
dant representatives of green macroalgae and can be found in coastal ben-
thic areas worldwide. They cause seasonal algal blooms in eutrophic waters
and even in areas contaminated with heavy metals (Gao et al. 2010). The
model organism, Ulva mutabilis, is a multicellular haplodiplontic alga, which
differentiates into a thallus and a holdfast. The thallus is made of blade
cells and the holdfast of stem and rhizoid cells (Løvlie 1964). The blade cells
develope into gametangia (sporangia) and release 16 biflagellated gametes
per gametangium (16 tetraflagellated zoospores) (Løvlie 1964, 1968; Wichard
and Oertel 2010). After merging of the two mating types of the gametes mt+
and mt–, zygotes are formed which develop into the sporophytes. However,
if the gametes do not find a mating partner, they develop into new gameto-
phytes, which allow the maintenance of haploid strains (Wichard 2015, see
Ulva′s life cycle in Chapter 9 by Califano and Wichard 2018).
The sporulation processes, gametogenesis and zoosporogenesis, are
key parts of Ulva reproduction and still an understudied field. Both the
gametophytes and sporophytes of the haplodiplontic Ulva have the same
morphological characteristics but form either biflagellated gametes or
quadriflagellated zoids. Sporulation can artificially be induced by cutting
the mature algae into smaller pieces and washing them. This treatment
removes sporulation inhibitors (SI-1 and SI-2), which regulate the differ-
entiation of blade cells into gametangia and sporangia in Ulva. (Nilsen
and Nordby 1975; Løvlie 1978; Stratmannet al. 1996). In addition, further
studies revealed that switching temperatures, as experienced by certain
tropical Ulva species, can induce the cell differentiation (Carl et al. 2014).
Moreover, Lüning et al. (2008) found that the reproduction rhythmicity of
U. pseudocurvata is also controlled by the moon phases (Lüning et al. 2008).
Chapter eight: Purification of sporulation and swarming inhibitors 141
Figure 8.1 All-in-one workflow for the partial purification of known inhibitors (sporu-
lation inhibitor 1 [SI-1], sporulation inhibitor 2 [SI-2], swarming inhibitor [SWI])
regulating the gametogenesis, and release of gametes in Ulva. On removal of SI-1
and SI-2, the SWI production can be induced by putting the fragmented algae into
fresh UCM. The best SI-1 extraction results can be achieved with axenic or with
young Ulva thalli.
8.3 Materials
8.3.1 Ulva culture
The material necessary for the standard cultivation of Ulva was described
previously (Stratmann et al. 1996; Alsufyani et al. 2017). Gametophytes of
the same species, U. mutabilis (Føyn), were also used in the protocols of the
Chapters 9, 18, and 31.
(g) (f ) (e)
(b)
(c)
(d)
(a)
Figure 8.2 Drawing of the 25 L bioreactor used for cultivation of Ulva mutabilis:
(a) air inlet; (b) sterile HEPA-Vent (Ø = 50 mm, Whatman) filter; (c) air outlet;
(d) sampling outlet; (e) bubbling tube (Duran glass, Ø = 4 mm); (f) sampling tube
(Teflon, Ø = 1 mm); and (g) hose clamp to control the sampling flow. (Adapted
from Alsufyani, T. et al. 2017, With permission, CC-BY.)
Chapter eight: Purification of sporulation and swarming inhibitors 147
1. Wash five U. mutabilis fertile thalli (3–5 cm length) with UCM and
incubate them in 0.2 L UCM for 15 min at room temperature.
2. Mince the alga into 3–5 mm² by using the herb chopper and incubate
the fragments in 50% UCM for 15 min at room temperature; repeat
the washing step twice (for further details see Chapter 9). Recover
the algal fragments through a mesh of 100 µm.
3. Prepare the dilution series of the potentially active test sample
with UCM in the 96-well plate. The total volume per well is 200 µL
(Figure 8.3a). The ratio of a standard dilution series might be, for
example, 1:5, 1:10, 1:50, and 1:100, but the dilution factor must be
adjusted if the SWI concentration is higher.
4. Do not forget a negative control: Only UCM (i.e., no inhibitor) is added
and, thus, all blade cells should differentiate into gametangia. In
the case of the positive control, a defined amount of SI (100 U mL−1,
see definition in step 9) of the laboratory quality control sample is
added. The blade cells should, thus, not differentiate into gametan-
gia or sporangia, respectively (Figure 8.3a). The quality control sam-
ples should be treated in the same manner as the test samples and
are used to validate the test run.
5. Transfer the sieved algal pieces with forceps into the multiwell plates
(40–80 pieces per well). When transferring the fragments (from step 2)
into the test well, remove the leftover liquid with a paper towel to
avoid the dilution of the test solution.
6. The algal fragments should be cultured under standard conditions
for the next three days.
7. On the morning of the third day after induction, the fragments must
be inspected in each test well under an inverted microscope. Those
fragments that show on blade cells are classified as noninduced; all
others with gametangia are classified as induced (Figure 8.3b). If the
positive control does not show any inhibition, the experiment needs to be
repeated with a new batch of younger algae, which can still perceive the
sporulation inhibitor.
148 Protocols for Macroalgae Research
1 2 3 4 5
(a)
(i) (ii)
Sample
without SI
100 μm 10 μm
(iii) (iv)
Sample
with SI
100 μm 100 μm
(b)
(i) (ii)
Sample
without SWI
Gametes
10 μm 10 μm
(iii) (iv)
Sample
with SWI
10 μm 10 μm
(c)
Figure 8.3 Set up for both sporulation and swarming inhibitor bioassays. (a) Multiwell-
based bioassay-guided fractionation and semiquantification through dilution series of
the biologically active samples. (a) The general principle of the bioassay activity test
for the sporulation inhibitors (SI-1 or SI-2) or the swarming inhibitor (SWI) is shown:
The negative control contains no inhibitor, all blade cells develop into gametangia. The
positive control contains a defined amount of SI or SWI. (b) Light microscopy reveals
the blade cells before the experiment (i, iii). Without SI-1 or SI-2 in the test sample,
(ii) cells develop into gametangia within three days. (c) Gametangia are shown (i, iii).
Without SWI in the test sample, (ii) gametangia are completely discharged within
15 min. The effect of inhibitory samples for both bioassays (b, c) is shown in (iv).
Chapter eight: Purification of sporulation and swarming inhibitors 149
9. 1 U of the SI-1 and SI-2 is hereby defined as the minimal amount of
the factor that inhibits differentiation of a blade cell into a gametan-
gium (=gametogenesis) with an inhibitory rate >50% in 1 mL UCM
within three days under standard conditions. If the test sample, for
example, diluted in a ratio of 1:5 reveals an inhibitory rate of 85%, but
only 40% in a ratio of 1:10, the sample contains 5 U mL−1 of the inhibi-
tor. Fragments (size: 1–5 mm²) sometimes show a mixture of the blade
and differentiated cells. Those fragments counted as induced, which
harbor more than 50% of differentiated cells (i.e., gametangia).
4000
6 6
Biological activity of SI-1 (Units mL )
5 5
3000
at 233 nm
4 4
3 3 2000
2 2
1000
1 1
(c)
0 0
2500
Biological
1500
1000
500
100
(d)
14
80
Biological activity of SI-2
60 10
8
40
6
4
20
2
0 0
0 5 10 15 20 25 14 16 18 20 22 24 26 28 30
(b) Elution volume (mL) (e) Retention time (min)
1. Harvest Ulva, dry it carefully with paper towels, and weigh the
biomass.
2. Wash the alga with UCM twice and incubate the biomass for 15 min
in 0.2 L UCM at room temperature to remove any leftover traces of
the SI-1.
3. An amount of 1 g of dried Ulva is suspended in 1 mL of 10 mM Tris–
HCl (pH 8) and minced into pieces with the herb chopper.
4. Add 3 mL of 10 mM Tris–HCl (pH 8) to the suspension and incubate
the alga for 15 min at room temperature.
5. Filter the algae with a stainless steel filter in a funnel.
6. Transfer the suspension to a 5 mL plastic cup (Eppendorf, Germany)
for centrifugation at 3800 rpm for 10 min at 4°C.
7. The supernatant is passed initially through a GF/C filter and then
through a cellulose-acetate membrane (0.45 µm).
8. All samples should be stored at −20°C.
Chapter eight: Purification of sporulation and swarming inhibitors 153
1. Wash the algal fragments, already used for the SI-2 extraction, three
times with half-strength UCM.
2. Transfer the fragments into UCM and incubate them for 24 h under
standard conditions (Note 7).
3. Remove the algal fragments, for example, with a stainless steel filter
in a funnel.
4. Pass the growth medium through a GF/C filter and subsequently
through a cellulose-acetate membrane (0.45 µm).
5. Load the filtrate onto a SepPaktC18 Plus Long Cartridge at 2 mL min−1.
Up to 2 L can be loaded onto one cartridge.
154 Protocols for Macroalgae Research
8.5 Notes
In several Ulva species, gametogenesis and subsequent gamete release
can be artificially induced by the removal of sporulation and swarming
inhibitors, but thus far, neither these inhibitors nor the signaling pathways
inducing gametogenesis have been characterized. Ongoing attempts to
elucidate the structure of the SIs will result in a deeper understanding of
the molecular mechanism in a midterm run. However, the compounds
already partially purified can be used as a tool to manipulate Ulva’s life
cycle to extend the vegetative growth of algal aquaculture.
Acknowledgments
This work was funded by the Deutsche Forschungsgemeinschaft (DFG)
through the Collaborative Research Centre 1127 “Chemical Mediators
in complex Biosystems” (T.W., R.K.) and the Deutsche Bundesstiftung
Umwelt for the PhD fellowship (R.K.).
References
Alsufyani, T., A. Weiss, and T. Wichard. 2017. Time course exo-metabolomic pro-
filing in the green marine macroalga Ulva (Chlorophyta) for identification
of growth phase-dependent biomarkers. Mar. Drugs 15 (1):14. doi: 10.3390/
md15010014.
Califano G. and T. Wichard. 2018. Preparation of axenic cultures in Ulva
(Chlorophyta). In Biotechnology Protocols for Macroalgae Research, B. Charrier,
T. Wichard, and C.R.K. Reddy (Eds.), Chapter 9. Boca Raton, FL: CRC Press,
Taylor & Francis Group.
Carl, C., R. de Nys, R. J. Lawton, and N. A. Paul. 2014. Methods for the induction
reproduction in a species of filamentous Ulva. PLoS One 9 (5). doi: 10.1371/
journal.pone.0097396.
Gao, S., X. Y. Chen, Q. Q. Yi, G. C. Wang, G. H. Pan, A. P. Lin, and G. Peng. 2010.
A Strategy for the proliferation of Ulva prolifera, main causative species of
green tides, with formation of sporangia by fragmentation. PLoS One 5 (1):
e8571. doi: 10.1371/journal.pone.0008571.
Jonsson, S., M. H. Laur, and L. Pham-Quang. 1985. Mise en évidence de differentes
types de glycoprotéines dans un extrait inhibiteur de la gamétogenése chez
Enteromorpha prolifera Chlorophycée marine. Cryptogamie/Algologie 6:253–264.
Løvlie, A. 1964. Genetic control of division rate and morphogenesis in Ulva muta-
bilis Føyn. CR Trav. Lab. Carlsb. Comptes. 34:77–168.
Løvlie, A. 1968. On the use of a multicellular alga (Ulva mutabilis Føyn) in the
study of general aspects of growth and differentiation. Nytt. Magasin.
Zoologi. 16:39–49.
Løvlie, A. 1978. Genetic control of cell cycles during morphogenesis in Ulva muta-
bilis. Develop. Biol. 64 (1):164–177.
Katsaros, C., A. Weiss, I. Llangos, I. Theodorou, and T. Wichard. 2017. Cell struc-
ture and microtubule organisation during gametogenesis of Ulva mutabilis
Føyn (Chlorophyta). Bot. Mar. 60 (2):123–135. doi: 10.1515/bot-2016-0073.
Kessler, R. W., A. C. Crecelius, U. S. Schubert, and T. Wichard. 2017. In situ moni-
toring of molecular changes during cell differentiation processes in marine
macroalgae through mass spectrometric imaging. Anal. Bioanal. Chem. 409 (20):
4893–4903. doi: 10.1007/s00216-017-0430-7.
Lüning, K., P. Kadel, and S. J. Pang. 2008. Control of reproduction rhythmicity by
environmental and endogenous signals in Ulva pseudocurvata (Chlorophyta).
J. Phycol. 44 (4):866–873. doi: 10.1111/j.1529-8817.2008.00535.x.
Nilsen, G. and O. Nordby. 1975. Sporulation inhibiting substance from vegetative
thalli of green alga Ulva mutabilis Føyn. Planta 125 (2):127–139.
Chapter eight: Purification of sporulation and swarming inhibitors 157
Contents
9.1 Introduction ........................................................................................... 159
9.2 State of the art .........................................................................................161
9.3 Materials................................................................................................. 163
9.3.1 Equipment .................................................................................. 163
9.3.2 Consumables and reagents...................................................... 163
9.3.2.1 Consumables............................................................... 163
9.3.2.2 PCR Reagents .............................................................. 164
9.3.3 Ulva culture medium ............................................................... 164
9.4 Experimental procedures .................................................................... 165
9.4.1 Induction of gametogenesis and release of gametes ........... 165
9.4.2 Enrichment of gamete density ................................................ 165
9.4.3 Purification of gametes from bacteria .................................... 166
9.4.4 Measuring gamete density: Flow cytometry .........................167
9.4.5 Axenicity proof of gametes ......................................................167
9.4.5.1 Test of axenicity on agar medium............................ 168
9.4.5.2 Test of axenicity by PCR ............................................ 168
9.5 Notes ....................................................................................................... 169
Acknowledgments ......................................................................................... 170
References........................................................................................................ 171
9.1 Introduction
The green marine alga Ulva, also commonly known as the sea lettuce, is a
cosmopolitan seaweed. It alternates between the isomorphic life stages of
gametophyte (haploid) and sporophyte (diploid). Gametophytes produce
biflagellate phototactic germ cells (gametes) through mitosis. Meanwhile,
diploid sporophytes generate photophobic tetraflagellate germ cells (zoo-
spores or zoids) (Figure 9.1) (Løvlie 1964). Hereby, sporulation inhibitors
control the gametogenesis/zoosporogenesis and production of germ cells
(Chapter 8 by Kessler et al. 2018).
159
160 Protocols for Macroalgae Research
Gametophyte
mt (+) mt (−)
Gametes
Zygote
Sporophyte
mt (+/−)
Zoids
Figure 9.1 The isomorphic life cycle of Ulva. All Ulva species are isomorphic and
alternate between gametophytic and sporophytic life stages with similar mor-
phologies. The gametophytes are haploid (n) and the sporophytes are diploid
(2n). The mating types (mt) are indicated by (+) and (−). Dashed arrows show the
parthenogenetic development of gametophytes derived from unfused gametes.
Blade cells differentiate into gametangium or sporangium (dark-gray margins).
(From Wichard, T., Front. Plant Sci., 6, 86, 2015. With permission CC-BY.)
Washing
Removal Collection of
of SWI gametes
Pipette
loading (× 3)
FL3 - Chlorophyll
Side scatter - H
gametes
Contaminated
sample
Figure 9.2 The workflow of obtaining axenic gametes from Ulva includes six
operational steps. The working time is given in parenthesis. Gametogenesis is
induced by removal of sporulation inhibitors (SI). On the morning of the third
day after induction and removal of a swarming inhibitor (SWI), phototactic gam-
etes can be collected and purified from the accompanying bacteria. Axenicity test:
Contaminated samples can be identified by plating gametes on agar (cultivable
bacteria) or by PCR (non-cultivable bacteria).
Chapter nine: Preparation of axenic cultures in Ulva (Chlorophyta) 163
9.3 Materials
9.3.1 Equipment
• Sterile laminar flow cabinet, providing a minimum working area of
0.55 m².
• Benchtop centrifuge with rotor adaptation for plastic microtubes
(Eppendorf-Centrifuge 5415D, maximum speed of 16,100 g).
• Thermocycler device for polymerase chain reaction (PCR).
• Flow cytometer (BD Accuri C6, USA) equipped with 488 nm argon
laser and emission long pass filters for the wavelength of 670 nm.
• Portable light source (e.g., fluorescent tube).
• Automatic pipettes.
1. Gametes gather at the spot light, forming a visible green line on the
flask wall directly exposed to the light (Note 3).
2. Collect the gametes with a pipette into a 15 mL plastic tube (the plas-
tic tube is covered with an aluminum foil except for the bottom part)
and again expose the gametes to light.
3. After approximately 10 min, remove the clear supernatant (without
gametes) carefully and transfer the green fraction into a 1.5 mL tube
(Figure 9.2).
Light
source
Pipette
support
(a)
(b)
Figure 9.3 Photographs of the two pipette supports: (a) Supports are used during
the purification process. The Pasteur glass pipette is placed on the supports inside
the laminar flow cabinet, and the light source is placed outside the enclosure. (b) A
plastic tube rack can be also used as safe support for the glass pipettes.
Chapter nine: Preparation of axenic cultures in Ulva (Chlorophyta) 167
(a) (b)
Figure 9.4 Ten−day−old cultures of Ulva mutabilis are shown: (a) axenic culture, (b)
typically contaminated culture (after purification) with bacteria possessing the same
activity as the Roseovarius sp. MS2. Although the typical cell wall protrusion of axenic
cultures are visible in (b) as well, the longitudinal cell divisions indicate the presence
of morphogenetic compounds released by bacteria. Magnification bar = 100 µm.
9.5 Notes
Note 1: An exact amount of reference beads is necessary for an accurate
quantification, depending on the flow cytometer available. Choose
the beads on the basis of the specific excitation and emission wave-
length of the laser and filters installed in the equipment.
Note 2: The SWI accumulates in the growth medium during gameto-
genesis and prevents the discharge of the gametes on the morning
of the third day after induction. Induced fragments will change their
color from the dark green of the blade cell to the brownish of the
gametangium.
Note 3: Most gametes are discharged from the gametangia on medium
exchange within the first minutes. However, if the cell division of
the cultures is not completely synchronic, the formation and release
of gametes will be affected, and the gametes will be continuously
released from the gametangium over a period of 2–4 h.
Note 4: There is a significant loss of gametes during the purification
steps (up to 90%). Not all gametes will reach the tip of the pipette. If
the density of gametes is too high, self-shading of the gametes will
prevent them from going through the pipette.
Note 5: Avoid the formation of gas bubbles, which will hinder the move-
ment of the gametes’ during the filling of UCM and loading of gam-
etes in the Pasteur pipettes.
Note 6: In the case of gamete suspension with low density, apply up to
800 µL of the suspension into the wide opening of the pipette.
Note 7: If the movement of the gametes takes longer than 30 min for 15 cm
because of slow-swimming, the separation from the accompanying
bacteria will be insufficient.
170 Protocols for Macroalgae Research
Acknowledgments
This work was funded by the Deutsche Forschungsgemeinschaft (DFG)
through the Collaborative Research Centre 1127 Chemical Mediators in
complex Biosystems and by the European Union’s Horizon 2020 research
and innovation program under the Marie Sklodowska−Curie grant
agreement No.642575—The ALgal Microbiome: Friends and Foes (ALFF)
(G.C. and T.W.).
Chapter nine: Preparation of axenic cultures in Ulva (Chlorophyta) 171
References
Alsufyani, T., A. Weiss, and T. Wichard. 2017. Time course exo−metabolomic pro-
filing in the green marine macroalga Ulva (Chlorophyta) for identification
of growth phase−dependent biomarkers. Mar. Drugs 15: 14. doi: 10.3390/
md15010014.
Andersen, A.R. 2005. Algal Culturing Techniques: A Book for All Phycologists. London,
UK: Elsevier Academic Press.
Kessler, R.W., T. Alsufyani, and T. Wichard. 2018. Purification of sporulation and
swarming inhibitors from Ulva: Application in algal life cycle controlling. In
Protocols for Macroalgae Research, Charrier, B., Wichard, T., and Reddy, C.R.K.
(Eds.), Chapter 8. Boca Raton, FL: CRC Press.
Løvlie, A. 1964. Genetic control of division rate and morphogenesis in Ulva muta-
bilis Føyn. C. R. Trav. Lab. Carlsberg 34: 77–168.
Spoerner, M., T. Wichard, T. Bachhuber, J. Stratmann, and W. Oertel. 2012. Growth
and thallus morphogenesis of Ulva mutabilis (Chlorophyta) depends on a
combination of two bacterial species excreting regulatory factors. J. Phycol.
48: 1433–1447. doi: 10.1111/j.1529−8817.2012.01231.x.
Stratmann, J., G. Paputsoglu, and W. Oertel. 1996. Differentiation of Ulva mutabilis
(Chlorophyta) gametangia and gamete release are controlled by extracellu-
lar inhibitors. J. Phycol. 32(6): 1009–1021. doi: 10.1111/j.0022−3646.1996.01009.x.
Tatewaky, M. and L. Provasoli. 1964. Vitamin requirements of three species of
antithamnion. Bot. Mar. 6: 193–203. doi: 10.1515/botm.1964.6.3−4.193.
Vesty, E.F., R. Kessler, T. Wichard, and J. Coates. 2015. Regulation of gametogen-
esis and zoosporogenesis in Ulva linza (Chlorophyta): Comparison with
Ulva mutabilis and potential for laboratory culture. Front. Plant Sci. 6: 15. doi:
10.3389/fpls.2015.00015.
Wichard, T. 2015. Exploring bacteria−induced growth and morphogenesis in the
green macroalga order Ulvales (Chlorophyta). Front. Plant Sci. 6: 86. doi:
10.3389/fpls.2015.00086.
Wichard, T. 2016. Identification of metallophores and organic ligands in the che-
mosphere of the marine macroalga Ulva (Chlorophyta) and at land-sea inter-
faces. Front. Mar. Sci. 3: 131. doi: 10.3389/fmars.2016.00131.
Wichard, T. and W. Oertel. 2010. Gametogenesis and gamete release of Ulva
mutabilis and Ulva lactuca (Chlorophyta): Regulatory effects and chemical
characterization of the “Swarming Inhibitor”. J. Phycol. 46(2): 248–259. doi:
10.1111/j.1529−8817.2010.00816.x.
Weiss, A., R. Costa, and T. Wichard. 2017. Morphogenesis of Ulva muta-
bilis (Chlorophyta) induced by Maribacter species (Bacteroidetes,
Flavobacteriaceae). Bot. Mar. 60(2): 197–206. doi: 10.1515/bot-2016-0083.
Woodman, M.E. 2008. Direct PCR of intact bacteria (colony PCR). Curr. Prot.
Microbiol. 9: A.3D.1–A.3D.6.
section two
Chemical composition
chapter ten
Contents
10.1 Introduction ......................................................................................... 175
10.2 State of the art ...................................................................................... 177
10.3 Materials ............................................................................................... 177
10.3.1 Extraction and pre-drying of seaweed biomass ................ 177
10.3.2 Sample preparation by pyrolysis ......................................... 177
10.3.3 Biochar generation by pyrolysis........................................... 180
10.3.4 Storage of biochar samples ................................................... 180
10.4 Experimental procedures .................................................................. 180
10.4.1 Extraction and pre-drying of seaweed biomass
(the case of Macrocystis pyrifera) ........................................... 180
10.4.2 Sample preparation for pyrolysis ........................................ 180
10.4.3 Biochar generation by pyrolysis........................................... 181
10.4.4 Storage of biochar samples ................................................... 181
10.5 Notes ..................................................................................................... 182
Acknowledgments ......................................................................................... 183
References........................................................................................................ 183
10.1 Introduction
Biochar is a carbon-rich solid derived from thermally stabilized organic
material, meaning that this product can be stored and applied to soils for
improving its quality. Biochar differs from other solid products derived
from thermochemical conversion, such as charcoal or active carbon, in
that long-term carbon storage is an objective, than the generation of raw
material for processing industries or fuels (Mašek et al. 2013). In technical
terms, biochar is produced through the thermal decomposition of organic
* Loretto Contreras-Porcia and Matías Araya: Both the authors have contributed equally.
175
176 Protocols for Macroalgae Research
10.3 Materials
10.3.1 Extraction and pre-drying of seaweed biomass
• Plastic bags.
• Raschel mesh.
• Tap water.
• Distilled water.
• Hemp or cotton rope, 10–20 mm in diameter.
• Ice pack.
• Knife.
• Bag sealer.
Table 10.1 Main physicochemical characteristics of biochar generated from different biomass sources
and different pyrolysis conditions
CECc SAd
Feedstock PTa (°C) C (%) H (%) O (%) N (%) S (%) pH b (cmol kg–1) Ash (%) (m 2 g–1) References
Algae
Cladophora vagabunda 450 21.80 1.20 – 2.00 – 9.87 23 54.20 – Bird et al. (2011)
Caulerpa taxifolia 450 24.80 1.20 – 2.40 – 9.65 17 20.90 – –
Cladophoropsis sp. 450 23.60 1.50 – 2.80 – 10.07 27 46.50 – –
Chaetomorpha linum 450 23.60 1.30 – 2.40 – 9.61 36 16.00 – –
Chaetomorpha indica 450 10.20 0.80 – 1.10 – 7.83 16 73.50 – –
Cladophora patentiramea 450 20.30 1.20 – 1.70 – 9.12 21 47.00 – –
Cladophora coelothrix 450 34.60 1.50 – 3.3 – 8.72 19 32.10 – –
Ulva flexuosa 450 22.60 1.20 – 2.70 – 10.00 41 42.6 – –
414 25.70 1.90 – 3.90 – 9.80 39 29.2 – –
305 28.90 2.80 – 5.00 – 8.00 29 26.5 – –
Undaria pinnatifida 500 56.60 2.70 33.00 4.80 3.00 – – 47.70 – Cho et al. (2013)
Gracilaria edulis 450 30.90 2.20 16.5 2.80 4.40 7.60 – – 2.02 Roberts et al. (2015c)
Eucheuma spinosum 450 25.60 1.8 24.90 0.80 9.30 8.20 – – 30.30 –
Kappaphycus alvarezii 450 31.30 2.10 23.80 0.70 7.00 8.80 – – 2.24 –
Sargassum sp. 450 28.90 2.10 18.20 1.10 2.80 10.80 – – 7.46 –
Unadaria pinnatifida 450 34.80 2.80 15.60 2.40 0.80 9.90 – – 1.33 –
Saccharina japonica 450 28.00 1.90 16.40 2.20 1.00 11.00 – – 1.29 –
(Continued)
Protocols for Macroalgae Research
Table 10.1 (Continued) Main physicochemical characteristics of biochar generated from different biomass sources
and different pyrolysis conditions
CECc SAd
Feedstock PTa (°C) C (%) H (%) O (%) N (%) S (%) pH b (cmol kg–1) Ash (%) (m 2 g–1) References
Undaria pinnatifida 200 30.58 3.820 34.70 3.40 – 7.37 – 32.83 1.909 Jung et al. (2016b)
400 31.92 1.74 13.56 2.35 – 8.38 – 41.85 70.29 –
600 36.64 0.99 11.76 2.31 – 10.36 – 48.71 61.80 –
800 40.51 0.70 10.02 2.00 – 11.09 – 50.41 44.49 –
Ulva compressa 500 57.60 2.60 22.40 5.60 11.80 – 157 43.30 – Kim et al. (2016)
Porphyra tenera 500 74.70 3.00 9.00 9.10 4.20 – – 21.30 – Park et al. (2016)
Oedogonium 450 49.80 – – 5.50 1.00 10.60 21.30 – – Cole et al. (2017)
Others
Poultry litter 400 – 3.08 15.62 4.48 – 9.49 48 – – Jassal et al. (2015)
Chapter ten: Biochar production from seaweeds
200 m
100 m
200 m
200 m 200 m
(b)
Figure 10.1 Open-air biomass dryer systems on a clean mesh (a) or hanging along
a rope (b).
300°C 450°C
600°C
Figure 10.2 Biochar obtained from Macrocystis pyrifera at 300°C, 450°C, and 600°C.
2. Wash the samples three times with deionized water (1:20 sample:water
ratio). The samples must then be filtered to obtain the solid product,
which must be subsequently dried in an oven at 60–80°C for 24 h
(Jung et al. 2015a; 2016b).
3. The obtained biochar must be cooled in a desiccator for 1 h and then
stored in sealed plastic bags at room temperature.
10.5 Notes
Note 1: To prevent the decomposition of algae biomass, refrigerated
storage and/or transportation should not exceed 48 h, whereas non-
refrigerated storage and/or transportation should not exceed 24 h.
Note 2: Initial washing is performed primarily to remove sand and
any other organisms. Maintaining the holdfast will facilitate algae
handling.
Note 3: Monitor and periodically move the algae biomass to ensure uni-
form drying (for ground or hanging drying methods). Algae bio-
mass must be covered with mesh for ground drying under windy
conditions. If a moisture content > 15% persists after two days, and if
environmental conditions will not improve, directly proceed to Step 4,
which will remove any moisture excess.
Note 4: If the next step will be carried out in less than 12 h, it is advisable
to store the biomass in a dry place so that they do not get wet again.
Note 5: Use perforated boxes or baskets to ensure that heat reaches all
algal tissue surfaces. If posterior experiments measure heavy metal
Chapter ten: Biochar production from seaweeds 183
contents, plastic boxes are advisable for the drying process. If pos-
terior experiments are related to the organic matter, stainless-steel
baskets are advisable for the drying process.
Note 6: If following analyses focus on heavy metals adsorption, grind-
ing the samples manually or with a mortar are recommended.
Note 7: An end pyrolysis temperature of 450°C is more efficient and
results in less solid biomass loss in the pyrolysis process (Bird et al.
2011). This temperature may vary depending on the purpose of the
study and pyrolysis conditions.
Note 8: To perform adsorption tests, it is advisable to grind up to pass
through a 0.5–1.0 mm sieve (Zhang et al. 2013). If the biochar will be
used to assess heavy metals adsorption, avoid crushing the samples
with metallic instruments. In the case of applying the biochar to the
soil, it a particle size of 0.5–2.0 mm is recommended (Bird et al. 2012;
Roberts et al. 2015c; Jung et al. 2016b).
Acknowledgments
This work was supported by FIC 30397482-0 Gobierno Regional de
Valparaíso and Project CONICYT FB-0002-2014 to LC-P. We acknowledge
the language support provided by Ashley VanCott, BioPub Ltd. We also
thank Ignacio Perez-Massad for providing Figure 10.1.
References
Bird, M.I., Wurster, C.M., de Paula, S.P.H., Bass, A.M., and de Nys, R. 2011. Algal
biochar production and properties. Bioresource Technol. 102:1886–1891.
Bird, M.I., Wurster, C.M., de Paula Silva, P.H., Paul, N.A., and de Nys, R. 2012.
Algal biochar: Effects and applications. GCB Bioenergy 4:61–69.
Bridgwater, A. and Peacocke, G. 2002. Fast pyrolysis processes for biomass.
Renew. Sust. Energy Rev. 4:1–73.
Case, S., McNamara, N.P., Reay, D., Stott, A., Grant, H., and Whitaker, J. 2015.
Biochar suppresses N2O emissions while maintaining N availability in a
sandy loam soil. Soil Biol. Biochem. 81:178–185.
Cha, A.B., Sung, H.P.C., Sang-Chul, J.C., Changkook, R.D., Jong-Ki, J.E. Min-Chul,
S.B., and Young-Kwon, P.A. 2016. Production and utilization of biochar: A
review. J. Ind. E. Chem. 40:1–15.
Cho, H.J., Baek, K., Jeon, J.-K., Park, S.H., Suh, D.J., and Park, Y.-K. 2013. Removal
characteristics of copper by marine macro-algae-derived chars. Chem. Eng. J.
217:205–211.
Cole, A.J., Paul, N.A., de Nys, R., and Roberts, D. 2017. Good for sewage treat-
ment and good for agriculture: Algal based compost and biochar. J. Environ.
Manag. 200:105–113.
Jassal, R., Johnson, M., Molodovskaya, M., Black, T.A., Jollymore, A., and Sveinson, K.
2015. Nitrogen enrichment potential of biochar in relation to pyrolysis tem-
perature and feedstock quality. J. Environ. Manag. 152:140–144.
184 Protocols for Macroalgae Research
Johansson, C.H., Paul, N.A., Nys, R., and Roberts, D.A. 2016. Simultaneous bio-
sorption of selenium, arsenic and molybdenum with modified algal-based
biochars. J. Environ. Manag. 165:117–123.
Jung, K.-W., Hwang, M.-H., Jeong, T.-U., and Ahn, K.-H. 2015a. A novel approach
for preparation of modified-biochar derived from marine macroalgae: Dual
purpose electro-modification for improvement of surface area and metal
impregnation. Bioresource Technol. 191:342–345.
Jung, K.-W., Jeong, T.-U., Hwang, M.-H. Kim, K., and Ahn, K.-H. 2015b. Phosphate
adsorption ability of biochar/Mg–Al assembled nanocomposites prepared
by aluminum-electrode based electro-assisted modification method with
MgCl2 as electrolyte. Bioresource Technol. 198:603–610.
Jung, K.W., and Kyu-Hong, A.K.-H. 2016a. Fabrication of porosity-enhanced MgO/
biochar for removal of phosphate from aqueous solution: Application of a
novel combined electrochemical modification method. Bioresource Technol.
200:1029–1032.
Jung, K.-W, Kim, K., Jeong, T.-U., and Ahn, K.-H. 2016b. Influence of pyrolysis
temperature on characteristics and phosphate adsorption capability of
biochar derived from waste-marine macroalgae (Undaria pinnatifida roots).
Bioresource Technol. 200:1024–1028.
Kim, B.-S., Lee, H.W., Park, S.H., Baek, K., Jeon, J.-K., Cho, H.J., Jung, S.-C., Kim,
S.C., and Park, Y.-K. 2016. Removal of Cu2+ by biochars derived from green
macroalgae. Environ. Sci. Pollut. Res. 23:985–994.
Lee, H., Park, R.-S., Lee, H.W., Hong, Y., Lee, Y., Park, S.H., Jung, S.C., Yoo, K.-S.,
Jeon, J.-K., and Park, Y.-K. 2016. Adsorptive removal of atmospheric pollut-
ants over Pyropia tenera chars. Carbon Lett. 19:79–88.
Lee, M.N. and Woo, S.H. 2014. Application of biochar produced from wood and
seaweed for the removal of dye in wastewater. 2nd FORBIOM Workshop
Potentials and Biochar to Mitigate Climate Change, Hanbat National University,
Korea.
Lehmann, J. and Joseph, S. 2009. Biochar for environmental management: An
Introduction. In Biochar for Environmental Management: Science and Technology,
Lehmann, J. and Joseph, S. (Eds.), pp. 1–9, Earthscan, London, UK.
Mandal, S., Thangarajan, R., Bolan, N.S., Sarkar, B., Khan, N., Ok, Y.S., and Naidu, R.
2016. Biochar-induced concomitant decrease in ammonia volatilization and
increase in nitrogen use efficiency by wheat. Chemosphere. 142:120–127.
Mašek, O., Brownsort, P., Cross, A., and Sohi, S. 2013. Influence of production con-
ditions on the yield and environmental stability of biochar. Fuel. 103:151–155.
Park, S.H., Cho, H.J., Ryu, C., and Park, Y.-K. 2016. Removal of copper (II) in aque-
ous solution using pyrolytic biochars derived from red macroalga Porphyra
tenera. J. Ind. Eng. Chem. 25:314–319.
Qian, K., Kumar, A., Zhang, H., Bellmer, D., and Huhnke, R. 2015. Recent advances
in utilization of biochar. Renew. Sustain. Energy Rev. 42:1055–1064.
Rajapaksha, A.U., Chen, S.S., Tsang, D.C.W., Ming, Z.M., Vithanage, M., Mandal, S.,
Gao, B., Bolanm N.S., and Ok, S.Y. 2016. Engineered/designer biochar for
contaminant removal/immobilization from soil and water: Potential and
implication of biochar modification. Chemosphere. 148:276–291.
Roberts, D.A., Paul, N.A., Bird, M.I., and de Nys, R. 2015a. Bioremediation for coal-
fired power stations using macroalgae. J. Environ. Manag. 153:25–32.
Chapter ten: Biochar production from seaweeds 185
Roberts, D.A., Paul, N.A., Cole, A.J., and de Nys, R. 2015b. From waste water treat-
ment to land management: Conversion of aquatic biomass to biochar for
soil amelioration and the fortification of crops with essential trace elements.
J. Environ. Manage. 157:60–68.
Roberts, D.A., Paul, N.A., Dworjanyn, S.A., Bird, M.I., and de Nys, R. 2015c. Biochar
from commercially cultivated seaweed for soil amelioration. Sci. Rep. 5:1–6.
Zhang, M., Gao, G., Varnoosfaderani, S., Hebard, A., Yao, Y., and Inyang, M. 2013.
Preparation and characterization of a novel magnetic biochar for arsenic
removal. Bioresource Technol. 130:457–462.
chapter eleven
Contents
11.1 Introduction......................................................................................... 187
11.2 State of the art ..................................................................................... 189
11.3 Materials .............................................................................................. 191
11.3.1 Extraction of laminarin from macroalgal material .......... 191
11.3.2 Precipitation of alginate ....................................................... 191
11.3.3 Liquid chromatography–mass spectrometrical
method measurement .......................................................... 192
11.4 Experimental procedures .................................................................. 192
11.4.1 Laminarin extraction............................................................ 192
11.4.2 LC–MS measurements ......................................................... 193
11.4.3 Laminarin reference and quantification............................ 194
11.5 Notes..................................................................................................... 195
Acknowledgment ........................................................................................... 196
References........................................................................................................ 197
11.1 Introduction
Brown algae exhibit a unique central storage carbon metabolism (Michel
et al. 2010). The photoassimilated d-fructose 6-phosphate is not used by
these algae to produce sucrose as in higher plants but is rather converted
into the polyol d-mannitol (Stenhouse 1844, Yamaguchi et al. 1966). Brown
algae lack or contain only very low concentrations of free sugars such as
glucose, fructose, or sucrose (Yamaguchi et al. 1966). These free sugars
most likely undergo immediate biochemical conversion into d-mannitol
and polysaccharides.
The long-term carbon storage polysaccharide in brown algae is lami-
narin, a vacuolar β-1,3-glucan, which chemically differs from storage prod-
ucts of most other living organisms that typically use glycogen or starch
187
188 Protocols for Macroalgae Research
CH2OH CH2OH
CH2OH
O O OH
O O
OH O
OH OH
OH OH OH
OH n
(a) G-chain laminarin
CH2OH
CH2OH O
O
O
OH O CH2(CHOH)4CH2OH
OH
Mannitol
OH OH
OH n
(b) M-chain laminarin
Detection
A B
Mass spectrometer:
Linear ion trap
LC–MS
Interface +
MS ion source
11.3 Materials
11.3.1 Extraction of laminarin from macroalgal material
• Freeze drier
• Laboratory mill
• Spatula
• Laboratory balance
• Pipettes
• 1 ml reaction vessels
• Ultrapure water
• Laboratory shaker
• Centrifuge (14,000 rpm) (Note 1)
1. For extraction, grind the samples (<1 mm grain size) with an analyti-
cal mill (Ika MF 10 Basic, Germany).
2. Extract the algal material (0.2 g DW) with ultrapure water (1.5 ml) on
a shaker (250 rpm) at 25°C for 2 h.
3. Centrifuge 1 ml of the water extract at 14,000 rpm for 5 min
(HettichMikro 22 R, Germany).
4. Transfer the supernatant into a 4 ml vial and add 1.5 ml of ultrapure
water. Then the procedure is repeated, and both supernatants are mixed
and converted into a microvial (300 µl) for direct LC–MS analysis.
5. If the algal sample contains a large amount of alginate, see Note 2.
Chapter eleven: Identification and quantification of laminarins 193
RT:0.00 − 9.99
2.54
100 2.74
80 Cystoseira tamariscifolia
60
40 3.10
20
Relative abundance
0.28 0.68 0.93 1.33 1.76 2.08 2.27 3.39 3.67 4.11 4.61 4.77 5.27 5.72 5.90 6.30 6.64 7.10 7.28 7.44 7.92 8.29 8.64 8.90 9.12 9.76 9.94
0
2.74
100
80 2.63 Saccharina latissima
60
40 3.70
20 3.28
0.26 0.39 1.04 1.29 1.45 1.67 2.18 4.06 4.30 4.53 4.90 5.27 5.72 5.91 6.34 6.82 7.09 7.38 7.76 8.25 8.59 9.13 9.48 9.69
0
3.70
100
80
Laminarin
60
40
20 2.87 2.97 3.57
0.15 0.39 0.90 1.06 1.47 1.98 2.33 2.68 4.15 4.34 4.61 5.09 5.58 6.01 6.33 6.49 6.76 6.89 7.32 7.80 7.94 8.48 8.80 9.31 9.87
0
0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 6.0 6.5 7.0 7.5 8.0 8.5 9.0 9.5
65
60
55
50
45 1790.31
1521.25
40
35
30
25
20
15 1469.02
2117.50
10 1844.47
2198.25
1414.52
5 2279.05
1360.58 2441.38 3418.90
0
600 800 1000 1200 1400 1600 1800 2000 2200 2400 2600 2800 3000 3200 3400 3600 3800 4000
m/z
(b)
11.5 Notes
Note 1: A hot standard extraction method completely extracts alginate
in addition to the laminarins, which results in very viscous extracts.
These extracts are completely unsuitable for LC–MS analysis. The
hot extraction follows a standardized microwave extraction method
for plant components in ultrapure water as a solvent. The conditions
for microwave extraction are 20% of 400 W (MarsXpress 5 CEM) at a
rate of 5°C min−1 to 60°C for 5 min. This microwave-based method is
very effective and can be used for fresh and freeze dried raw material
with water as the solvent. It takes only 20 min for a complete extrac-
tion. However, this standard extraction method is proved to be com-
pletely unsuitable for extraction of laminarin from brown macroalgal
samples.
196 Protocols for Macroalgae Research
Note 2: Certain brown macroalgal species (e.g., Fucales) contain very high
amounts of alginates. Thus, even the application of the described cold
water method extracts substantial amounts of alginate. The extrac-
tion of alginates can be recognized by a higher viscosity and a
visible turbidity of the extract. To avoid disturbance of the LC–MS
measurements, a method was developed to precipitate alginates.
From the aqueous extract, 500 µL is transferred into a reaction vessel,
and the alginates are precipitated with 450 µL ethanol (>99.7%), and
50 µL formic acid (98%). This mixture is mixed for 10 s (Vortex mixer),
then stored for 12 h in a refrigerator and finally centrifuged for 5 min
at 14,000 rpm (Hettich micro 22 R, Germany). The clear and liquid
supernatant is transferred into a microvial (300 µl) for later LC–MS
analysis. To achieve comparable analysis conditions, the commercial
laminarin substance used for the reference measurements is treated
in the same way.
Note 3: The received extracts have to be measured either immediately or
after 4 h. Longer storage of the extracts, even in the refrigerator, leads
to noticeable decomposition of the polysaccharides, and thus lower
laminarin content will be detected.
Note 4: LC–MS analytics have some particular characteristics, which have
to be considered. If chemically different laminarin species, number
of different chain lengths, and presence of branched chains have to
be studied and investigated with the HPLC-measuring system, we
recommend the use of a column for size-exclusion chromatography.
Therefore, we use for the chromatographic separation of individual
laminarin chains a Zenix SEC 150 column (Sepax Technologies, Inc.,
250 mm length, 4.6 mm ID, 3 µm particle size, 150 An intermediate vol-
ume) with ultrapure water and 0.1% formic acid as eluents. However,
for the quantitative determination of the total laminarin content, this
column is not suitable because an integration of split or shoulder peaks
is not possible even after smoothing. For quantification purposes, the
use of the column described in Methods is better suitable.
Note 5: During an analysis sequence with the LC–MS system, a lami-
narin reference sample has to be inserted as a quality control-run
(QC-run) after at least six algal sample runs. Despite careful sam-
ple preparation, both the spray shield and the transfer capillary of
the mass spectrometer get blocked relatively fast, which leads to a
smaller ion transfer into the mass spectrometer. A QC-run permits
clear measurements because of an unhindered ion transfer.
Acknowledgment
This research was funded by the Project BIOACID Phase II of the German
Federal Ministry of Education and Research (BMBF; FKZ 03F0655L).
Chapter eleven: Identification and quantification of laminarins 197
References
Alderkamp, A.-C., M. van Rijssel, and H. Bolhuis. 2007. Characterization of marine
bacteria and the activity of their enzyme systems involved in degradation of
the algal storage glucan laminarin. FEMS Microbiol. Ecol. 59:108–117.
Annan, W. D., E. Hirst, and D. J. Manners. 1965. 162. The constitution of lami-
narin. Part V. The location of 1,6-glucosidic linkages. J. Chem. Soc. 885–891.
doi:10.1039/JR9650000885.
Beattie, A., E. L. Hirst, and E. Percival. 1961. Studies on the metabolism of the
Chrysophyceae. Comparative structural investigations on leucosin
(Chrysolaminarin) separated from diatoms and laminarin from the brown
algae. Biochem. J. 79:531–537.
Chapman, A. R. O., and J. S. Craigie. 1977. Seasonal growth in Laminaria longi-
cruris: Relations with dissolved inorganic nutrients and internal reserves of
nitrogen. Mar. Biol. 40:197–205.
Chizhov, A. O., A. Dell, H. R. Morris et al. 1998. Structural analysis of laminarans
by MALDI and FAB mass spectrometry. Carbohydr. Res. 310:203–210.
Craigie, J. S. 1974. Storage products. In Algal Physiology and Biochemistry, W. P. D.
Stewart (Ed.), pp. 206–235. Berkley, CA: University of California Press.
Dethier, M. N., and S. L. Williams. 2009. Seasonal stresses shift optimal intertidal
algal habitats. Mar. Biol. 156:555–567.
Devillé, C., J. Damas, P. Forget, G. Dandrifosse, and O. Peulen. 2004. Laminarin in
the dietary fibre concept. J. Sci. Food Agricult. 84:1030–1038.
Gagné, J. A., K. H. Mann, and A. R. O. Chapman. 1982. Seasonal patterns of growth
and storage in Laminaria longicruris in relation to differing patterns of avail-
ability of nitrogen in the water. Mar. Biol. 69:91–101.
Gómez, I., and P. Huovinen. 2012. Morpho-functionality of carbon metabolism in
seaweeds. In Seaweed Biology: Novel Insights into Ecophysiology, Ecology and
Utilization, C. Wiencke, and K. Bischof (Eds.), pp. 25–46. Berlin, Germany:
Springer Verlag.
Gómez, I., and C. Wiencke. 1998. Seasonal changes in C, N and major organic
compounds and their significance to morpho-functional processes in the
endemic Antarctic brown alga Ascoseira mirabilis. Polar Biol. 19:115–124.
Graiff, A., W. Ruth, U. Kragl, and U. Karsten. 2016. Chemical characterization and
quantification of the brown algal storage compound laminarin—A new
methodological approach. J. Appl. Phycol. 28:533–543.
Johnston, C. S., R. G. Jones, and R. D. Hunt. 1977. A seasonal carbon budget for a
Laminarian population in a scottish sea-loch. Helgoländer Wissenschaftliche
Meeresuntersuchungen 30:527–545.
Michel, G., T. Tonon, D. Scornet, J. M. Cock, and B. Kloareg. 2010. Central and storage
carbon metabolism of the brown alga Ectocarpus siliculosus: Insights into the ori-
gin and evolution of storage carbohydrates in eukaryotes. New Phytol. 188:67–81.
Nelson, T. E., and B. A. Lewis. 1974. Separation and characterization of the soluble
and insoluble components of insoluble laminaran. Carbohydr. Res. 33:63–74.
Peat, S., W. J. Whelan, and H. G. Lawley. 1958. 141. The structure of laminarin.
Part I. The main polymeric linkage. J. Chem. Soc. 724–728. doi:10.1039/
JR9580000724.
Percival, E. G. V., and A. G. Ross. 1951. 156. The constitution of laminarin. Part II.
The soluble laminarin of Laminaria digitata. J. Chem. Soc. 720–726. doi:10.1039/
JR9510000720.
198 Protocols for Macroalgae Research
Read, S. M., G. Currie, and A. Bacic. 1996. Analysis of the structural heterogeneity
of laminarin by electrospray-ionisation-mass spectrometry. Carbohydr. Res.
281:187–201.
Rioux, L.-E., S. L. Turgeon, and M. Beaulieu. 2007. Characterization of polysaccha-
rides extracted from brown seaweeds. Carbohydr. Polm. 69:530–537.
Rioux, L.-E., S. L. Turgeon, and M. Beaulieu. 2010. Structural characterization of
laminaran and galactofucan extracted from the brown seaweed Saccharina
longicruris. Phytochem. 71:1586–1595.
Rupérez, P., O. Ahrazem, and J. A. Leal. 2002. Potential antioxidant capacity of sul-
fated polysaccharides from the edible marine brown seaweed Fucus vesiculo-
sus. J. Agr. Food Chem. 50:840–845.
Schiener, P., K. D. Black, M. S. Stanley, and D. H. Green. 2015. The seasonal varia-
tion in the chemical composition of the kelp species Laminaria digitata,
Laminaria hyperborea, Saccharina latissima and Alaria esculenta. J. Appl. Phycol.
27:363–373.
Schmiedeberg, W. 1885. Über die Bestandteile der Laminaria. Gesellschaft deutscher
Naturforscher und Ärzte, Leipzig, Tageblatt der 58. Versammlung, p. 427.
Stenhouse, J. 1844. Über das Vorkommen von Mannit in Laminaria saccharina und
einigen anderen Seegräsern. Justus Liebigs Annalen Der Chemie 51:349–354.
Templeton, D. W., M. Quinn, S. van Wychen, D. Hyman, and L. M. L. Laurens. 2012.
Separation and quantification of microalgal carbohydrates. J. Chromatogr. A.
1270:225–234.
Yamaguchi, T., T. Ikawa, and K. Nisizawa. 1966. Incorporation of radioactive car-
bon from H14CO3− into sugar constituents by a brown alga, Eisenia bicyclis,
during photosynthesis and its fate in the dark. Plant Cell Physiol. 7:217–229.
Yemm, E. W., and A. J. Willis. 1954. The estimation of carbohydrates in plant
extracts by anthrone. Biochem. J. 57:508–514.
Yvin, J.-C., F. LeVasseur, and F. Hud`Homme. 1999. Use of laminarin and oligosac-
charides derived there from in cosmetics and for preparing a skin treatment
drug. US Patent 5980916A.
chapter twelve
Determination of carbohydrate
composition of macroalgae
Wouter J.J. Huijgen, E.M. Cobussen-Pool,
B.F. van Egmond, and J.W. van Hal
Contents
12.1 Introduction ......................................................................................... 200
12.2 State of the art ...................................................................................... 201
12.3 Materials ............................................................................................... 202
12.3.1 Seaweed .................................................................................. 202
12.3.2 Reagents.................................................................................. 202
12.3.3 Equipment .............................................................................. 203
12.4 Experimental procedures................................................................... 203
12.4.1 Preparation seaweed sample ............................................... 204
12.4.2 Hydrolysis .............................................................................. 204
12.4.2.1 Prehydrolysis ........................................................ 204
12.4.2.2 Hydrolysis ............................................................. 205
12.4.3 Analysis monomeric carbohydrates ................................... 205
12.4.3.1 Sample preparation .............................................. 205
12.4.3.2 Gradient ................................................................. 206
12.4.4 Calculations ........................................................................... 208
12.5 Notes ..................................................................................................... 208
12.5.1 General remarks .................................................................... 208
12.5.2 Seaweed hydrolysis............................................................... 209
12.5.3 High-performance anion-exchange chromatography
with pulsed amperometric detector ................................... 209
Acknowledgments ......................................................................................... 210
References........................................................................................................ 210
199
200 Protocols for Macroalgae Research
12.1 Introduction
Carbohydrates are a major constituent of macroalgae (in addition to min-
erals and proteins) (Van Hal et al. 2014). In addition, carbohydrates are key
target components in the application of macroalgae for production of, for
example, biobased chemicals and biofuels (Van Hal et al. 2014). However,
a generally accepted protocol for adequate determination of the content
of individual carbohydrates is lacking. The EU-H2020 MacroFuels project
aims to develop such a protocol.
The carbohydrates that occur in seaweeds vary between the major
classes of seaweeds: that is, the brown seaweeds (Phaeophycaea includ-
ing Kelps), the red seaweeds (Rhodophyta), and the green seaweeds
(Chlorophyta). Within the group of red seaweeds, at least three different
main structural carbohydrates occur: that is, (1) xylan (e.g., Palmaria pal-
mata), (2) agar (e.g., Gracilaria sp.), and (3) carrageenan (e.g., Chondrus
crispus). Table 12.1 gives an overview of carbohydrates reported in the lit-
erature for the seaweeds used in our study.
Here, we present a method for determination of the carbohydrate com-
position of macroalgae based on hydrolysis and subsequent determina-
tion of resulting monosaccharides by high-performance anion-exchange
chromatography with pulsed amperometric detector (HPAEC–PAD). The
presented approach is intended to be uniform for all seaweeds, with opti-
mum hydrolysis conditions that have to be determined for each (class of)
seaweed. As an example, the optimum hydrolysis times for three differ-
ent seaweeds have been identified. This optimization procedure can also
be extended to other seaweeds. In addition, a gradient and elution order
for the HPAEC–PAD is described that allows determination of the major
sugar alcohols, reducing sugars, deoxy sugars, and uronic acids occurring
in tested seaweeds in a single run.
The reaction times of both steps have been varied to determine opti-
mum conditions. It was found that prehydrolysis is required to have ade-
quate hydrolysis of structural components such as cellulose and ulvan.
Optimum prehydrolysis time determined for all the three seaweeds was
60 min (identical to lignocellulosic biomass).
The optimum hydrolysis time was found to depend on both the sea-
weed type and specific monosaccharides of interest. For each monosac-
charide, a balance between formation and degradation (e.g., to furans),
which are both acid catalyzed, needs to be found. Recommended optimum
hydrolysis times based on the maximum total carbohydrate response are
given in Table 12.2.
202 Protocols for Macroalgae Research
12.3 Materials
12.3.1 Seaweed
• In the case of nondried seaweed, immediately dry it to prevent com-
positional changes.
• Optionally, remove large contaminants such as stones and snails.
• Cut seaweed, if needed.
• Dry at mild conditions, preferably by freeze-drying (alterna-
tively, in a [vacuum] oven at ≤50°C).
• Check moisture content. Ambient-dry conditions (about 5%–15%
moisture content, depending on seaweed species) are sufficient.
• Crush or cut dried seaweed to <1 cm and homogenize.
• Store the dried seaweed in closed containers at ambient temperature
until compositional analysis.
12.3.2 Reagents
• 72% w/w (12 M) sulfuric acid (stored at 4°C)
• BaCO3
• Bromophenol blue indicator (20 mg L –1)
• Standards analysis monomeric carbohydrates:
• Sugar alcohols:
– Mannitol
– Glycerol
• Reducing sugars:
– Glucose
– Xylose
– Arabinose (might be left out as not detected in seaweeds)
– Galactose
– Fructose (internal standard)
Chapter twelve: Determination of carbohydrate composition of macroalgae 203
• Deoxy sugars:
– Rhamnose (as L-rhamnose monohydrate)
– Fucose
• Uronic acids:
– Guluronic acid as the sodium salt (CarboSynth Limited, UK)
– Mannuronic acid as the sodium salt (CarboSynth Limited, UK)
– Iduronic acid (CarboSynth Limited, UK)
– Galacturonic acid
– Glucuronic acid
12.3.3 Equipment
• Halogen moisture analyzer.
• 50 mL centrifuge plastic tubes with a screw cap.
• Use centrifuge tubes that are heat resistant to maximum operat-
ing temperature (i.e. 100°C).
• Use centrifuge tubes with a cap that are liquid tight at elevated
temperature, to prevent leaking while shaking. Centrifuge tubes
with a flat screw cap tend to work fine.
• 10 mL plastic centrifuge tubes.
• Water bath set at 30°C.
• Water bath set at 100°C.
• Centrifuge applicable for 50 mL centrifuge tubes.
• Centrifuge applicable for 10 mL centrifuge tubes.
• HPAEC setup (Dionex, Sunnyvale, CA, USA) consisting of
• ICS3000 Ion Chromatography System
• CarboPac PA1 column (2 × 250 mm)
• CarboPac guard column (2 × 50 mm)
• Pulsed amperometric detector (PAD)
• Pump 1 (used for gradient over column, 0.25 mL/min)
– Eluent A: Ultrapure water
– Eluent B: 0.1 M NaOH in ultrapure water
– Eluent C: 0.1 M NaOH and 1 M NaAc in ultrapure water
• Pump 2 (used for postcolumn addition of NaOH, 0.15 mL min–1)
– 0.25 M NaOH in ultrapure water.
12.4.2 Hydrolysis
12.4.2.1 Prehydrolysis
1. Place tube with the sample in ice water and let it cool down (proper
cooling is crucial!).
2. Open the tube and carefully add 3 mL of prechilled 72% w/w sulfu-
ric acid.
3. Carefully mix the sample with the sulfuric acid using a sealed glass
Pasteur pipette. Leave the pipette in the tube.
4. Transfer the open tube to a preheated water bath and incubate for
60 min at 30°C (Note 6).
Chapter twelve: Determination of carbohydrate composition of macroalgae 205
5. Mix sample every 10–15 min using the sealed glass Pasteur pipette.
6. After incubation, transfer the tube to ice water to stop the prehy-
drolysis reaction.
12.4.2.2 Hydrolysis
1. Add demineralized H2O to the tube to a final volume of 30 mL.
While adding the water, clean the sealed glass Pasteur pipette and
take it out.
2. Close the tube with the screw cap and mix well by shaking.
3. Transfer tube, placed in a rack, to a preheated water bath, and incu-
bate at 100°C for 180 min (Laminaria digitata and Ulva lactuca) or
120 min (Palmaria palmata) (Notes 7 and 8).
4. Mix every 45–60 min by shaking. Be cautious and shake gently as
centrifuge tube is hot and tube and/or screw cap might have become
softer at elevated temperature (Note!)
5. After incubation, transfer the tube to ice water to cool down and stop
the hydrolysis.
6. Take tube out of ice water, dry the outside, and weigh the tube (mfinal).
7. Centrifuge tube for 5 min at 2500–3000 g at 20°C.
8. Alternatively, the tube can be placed overnight in a refrigerator after
initial cooling in ice water to cool down further and allow the resid-
ual solids to settle. In general, thus a clear supernatant is obtained,
and centrifugation is not needed.
9. Take a sample from the supernatant (Note 9).
10. Store sample in the freezer at −20°C until analysis. The sample
should preferably be analyzed within a few weeks but might be kept
for substantially longer times.
12.4.3.2 Gradient
After sample injection, while using 15 mM NaOH as mobile phase, pure
water is used first as eluent for successively sugar alcohols, deoxy sug-
ars, and reducing sugars and, subsequently, a gradient of NaOH and Na
acetate is used as eluent for uronic acids (Notes 10 and 11).
Glycerol
Mannitol
Fucose
Galactose
Rhamnose
Arabinose
Glucose
Xylose
Galacturonic acid
Mannuronic acid
Glucuronic acid
Guluronic acid
IS (Fructose)
1 2 3 4 5 6 7 8 9 10 11 12 13
0.1 2.0 4.0 6.0 8.0 10.0 12.0 14.0 16.0 18.0 20.0 22.0 24.0 26.0 28.0 30.0 32.0
t (min)
12.4.4 Calculations
The content of individual carbohydrates is calculated by
ccarb =
[carb] ⋅ ( mfinal − mempty ) ⋅ 100% (12.1)
mseaweed ⋅ ( 1 − xmoisture )
where:
ccarb is the content of carbohydrate in seaweed (% w/w on dry basis).
[carb] is the concentration carbohydrate in hydrolysate as determined
by HPAEC–PAD (g/g).
mfinal is the weight of the centrifuge tube and hydrolysate after two-
stage hydrolysis (g).
mempty is the weight of the empty centrifuge tube (g).
mseaweed is the weight of the seaweed sample used for hydrolysis (g).
xmoisture is the moisture content of the seaweed sample (−).
12.5 Notes
12.5.1 General remarks
Note 1: Perform compositional analysis at least in duplicate.
Note 2: The summative composition of seaweeds might be further com-
pleted by determination of, among others, proteins (e.g., based on
total nitrogen) (Safi et al. 2017), phenolic components (Hou et al.
2017), and ash (by calcination of seaweed at 550°C).
Note 3: In a compositional analysis of lignocellulosic biomass, recovery
factors are sometimes used to correct for acid degradation of formed
monosaccharides (Sluiter et al. 2010). A similar approach might be
followed for seaweeds, although it should be realized that (1) the
use of recovery factors tends to lead to an overestimation of the con-
tent of individual carbohydrates (Whitfield et al. 2016) and (2) uronic
acids are more sensitive to acid degradation than reducing sugars.
(Moreover, the sensitivity of each uronic acid toward acid degrada-
tion differs.)
Note 4: No distinction between different glucans in, for example, Kelps,
that is, laminarin and cellulose, can be made with this method. If
required, see Chapter 11 by Graiff et al. (2017).
Note 5: The presented protocol has not been verified for agarophytes,
such as Gracilaria sp., carrageenan-containing Rhodophyta such as
Chapter twelve: Determination of carbohydrate composition of macroalgae 209
Acknowledgments
The authors thank Mark Smit for his contribution to the experimental
work and Arjan Smit for his ideas and input. The current chapter is part
of the MacroFuels project. This project has received funding from the
European Union’s Horizon 2020 research and innovation program under
grant agreement No 654010.
References
Graiff, A., W. Ruth, and U. Karsten. 2017. Identification and quantification of
laminarin in brown algae. In Protocols for Macroalgae Research, B. Charrier,
T. Wichard, and C. R. K. Reddy, (Eds.), Chapter 11. Boca Raton, FL: CRC
Press, Taylor & Francis Group.
Hou, X., R. Neerup, and A. B. Bjerre. 2017. Total phenol content and antioxidant
capacity analysis of seaweed biomass. In Biotechnology Protocols for Macroalgae
Research, B. Charrier, T. Wichard, and C. R. K. Reddy (Eds.), Chapter 15. Boca
Raton, FL: CRC Press, Taylor & Francis Group.
Manns, D., A. L. Deutschle, B. Saake, and A. S. Meyer. 2014. Methodology for
quantitative determination of the carbohydrate composition of brown sea-
weeds (Laminariaceae). RSC Advances 4:25736–25746.
Safi, C., J. van Leeuwen, Y. Telleman, N. Engelen-Smit, L. van den Broek, and
P. Harmsen. 2017. Quantification of proteins in seaweeds. In Biotechnology
Protocols for Macroalgae Research, B. Charrier, T. Wichard, and C. R. K. Reddy
(Eds.), Chapter 13. Boca Raton, FL: CRC Press, Taylor & Francis Group.
Sluiter, J. B., R. O. Ruiz, C. J. Scarlata, A. D. Sluiter, and D. W. Templeton. 2010.
Compositional analysis of lignocellulosic feedstocks. 1. Review and descrip-
tion of methods. J. Agric. Food Chem. 58:9043–9053.
TAPPI (Technical Association of the Pulp and Paper Industry) 2009. Standard test
methods. http://www.tappi.org. Norcross, GA.
Van der Wal, H., B. L. H. M. Sperber, B. Houweling-Tan, R. R. C. Bakker,
W. Brandenburg, and A. M. López-Contreras. 2013. Production of acetone,
butanol, and ethanol from biomass of the green seaweed Ulva lactuca.
Bioresour. Technol. 128:431–437.
Van Hal, J. W., A. B. Bjerre, A. M. López-Contreras, M. Stanley, G. O. Hreggvidsson,
and W. J. J. Huijgen. 2016. Carbohydrate analysis of seaweed in the biorefin-
ery to chemicals and fuel context. Paper Presented at the International Seaweed
Symposium, Copenhagen, Denmark.
Van Hal, J. W., W. J. J. Huijgen, and A.M. López-Contreras. 2014. Opportunities
and challenges for seaweed in the biobased economy. Trends Biotechnol.
32:231–233.
Whitfield, M. B., M. S. Chinn, and M. W. Veal. 2016. Improvement of acid hydro-
lysis procedures for the composition analysis of herbaceous biomass. Ener.
Fuels 30:8260–8269.
Wildschut, J., A. T. Smit, J. H. Reith, and W. J. J. Huijgen. 2013. Ethanol-based
organosolv fractionation of wheat straw for the production of lignin and
enzymatically digestible cellulose. Bioresour. Technol. 135:58–66.
chapter thirteen
Quantification of
proteins in seaweeds
Carl Safi, Jelle van Leeuwen, Yvette Telleman,
Nicole Engelen-Smit, Lambertus van den Broek,
and Paulien Harmsen
Contents
13.1 Introduction ......................................................................................... 212
13.2 State of the art ...................................................................................... 212
13.3 Materials ............................................................................................... 213
13.3.1 Kjeldahl method: Total protein determination ................. 213
13.3.2 Dumas method: Total protein determination ................... 213
13.3.3 Amino acid determination ...................................................216
13.3.4 Nitrogen-to-protein-conversion factor determination......216
13.3.5 Lowry method: Soluble protein determination .................216
13.3.6 Bradford method: Soluble protein assay ........................... 217
13.4 Experimental procedures................................................................... 217
13.4.1 Kjeldahl method: Total protein determination ................. 217
13.4.1.1 Digestion ............................................................... 217
13.4.1.2 Distillation ............................................................ 217
13.4.1.3 Titration ................................................................. 218
13.4.1.4 Calculation ............................................................ 218
13.4.2 Dumas method: Total protein determination ................... 218
13.4.2.1 Procedure .............................................................. 218
13.4.3 Amino acid determination .................................................. 219
13.4.3.1 Preparation of reagents ....................................... 219
13.4.3.2 Preparation of eluents ......................................... 219
13.4.3.3 Preparation of standards .................................... 219
13.4.3.4 Acid hydrolysis..................................................... 220
13.4.3.5 Analysis ................................................................. 220
13.4.4 Nitrogen-to-protein-conversion factor determination..... 220
13.4.4.1 Procedure .............................................................. 220
13.4.5 Lowry method: Soluble protein determination ................ 221
13.4.5.1 Preparation of the standard solution ................ 221
211
212 Protocols for Macroalgae Research
13.1 Introduction
Seaweeds have been long exploited as a marine vegetable and food
additive owing to their rich composition of unique functional polysac-
charides, minerals, and proteins (Fujiwara-Arasaki et al. 1984; Fleurence
1999). Though proteins are an important component of seaweed compo-
sition, additional value can be created by extracting proteins together
with high-value compounds, such as health-improving components
and hydrocolloids (Jung et al. 2013). Aside from the economic benefits of
protein extraction, seaweed-based proteins can also alleviate the grow-
ing global demand for protein that is expected in the coming years.
As seaweeds only need water, CO2, and the fertilizers present in the
sea, they can also contribute to reducing the environmental hazards
associated with the current production of animal proteins (Steinfeld
et al. 2006; Gerber et al. 2013). The protein content of seaweeds varies
through species, cultivation location, and season (Black 1950, Patarra
et al. 2011). Studies have shown that the protein content in green spe-
cies can fluctuate from 5% to 32% (w/dw), brown species from 3% to 16%
(w/dw), and red seaweeds can contain from 2% to 29% (w/dw) protein
at 90% confidence level (Angell et al. 2016). Combining the interest in
seaweed protein with these observed fluctuations stresses, the need for
reliable methods to determine the protein content in seaweed samples
and fractions.
assays such as Lowry and Bradford (Barbarino and Lourenco 2005). Here,
the presence of the peptide bonds or the presence of certain AAs plays a
role. Another approach is the determination of the total amount of nitrogen
present in your sample. Kjeldahl and Dumas can be used to estimate the
total nitrogen content, which can be converted into total protein content.
This is because each peptide bond contains a nitrogen atom, although
some AAs have an additional nitrogen atom in their structure (e.g.,
arginine and histidine). A nitrogen-to-protein (NTP)-conversion factor
is required, and this is calculated from the total amount of AAs present
in the sample (Shuuluka et al. 2013; Akgul et al. 2015; Angell et al. 2016;
Biancarosa et al. 2017). If insoluble proteins are present, it is not possible to
use the colorimetric assays to determine the total protein content because
the peptide bonds and/or AAs are hardly accessible and are more or less
contained in the matrix. In these cases, Kjeldahl or Dumas method is the
most suitable one. Be aware that you should be careful to compare the
outcome of the different protein assays with each other as the methods
are based on different principles and as such can result in different values
(Barbarino and Lourenco 2005). For total proteins, especially Kjeldahl is
most often used, whereas for soluble protein Lowry is the preferred protein
method. On the other hand, Dumas (total protein) and Bradford (soluble
protein) are also suitable, and their advantage is the short assay time. All
methods described in this chapter are applicable to all seaweed species
regardless of its color (red, green, or brown) or structural characteristics.
Furthermore, despite the satisfying efficiency these methods exhibit, they
also feature weaknesses that should also be taken into consideration
(Table 13.1).
13.3 Materials
13.3.1 Kjeldahl method: Total protein determination
• Sulfuric acid: 95%–97% sulfuric acid.
• Kjeltab: Kjeltabs Type CK, each tablet contains: 3.5 g K2SO4 + 0.4 g
CuSO4·5H2O.
• Boric acid: Boric acid is dissolved in a solution of 1%. To prepare the
solution, 50 g boric acid is dissolved in 5 L distilled water.
• Sodium hydroxide: 33% NaOH with 16.5 g L–1 sodium thiosulfate.
• Hydrochloric acid: 0.1 M (0.1 N) HCl.
Table 13.1 Screening of the main advantages and disadvantages of the methods described in this chapter
Methods Advantages Disadvantages
Kjeldahl Suitable for both dry and liquid samples Time-consuming
Measures small amount samples, if needed Measures total nitrogen including nonprotein nitrogen
Measures total proteins (soluble and insoluble) Generates a lot of waste
Dumas Suitable for both dry and liquid samples Measures total nitrogen, including nonprotein nitrogen
Measures total proteins (soluble and insoluble) A drying step must be performed before the analysis step of
the method
Amino acids Accurate measurement Time-consuming
determination The acid hydrolysis denatures tryptophan and renders very
difficult for the determination of cysteine
NTP Gives more accurate protein measurements It is dependent on the quality of the amino acid analysis
calculation It only targets protein nitrogen previously performed
The NTP is highly dependent on the nitrogen-containing
components present in the sample
Its value can vary between seaweed species, growth conditions,
and growth phase at which the seaweed has been harvested
(Continued)
Protocols for Macroalgae Research
Table 13.1 (Continued) Screening of the main advantages and disadvantages of the methods described in this chapter
Methods Advantages Disadvantages
Lowry Selective measurement given that it only Risk of no color formation because of the interference of other
determines soluble proteins substances such as chelating agents
Measures/detects very small amount of The formation of purple color instead of blue is due to the
protein present in the sample presence of reducing agents or thiol in the sample
The formation of precipitate is because of the presence of
surfactant in the sample
Bradford Fast and simple assay Detergents such as SDS and/or basic conditions affect the color
Measures very small amount of protein present in The proteins should contain arginine and/or aromatic residues
the sample as the dye binds to these amino acids to form the color
The color is not affected by the presence of reducing The calculation of the amount of proteins is dependent on the
Chapter thirteen: Quantification of proteins in seaweeds
agents such as DTT and β-mercaptoethanol type of standard protein used in the assay
215
216 Protocols for Macroalgae Research
13.4.1.1 Digestion
1. Switch on the heater and set the temperature at 420°C.
2. Weigh the sample in the tubes and add a Kjeltabs tablet and 9 mL
H2SO4 (Notes 1–3).
3. Once the heater is at the desired temperature, insert the tubes and
put the exhaust system on the top.
4. Switch on the scrubber and water the scrubber.
5. Leave the tubes for 50 min in the heater.
6. Switch off the scrubber and lift the rack with the tubes.
7. Leave the exhaust system on top until no further fumes come out.
8. Add 75 mL distilled water in each tube (maximum cooling time is
10 min).
9. The samples are ready for the next step: distillation.
13.4.1.2 Distillation
1. Switch on the VAPODEST 45 and the water.
2. Double check if the vessels are full with distilled water, boric acid,
and sodium hydroxide.
3. Put a tube with 75 mL water in the machine and an empty Erlenmeyer.
This tube is only to warm up the machine.
4. Select the distillation program on the screen and press enter to start
measuring.
5. Wait for 5 min, and the sample is ready.
6. Put a new tube with water and an empty Erlenmeyer to determine
the blank value.
218 Protocols for Macroalgae Research
7. Repeat this step until the value will be 0.12–0.15 M HCl (titration).
8. Now tubes with the protein samples can be measured.
9. After all samples are tested, activate the cleaning system.
13.4.1.3 Titration
1. Always start with calibrating the electrode by selecting the calibra-
tion mode.
2. Set the temperature at 25°C.
3. First, calibrate with the pH 7 buffer, then calibrate with the pH 4
buffer.
4. After the calibration step is over, select Kjeldahl method and start
with titration.
5. Record the amount of HCl (mL) titrated.
13.4.1.4 Calculation
Use the following equation:
13.4.2.1 Procedure
1. Dry the samples completely before analysis, to guarantee complete
combustion (Note 4). However, applying very high temperatures is
discouraged to prevent evaporation of ammonium or nitrous gas.
Usually, lyophilization is used, and if this is not possible, low-tem-
perature drying (60°C–80°C) is to be allowed.
2. Make a calibration curve with the stock chemical containing nitro-
gen from 1 to 20 mg and should cover the detection range.
3. Weigh the samples in aluminum or tin cups folded shut to prevent
sample loss or adsorption of water from the air. For optimal accuracy,
Chapter thirteen: Quantification of proteins in seaweeds 219
13.4.3.5 Analysis
1. Set the column temperature at 50°C.
2. UV_1 set at wavelength 338 nm.
3. UV_2 set at wavelength 263 nm.
4. Flow gradient (Table 13.2).
6. If the NTP conversion factor is known (Note 9), then you can calcu-
late the amount of proteinogenic protein in your sample by multi-
plying the nitrogen content of your sample (obtained by Kjeldahl or
Dumas) with the NTP conversion factor.
13.4.6.1 Procedure
1. Pipette 30 µL sample or standard into a 1 mL plastic cuvette (Note 15).
2. Add 1.0 mL Bradford reagents and mix well (Note 16).
3. Measure the absorbance at 595 nm.
4. Plot the calibration curve (at least five different BSA concentrations
in duplicate) to determine the protein concentration of the unknown
samples (Note 17).
13.5 Notes
Note 1: To keep the measurement within the appropriate range, there
must be between 10 and 100 mg seaweed protein in the sample.
Note 2: It is possible to use liquid samples in this method. Therefore,
the digestion part of the protocol has to be changed by following the
next three steps:
a. Incubate the sample for 1 h at 180°C.
b. Then incubate the same sample for 1 h at 280°C.
c. Finally, incubate the same sample for 50 min at 420°C.
Chapter thirteen: Quantification of proteins in seaweeds 223
Note 3: Another alternative is that the liquid sample can be put in the
tube and dried at 105°C overnight. Afterward, it can be treated as a
solid sample.
Note 4: For seaweeds, inhomogeneity can be an issue, which could
partially be solved by (freeze-) drying a larger batch of material and
grinding the dried material.
Note 5: Make sure to add ethanethiol inside the fume hood.
Note 6: Not all the AAs present in the seaweed will survive the hydro-
lysis process.
Note 7: Asparagine is converted into aspartic acid and glutamine into
glutamic acid.
Note 8: Tryptophan is completely destroyed, and it is impossible to
accurately determine cysteine concentrations in acid hydrolyzed
samples.
Note 9: Be aware that the NTP conversion factor for Kjeldahl and Dumas
slightly differ as both have another principle to determine the total
nitrogen content.
Note 10: Use a new pipette for each reagent and sample, do not use the
same pipette for all the samples and reagents.
Note 11: While adding the reagents, it is essential to respect the waiting
time between each sample.
Note 12: Respecting the incubation time is important.
Note 13: If the solution is not turning blue, it means that there is
interference from undesired components in the sample such as che-
lating agents.
Note 14: If precipitate forms while adding the reagents, this implies that
the samples contain a surfactant or potassium ions.
Note 15: Dissolve the BSA well in glassware. Dissolving needs some
time, so only shake gently afterward. Stirring will result in foaming.
Note 16: Coomassie Brilliant Blue G-250 stains glass or quartz cuvettes;
the use of disposable polystyrene cuvettes is therefore recommended.
Note 17: Basic conditions and detergents influence the outcome negatively.
Acknowledgment
The authors would acknowledge MacroFuels project. This project has
received funding from the European Union’s Horizon 2020 research and
innovation program under grant agreement No 654010.
References
Akgul, R., Kizilkaya, B., Akgul, F. et al. 2015. Crude protein values and amino acid
profiles of some red algae collected from Canakkale. Indian J. Geo. Mar. Sci.
44: 527–530.
224 Protocols for Macroalgae Research
Angell, A.R., Mata, L., de Nys, R. et al. 2016. The protein content of seaweeds:
A universal nitrogen-to-protein conversion factor of five. J. Appl. Phycol. 28:
511–524.
Angell, A.R., Paul, N.A., and de Nys, R. 2017. A comparison of protocols for isolat-
ing and concentrating protein from the green seaweed Ulva ohnoi. J. Appl.
Phycol. 29: 1011–1026.
Barbarino, E. and Lourenco, S.O. 2005. An evaluation of methods for extraction
and quantification of protein from marine macro- and microalgae. J. Appl.
Phycol. 17: 447–460.
Biancarosa, I., Espe, M., Bruckner, C.G. et al. 2017. Amino acid composition, pro-
tein content, and nitrogen-to-protein conversion factors of 21 seaweed spe-
cies from Norwegian waters. J. Appl. Phycol. 29: 1001–1009.
Black, W.A.P. 1950. The seasonal variation in weight and chemical composition of
the common British Laminariaceae. J. Mar. Biol. Assoc. UK 29: 45–72.
Bradford, M.M. 1976. Rapid and sensitive method for quantitation of microgram
quantities of utilizing principle of protein-dye binding. Anal. Biochem. 72:
248–254.
Fleurence, J. 1999. Seaweed proteins: Biochemical, nutritional aspects and
potential uses. Trends. Food. Sci. Tech. 10: 25–28.
Fujiwara-Arasaki, T., Mino, N., and Kuroda, M. 1984. The protein value in human
nutrition of edible marine algae in Japan. Hydrobiologia 116: 513–516.
Gerber, P.J., Steinfeld, H., Henderson, B. et al. 2013. Tackling Climate Change Through
Livestock: A global Assessment of Emissions and Mitigation Opportunities. Food
and Agriculture Organization of the United Nations (FAO), Rome, Italy.
Jung, K.A., Lim, S.R., Kim, Y. et al. 2013. Potentials of macroalgae as feedstocks for
biorefinery. Bioresour. Technol. 135: 182–190.
Meussen, B.J., Zeeland, A.N.T., Bruins, M.E. et al. 2014. A fast and accurate UPLC
method for analysis of proteinogenic amino acids. Food Anal. Method 7:
1047–1055.
Patarra, R., Paiva, L., Net, A.I. et al. 2011. Nutritional value of selected macroalgae.
J. Appl. Phycol. 23: 205–208.
Shuuluka, D., Bolton, J.J., and Anderson, R.J. 2013. Protein content, amino acid
composition and nitrogen-to-protein conversion factors of Ulva rigida and
Ulva capensis from natural populations and Ulva lactuca from an aquaculture
system, in South Africa. J. Appl. Phycol. 25: 677–685.
Steinfeld, H., Gerber, P., Wassenaar, T. et al. 2006. Livestock’s long shadow:
Environmental issues and options. Food and Agriculture Organization of the
United Nations (FAO), Roman, Italy.
Vilg, J.V. and Undeland, I. 2017. pH-driven solubilization and isoelectric precipi-
tation of proteins from the brown seaweed Saccharina latissima-effects of
osmotic shock, water volume and temperature. J. Appl. Phycol. 29: 585–593.
chapter fourteen
Comprehensive phytohormone
quantification in the red
alga Pyropia yezoensis by
liquid chromatography–mass
spectrometry
Takakazu Matsuura, Izumi C. Mori, Yoko Ikeda,
Takashi Hirayama, and Koji Mikami
Contents
14.1 Introduction ......................................................................................... 226
14.2 State of the art ...................................................................................... 226
14.3 Materials ............................................................................................... 227
14.3.1 Extraction and purification of phytohormones ................ 227
14.3.2 Liquid chromatography–mass spectrometry ................... 228
14.4 Experimental procedures................................................................... 228
14.4.1 Extraction ............................................................................... 229
14.4.2 Partial purification of phytohormones with
solid- phase extraction .......................................................... 229
14.4.3 Liquid chromatography–mass spectrometry analysis .... 231
14.4.4 Data analysis .......................................................................... 234
14.5 Notes ..................................................................................................... 234
References........................................................................................................ 236
225
226 Protocols for Macroalgae Research
14.1 Introduction
Phytohormones are structurally unrelated compounds, including auxin,
abscisic acid (ABA), gibberellins, cytokinins, ethylene, jasmonates, sali-
cylic acid (SA), brassinosteroids, and strigolactones (Kende and Zeevaart
1997; Kamiya 2010). Emerging studies suggest the critical involvement
of these phytohormones in the regulation of growth, development, and
environmental stress response in plants (Weyers and Paterson 2001;
Vanstraelen and Benková 2012). To date, phytohormones are known to
exist in seaweeds as in terrestrial plants (Basu et al. 2002; Le Bail et al.
2010; Wang et al. 2014; Mikami et al. 2016), although functions of phyto-
hormones are not entirely understood.
Analysis of phytohormones in algae has a long history, and small
molecules identical to phytohormones of terrestrial plants have been
detected in a variety of algae (reviewed in Bradley 1991; Evans and
Trewavas 1991; Tarakhovskaya et al. 2007; Lu and Xu 2015). As the pre-
vious analyses generally focused on individual phytohormones, the
overall composition of phytohormones within a given algal species
has not been elucidated well. However, efforts have made to develop
methods employing gas chromatography–tandem mass spectrogram
and liquid chromatography–tandem mass spectrometry (LC–MS/MS)
for comprehensive phytohormone analysis in algae, and phytohor-
mone compositions have been characterized in a few algae (Yokoya
et al. 2010; Gupta et al. 2011; Wang et al. 2014). In addition, we recently
improved comprehensive analysis methods for phytohormones by liquid
chromatography– electrospray ionization–tandem mass spectroscopy
(LC–ESI–MS/MS) using the red seaweeds Pyropia yezoensis and Bangia
fuscopurpurea (Mikami et al. 2016; Mori et al. 2017). On the basis of our
current findings, we describe an updated procedure for comprehensive
analysis of phytohormones in P. yezoenssis as a model.
14.3 Materials
14.3.1 Extraction and purification of phytohormones
• Extraction solvent: Mixture of acetonitrile (MeCN)/water (80:20) con-
taining 1% (v/v) acetic acid (AcOH). Prepare daily.
• Internal standard stock solution: Prepare a stock solution of the extrac-
tion solvent containing all four internal standards in amounts
shown in Table 14.1. The sources of the internal standards are shown
in Table 14.1. Store the internal standard stock solution in a light-
proof bottle and replace after approximately six months.
• Solid-phase extraction cartridges:
• Oasis® HLB solid-phase extraction cartridge (1 cm3, Waters,
Milford, MA, USA)
• Oasis® MCX solid-phase extraction cartridge (1 cm3, Waters,
Milford, MA, USA)
• Oasis® WAX solid-phase extraction cartridge (1 cm3, Waters,
Milford, MA, USA).
• Solvents for solid-phase extraction:
• MeCN (HPLC grade or higher)
• Methanol (HPLC grade or higher, MeOH)
• 0.1 N HCl
• 0.1 N KOH
14.4.1 Extraction
All procedures should be carried out in a chemical hood:
Acidic extract
Oasis® WAX
Oasis® MCX
Oasis® HLB
LC method 1
100
0
0 10 20 30
LC method 2
100
0.25 ml min−1
50
0
0 10 20 30
LC method 3
100
0
0 10 20
Time (min)
Figure 14.2 Parameters for liquid chromatography. LC method 1 is used for the
analysis of indoleacetic acid and abscisic acid. LC methods 2 and 3 are used for
analyses of isopentenyladenine and salicylic acid, respectively. The proportion of
solvent B increases from 3% to 55% in 22 min in LC method 1. It increases from
3% to 97% in 16 min in LC method 2, and from 3% to 98% in 10 min in LC method
3. After a wash of the column with a high concentration of solvent B for approxi-
mately 5 min, the column is washed with 3% solvent B to condition the column.
LC, liquid chromatography. AcOH, acetic acid. MeCN, acetonitrile. MeOH, meth-
anol. FA, formic acid. The flow rate of LC methods 1 and 3 is 0.4 mL min−1, and that
of LC method 2 is 0.25 mL min−1.
Table 14.2 Parameters for mass spectrometry for quantification of IAA, ABA, iP, and SA
Retention time Time segment Multiple reaction Fragmentor Collision
Compound LC method (min) (min) ESI mode monitoring transition (m/z) voltage (V) energy (V)
IAA 1 9.7 9.3–11.8 Positive 176/130 90 15
D7-IAA 183/135, 136, 137
ABA 1 12.6 11.8–13.5 Negative 263/153 130 4
D6-ABA 269/159
iP 2 12.5 11–16 Positive 204/136 110 11
D6-iP 210/137
SA 3 5.6 2–10 Negative 137/93 90 12
D4-SA 141/97
See Figure 14.2 for LC methods.
Chapter fourteen: Comprehensive phytohormone quantification
233
234 Protocols for Macroalgae Research
14.5 Notes
Note 1: The dilution factor can be modified according to the actual con-
centrations of plant hormones detected in the algal sample.
Note 2: Grinding the tissue with stainless steel beads or tungsten beads
can reduce or abolish the detection of some hormones. This may be
because of tight binding of hormones to the beads and/or scraping
the plastic tube, which increases the surface area of plastic.
Note 3: The extracts may become very viscous and aggregate during the
suspension of powdered tissue in the extraction solvent. The extracts
will become more fluid as incubation progresses. Stir the extracts in
the mortar intermittently to disperse the aggregates.
Note 4: Samples can be stored at −20°C for a week or 4°C for overnight
at this step.
Note 5: The volume of the aqueous solution should be adjusted to
1 mL with 1% AcOH, if the volume of the solution is less than that.
Hormones may be lost during loading.
Note 6: The addition of 0.6 mL of 1% AcOH aqueous solution prevents
the sample from overdrying.
Note 7: Fill the cartridge up to the mouth with 1% AcOH aqueous solu-
tion to remove any remaining drops of HCl or KOH on the wall of
the cartridge that could affect binding of chemicals to the cartridge.
Note 8: As SA binds to Oasis® WAX cartridge very strongly, the recov-
ery rate is very low. Therefore, it is analyzed by using the eluate from
Oasis® MCX cartridge by LC–MS. SA can be eluted by 2% FA/98%
MeCN.
Note 9: Parameters for phytohormones that are not detected in P. yezoen-
sis but were examined in a previous paper (Mikami et al. 2016) are
listed in Table 14.3.
Table 14.3 Parameters for mass spectrometer for quantification of phytohormones measured in Mikami et al. (2016) and
12-oxo-phytodienoic acid (OPDA), which were not detected in gametophytes and sporophytes of Pyropia yezoensis
Retention Time segment Multiple reaction Fragmentor Collision
Compound LC method time (min) (min) ESI mode monitoring transition (m/z) voltage (V) energy (V)
GA1 1 8.9 8–9.3 Negative 347/273 160 18
D2-GA1 349/275
JA 1 14.4 13.5–15.7 Negative 209/59 135 11
D2-JA 211/59
GA4 1 17 16–17.3 Negative 331/257 160 20
D2-GA4 333/259
JA-Ile 1 16.2 17.5–20 Negative 322/130 140 14
13C -JA-Ile 328/136
6
OPDA 1 22.4 20–25 Negative 291/165 150 11
D5-OPDA 296/170
tZ 2 8.3 5–11 Positive 220/136 100 12
D5-tZ 225/136, 137
Chapter fourteen: Comprehensive phytohormone quantification
References
Basu, S., Sun, H., Brian, L., Quatrano, R.L. and Muday, G.K. 2002. Early embryo
development in Fucus distichus is auxin sensitive. Plant Physiol. 130:292–302.
Bradley, P.M. 1991. Plant hormones do have a role in controlling growth and devel-
opment of algae. J. Phycol. 27:317–321.
Evans, L.V. and Trewavas, A.J. 1991. Is algal development controlled by plant
growth substances? J. Phycol. 27:322–326.
Gupta, V., Kumar, M., Brahmbhatt, H., Reddy, C.R., Seth, A. and Jha, B. 2011.
Simultaneous determination of different endogenetic plant growth regula-
tors in common green seaweeds using dispersive liquid–liquid microextrac-
tion method. Plant Physiol. Biochem. 49:1259–1263.
Kamiya, Y. 2010. Plant hormones: Versatile regulators of plant growth and devel-
opment. Annu. Rev. Plant Biol. 61:Special Online Compilation.
Kende, H. and J.A.D. Zeevaart. 1997. The five “classical” plant hormones. Plant Cell
9:1197–1210.
Le Bail, A., Billoud, B., Kowalczyk, N., Kowalczyk, M., Gicquel, M., Le Panse, S.,
Stewart, S. et al. 2010. Auxin metabolism and function in the multicellular
brown alga Ectocarpus siliculosus. Plant Physiol. 153:128–144.
Lu, T. and Xu, J. 2015. Phytohormones in microalgae: A new opportunity for
microalgal biotechnology? Trends Plant Sci. 20:275–282.
Mikami, K., Mori, I.C., Matsuura, T., Ikeda, Y., Kojima, M., Sakakibara, H. and
Hirayama, T. 2016. Comprehensive quantification and genome survey reveal
the presence of novel phytohormone action modes in red seaweeds. J. Appl.
Phycol. 28:2539–2548.
Mori, I.C., Ikeda, Y., Matsuura, T., Hirayama, T. and Mikami, K. 2017. Phytohormones
in red seaweeds: A technical review of methods for analysis and a consider-
ation of genomic data. Bot. Mar. 60:153–170.
Tarakhovskaya, E.R., Maslov, Y.I. and Shishova, M.F. 2007. Phytohormones in
algae. Russ. J. Plant Physiol. 54:163–170.
Vanstraelen, M. and Benková, E. 2012. Hormonal interactions in the regulation of
plant development. Annu. Rev. Cell Dev. Biol. 28:463–487.
Wang, X., Zhao, P., Liu, X., Chen, J., Xu, J., Chen, H. and Yan, X. 2014. Quantitative
profiling method for phytohormones and betaines in algae by liquid chro-
matography electrospray ionization tandem mass spectrometry. Biomed.
Chromatogr. 28:275–280.
Weyers, J.D.B. and Paterson, N.W. 2001. Plant hormones and the control of physi-
ological processes. New Phytol. 152:375–407.
Yokoya, N.S., Stirk, W.A., van Staden, J., Novák, O., Turečková, V., Pěnčík, A. and
Strnad, M. 2010. Endogenous cytokinins, auxins, and abscisic acid in red
algae from Brazil. J. Phycol. 46:1198–1205.
chapter fifteen
Contents
15.1 Introduction ......................................................................................... 237
15.2 State of the art ...................................................................................... 238
15.3 Materials ............................................................................................... 239
15.3.1 Seaweed biomass .................................................................. 239
15.3.2 Equipment .............................................................................. 240
15.3.3 Chemicals ............................................................................... 240
15.3.3.1 Total phenolic content analysis of seaweed
biomass .................................................................. 240
15.3.3.2 Antioxidant capacity analysis of seaweed
biomass: 2,2-Diphenyl-1-picrylhydrazyl
assay ....................................................................... 241
15.4 Experimental procedures................................................................... 241
15.4.1 Total phenolic content analysis of seaweed biomass ....... 241
15.4.1.1 Folin–Ciocalteu assay .......................................... 241
15.4.1.2 Prussian blue assay .............................................. 243
15.4.2 Antioxidant capacity analysis of seaweed biomass:
2,2-Diphenyl-1-picrylhydrazyl assay .................................. 245
15.5 Tips and tricks ..................................................................................... 247
Acknowledgment ........................................................................................... 247
References........................................................................................................ 247
15.1 Introduction
Seaweed is a known source of value-added components such as dietary
fibers, proteins, minerals, vitamins, and phenolic compounds. These phe-
nolic compounds are considered as one of the most important classes
of natural antioxidants (Machu et al. 2015). A worldwide trend to apply
237
238 Protocols for Macroalgae Research
polyphenols but could also react with some other reducing compounds
that possibly exist in the crude extracts, and thus skew the results of TPC
determination. To increase the specificity for TPC determination, increasing
the purification of phenolic compounds extract is one of the possibilities.
Another colorimetric method commonly used for determining the TPC is
the PB assay (Price and Butler 1977). This method is based on the color-
formation reaction between the phenolic compounds and ferricyanide ion
(Fe(CN)6)3−. Phenols are oxidized, whereas Fe(CN)63− is reduced to the fer-
rocyanide ion Fe(CN)64−. Fe(CN)64− then reacts with the ferric ion (Fe3+) to
form PB metallic complex, Fe4[Fe(CN)6]3. Compared with the FC assay,
it has the advantages of lower time consumption and higher selectivity
(Margraf et al. 2015). The main drawbacks of the original PB assay (Price
and Butler 1977) are the formation of precipitates after incubation and an
increase in color intensity with time. Graham (1992) developed a stabilized
reagent in combination of phosphoric acid (or tetrasodium ethylenediami-
netetraacetate) and gum arabic: Phosphoric acid (or tetrasodium ethyl-
enediaminetetraacetate) is used for stopping the reaction by complexing
the excess of Fe3+ in the mixture, and gum arabic is used for preventing
the precipitation by impeding the coalescence of the colloidal particles.
However, running a preanalysis to ensure a proper phenolic concentration
range is recommended for preventing the potential precipitation of the PB
metallic complex, especially when the phenolic content of sample is high.
Although the selectivity of phenolic compound is higher in PB assay than
in FC assay (Margraf et al. 2015), the purity of the extract still affects the
measured result of TPC from PB assay.
The antioxidant capacity of extracted phenolic compounds from sea-
weed can be determined by, for example, DPPH assay (first developed
by Blois 1958). This assay is one of the most widely used assays and has
been modified and adapted in many different natural antioxidant stud-
ies, because of its simplicity, speed, and sensitivity (Kedare and Singh
2011). DPPH• (free radical) is a stable and commercially available organic
nitrogen radical that shows a strong absorption maximum at 517 nm
(purple). The color turns from purple to yellow followed by the formation
of DPPH by trapping hydrogen from a hydrogen donor (i.e., antioxidant).
The antioxidant effect is proportional to the disappearance of DPPH• in
test samples and can be spectrophotometrically monitored at 517 nm.
15.3 Materials
15.3.1 Seaweed biomass
Fresh or defrosted seaweed biomass is dried in a 50°C drying oven for
two nights (until the biomass dry matter content reaches ~90%) and is then
ground and passed through a Ø = 1.0 mm screen.
240 Protocols for Macroalgae Research
15.3.2 Equipment
• A UV–vis spectrophotometer with a 96-well flat-bottom microtiter
plate reader.
• Moisture content analyzer.
• Thermostatically controlled drying oven (50°C).
• Incubator/water bath.
• Benchtop centrifuge.
• Vortex mixer.
• Magnetic stirrer with temperature controller.
• Analytical balance.
• Single- and multichannel pipette and tips.
• Magnetic stirrer bars.
• Stopwatch.
• Measuring cylinders.
• Volumetric flasks.
• Filter funnels.
• 1.5 and 2 mL plastic microtubes with caps.
• 15/50 mL plastic tubes with screw caps.
• 100 mL autoclavable glass bottles with caps.
• Racks for 1.5–2 mL microtubes and 15/50 mL tubes.
• Disposable 96-well flat-bottom microplate.
• Aluminum foil.
15.3.3 Chemicals
15.3.3.1 Total phenolic content analysis of seaweed biomass
15.3.3.1.1 Folin–Ciocalteu assay
• FC reagent: 0.2 N in ultrapure water.
• Sodium bicarbonate (NaHCO3) solution: 6% w/v in ultrapure water.
• Absolute ethanol: 99% (w/w).
• PG stock solution: 2 mg mL−1 in absolute ethanol.
1. Measure the dry matter content of the dried, ground, and screened
seaweed (preparation described in Section 15.3.1) by using the
moisture content analyzer (following the instruction manual of the
analyzer you use). Record the moisture content of the sample p (%).
Calculate and record the dry matter content of the sample q (%):
q = 100% – p (15.1)
2. Mix the dried seaweed with 99% ethanol at a ratio of 1:20 (seaweed
biomass [g, dry weight]:ethanol [mL]) in, for example, a 100 mL glass
bottle with cap, under continuous stirring at 400 rpm at 22°C ± 1.0°C
242 Protocols for Macroalgae Research
for 24 h. Record the weight of the seaweed biomass m (mg) and total
volume V (L). Calculate and record the dry weight of seaweed bio-
mass M (mg):
M=m×q (15.2)
Y = kX + b (15.3)
Chapter fifteen: Total phenolic content and antioxidant capacity analysis 243
15. The concentration of the TPC in the seaweed extract sample, that is,
TPCSE (mg L−1) is calculated from the generated formula, based on its
measurement of A725 nm:
AE − A0
TPC SE = (15.4)
k
where:
TPCSE is the TPC in the extracted sample for measurement
AE is the absorbance of the seaweed extract sample
A0 is the absorbance of the blank
16. The TPC in seaweed biomass, that is, TPCSW (%, w/w) is
TPCSE × d × V
TPC SW(%) = (15.5)
M ×100
where:
d is the (possible) dilution factor recorded in step 6
V (L) and M (mg) are the extraction volume and seaweed bio-
mass dry weight recorded in step 2
1. Measure the dry matter content of the dried, ground, and screened
seaweed (preparation described in Section 15.3.1) by using the
moisture content analyzer (following the instruction manual of the
analyzer you use). Record the moisture content of the sample p (%).
Calculate and record the dry matter content of the sample q (%) fol-
lowing Equation 15.1.
2. Mix the dried seaweed with 99% ethanol at a ratio of 1:20 (seaweed
biomass [g, dry weight]:ethanol [mL]) in, for example, a 100 mL blue-
cap bottle, under continuous stirring at 400 rpm at 22°C ± 1.0°C for
24 h. Record the weight of the seaweed biomass m (mg) and total vol-
ume V (L). Calculate and record the dry weight of seaweed biomass
M (mg) following Equation 15.2.
3. Collect the liquid fraction (i.e., seaweed extract sample) after cen-
trifugation in a benchtop centrifuge at 2200 (×g) for 10 min.
4. Prepare other reagents and stock solutions as listed in Section
15.3.3.1.2.
5. Dilute the catechol stock solution with ethanol to prepare the stan-
dard solutions within the range of 0.6–15 mg L−1, for example, add
0.2, 0.5, 1.0, 2.0, and 5.0 mL of stock solution, and then add absolute
ethanol to the 50 mL mark of the volumetric flask.
244 Protocols for Macroalgae Research
6. If necessary, dilute the extracted sample with ethanol with the appro-
priate dilution factor to keep phenolic content in the diluted extract
sample within the linear range of concentrations of the standard
solution series, that is, 0.6–15 mg L−1. Record this dilution factor d.
7. Pipette 300 µL each of the below samples to one 2 mL microtube (and
make triplicate tubes per sample, as indicated):
• Diluted seaweed extract (×3)
• Blank (ethanol) (×3)
• Standard solution series: 0.6 mg L−1 (×3), 1.5 mg L−1 (×3), 3 mg L−1
(×3), 6 mg L−1 (×3), and 15 mg L−1 (×3).
Label the microtubes by, for example, seaweed extract, blank,
Catechol standard 0.6 mg L−1, Catechol standard 1.5 mg L−1, Catechol
standard 3 mg L−1, Catechol standard 6 mg L−1, and Catechol standard
15 mg L−1.
8. Pipette into each of the above microtubes 100 µL of 0.016 M K3Fe(CN)6
followed by 100 µL of 0.02 M FeCl3 in 0.1 M HCl.
9. Mix the microtubes properly by vortex mixing and incubate the
tubes in a water bath at 25°C for 15 min.
10. Pipette 300 µL 6.03 M H3PO4 into each of the above-mentioned tubes.
Vortex the tubes and incubate the tubes in a water bath at 25°C for
2 min.
11. Pipette 200 µL 1% gum arabic solutions to each of the microtubes and
mix it well by vortex mixing.
12. After that, pipette 200 µL of the reacted solution from each of the
labeled microtube to a well in the 96-well microtiter plate. Put the
plate to a UV–vis spectrophotometer.
13. Record the absorbance at 700 nm (A700 nm). Mean of A700 nm from the
triplicate of each sample (i.e., seaweed extract, blank, and catechol
standard series) is calculated and used for the following calculations.
14. Plot a catechol standard curve (X-axis: TPC [mg L−1], Y-axis: A700 nm).
A linear correlation formula is then generated from the curve, that is,
Y = aX + c (15.6)
15. The concentration of the TPC in the seaweed extract sample, that is,
TPCSE (mg L−1) is calculated from the generated formula, based on its
measurement of
TPC SE =
( AE − A0 ) (15.7)
a
where:
AE is the absorbance of the seaweed extract sample
A0 is the absorbance of the blank
Chapter fifteen: Total phenolic content and antioxidant capacity analysis 245
The TPC in seaweed biomass, that is, TPCSW (%, w/w) is then calcu-
lated as
TPCSE × d × V
TPC SW(%) = (15.8)
M ×100
where:
d is the dilution factor recorded in step 6
V (L) and M (mg) are the extraction volume and seaweed bio-
mass dry weight recorded in step 2
1. Measure the dry matter content of the dried, ground, and screened
seaweed biomass (preparation described in Section 15.3.1) by using
the moisture content analyzer (following the instruction manual of
the analyzer you use). Record the moisture content of the sample
p (%). Calculate and record the dry matter content of the sample q (%)
following Equation 15.1.
2. Mix the dried seaweed with 99% ethanol at a ratio of 1:20 (seaweed
biomass [g, dry weight]:ethanol [mL]) in, for example, a 100 mL glass
bottle with cap, under continuous stirring at 400 rpm at 22°C ± 1.0°C
for 24 h. Record the weight of the seaweed biomass m (mg) and
total volume V (L). Calculate and record the dry weight of seaweed
biomass M (mg) following Equation 15.2.
3. Collect the liquid fraction (i.e., seaweed extract sample) after cen-
trifugation in a benchtop centrifuge at 2200 (×g) for 10 min.
4. Prepare other regents and stock solutions as listed in Section 15.3.3.2.
5. Prepare AA standard: Mix the prepared AA standard with ethanol to
achieve the final concentration of AA standard series in concentra-
tion of 1 ~ 10 mg L−1: for example, add 1.0, 2.0, 5.0, 7.5, and 10 mL of
stock solution, and then add absolute ethanol to the 100 mL mark of
the volumetric flask.
6. Pipette 100 µL each of the below samples to a well in a 96-well
microtiter plate (and make triplicate tubes per sample, as indicated):
• Seaweed extract (×3)
• Sample control (ethanol) (×3)
• Standard solution series: 1 mg L−1 (×3), 2 mg L−1 (×3), 5 mg L−1 (×3),
7.5 mg L−1 (×3), and 10 mg L−1 (×3)
7. Pipette 100 µL of the prepared DPPH solution to the above-mentioned
wells.
246 Protocols for Macroalgae Research
( A − A0 )
I (%) = 1 − S × 100 (15.9)
AC
where:
AS is the absorbance in the seaweed extract sample
A0 is the absorbance in the blank for seaweed sample
AC is the absorbance in the sample control
OR
b. The AA equivalent antioxidant capacity (AEAC) by the following
way:
– Calculate the percentage inhibition (I%) in each of the AA
standard sample:
( A − A0 ST )
I ST (%) = 1 − ST × 100 (15.10)
AC
where:
AST is the absorbance of the standard sample
A0ST is the absorbance in the blank for the corresponding
standard sample
AC is the absorbance in the sample control
Y = gX + f (15.11)
Chapter fifteen: Total phenolic content and antioxidant capacity analysis 247
Acknowledgment
The current chapter is part of the MacroFuels project. The project has
received funding from the European Union’s Horizon 2020 research and
innovation program under grant agreement No 654010.
References
Amsler, C.D., and V.A. Fairhead. 2006. Defensive and sensory chemical ecology of
brown algae. Adv. Bot. Res. 43: 1–91.
Blois, M.S. 1958. Antioxidant determinations by the use of a stable free radical.
Nature 181: 1199–1200.
Chater, P.I., Wilcox, M.D., Houghton, D., and J.P. Pearson. 2015. The role of seaweed
bioactives in the control of digestion: Implications for obesity treatments.
Food Funct. 6: 3420–3427.
Connan, S., Delisle, F., Deslandes, E., and E.A. Gall. 2006. Intra-thallus phlorotan-
nin content and antioxidant activity in Phaeophyceae of temperate waters.
Bot. Mar. 49: 39–46.
248 Protocols for Macroalgae Research
Extraction of phycocyanin
and phycoerythrin pigments
Stewart William Beattie, Michèle Morançais,
Paul Déléris, Joël Fleurence, and Justine Dumay
Contents
16.1 Introduction ......................................................................................... 250
16.2 State of the art ...................................................................................... 251
16.3 Materials ............................................................................................... 252
16.3.1 Algal harvesting .................................................................... 252
16.3.2 Algal conditioning ................................................................ 253
16.3.3 Buffer preparation................................................................. 254
16.4 Experimental procedures................................................................... 254
16.4.1 Phycobiliproteins extraction................................................ 254
16.4.1.1 Algal treatment ..................................................... 254
16.4.1.2 Maceration ............................................................. 254
16.4.1.3 Acid extraction ...................................................... 255
16.4.1.4 Ultrasonic treatment ............................................ 255
16.4.1.5 Enzymatic hydrolysis .......................................... 255
16.4.2 Phycobiliprotein purification .............................................. 256
16.4.2.1 Adsorption bed chromatography ...................... 256
16.4.2.2 Ammonium sulphate precipitation ................... 257
16.4.2.3 Ion-exchange column........................................... 257
16.4.2.4 Hydroxyapatite chromatography....................... 257
16.4.2.5 Aqueous two-phase extraction........................... 258
16.4.3 Analytical method ................................................................ 258
16.5 Concluding remarks ........................................................................... 259
16.6 Notes ..................................................................................................... 260
References........................................................................................................ 262
249
250 Protocols for Macroalgae Research
16.1 Introduction
Phycobiliproteins (PBP) are an oligomeric water-soluble algal protein fam-
ily constituting the main light-harvesting chromoproteins of photosys-
tem II in Rhodophyta and Cyanobacteria. PBPs function as light-capture
and transfer systems, comprising up to 50% of water-soluble proteins in
Rhodophyta (Dumay et al. 2014) and 60% in Cyanobacteria (Cohen-Bazire
and Bryant 1982). These exist on the outer thylakoid membrane in the
stroma and are largely responsible for the reflected colors in Rhodophyta
(red algae), Cyanophycaea (blue–green algae), Cryptomonads and some
Pyrrophyceae, as aggregates of phycobilisomes (Cohen-Bazire and Bryant
1982, Isailovic et al. 2006). These PBPs function predominantly to absorb
sunlight in the wavelength range of 470–660 nm, allowing for the species
exploiting them to survive at very low light intensities, mainly at depths
in the water where chlorophyll-a becomes inefficient in light absorption.
PBPs are generally split into four groups according to their absorbance
maxima, existing as a complex of cores and rods (Figure 16.1). The quan-
tum efficiency with regards to energy transfer between PBPs has been
quoted as being close to 100%, with Phycoerythrin (PE) being located at
the tip of the rod, Phycocyanin (PC) in the center, and Allophycocyanin
(APC) comprising the core that then transfers energy to photosystem II
(Figure 16.2) (Galland-Irmouli et al. 2000).
Light (photons)
480–570 nm 550–650 nm
Light energy
transfer
6 nm
Stroma 12 nm
Thylakoid
membrane
Reaction center
chlorophyll
(chlorophyll a)
Lumen
Phycoerythrin/
Phycocyanin Allophycocyanin Chlorophyll a
Phycoerythrocyanin
Figure 16.2 Schematic of quantum energy transfer from the tip of a phycobilis.
CF1
Lipid bilayer
Allophycocyanin
Phycobilisome
Phycocyanin, phycoerythrin
This efficacy also depends on the number of steps taken to complete the
extraction as it is well known that a greater number of steps will result in
a lower final product yield (Kula et al. 1982).
The purity of the extracted PE and PC will ultimately determine which
use it is best suited for. Low purity (0.7) will be used as a food colorant as
it is nontoxic and noncarcinogenic (Doke 2005). High purity (3.9) can be
considered reactive grade, and purity (>3.2) will be implemented into pro-
cesses involving analyzing fluorescent energy transfer as PBPs have a nota-
bly large stokes shift, making then easily detectable through spectroscopy
(Rito-Palomares et al. 2001, Patil and Raghavaroa 2007, Dumay et al. 2015).
16.3 Materials
16.3.1 Algal harvesting
PBPs could be recovered from both seaweeds and microalgae.
For seaweeds (Note 1)
1. Collect the samples in the shore or from seaweed farms (Note 2).
2. Remove epiphytes when required.
3. Rinse thoroughly with tap water and spin dry the sample.
4. Store until use.
Chapter sixteen: Extraction of phycocyanin and phycoerythrin pigments 253
16.4.1.2 Maceration
This technique is arguably the most widely employed phycobiliprotein
extraction protocol, attributable to low economic impact, short extraction
time, and easiness of setup.
Chapter sixteen: Extraction of phycocyanin and phycoerythrin pigments 255
Silveira et al. (2007): The following equation can be used to calculate the
extraction yield of PBP:
EPP
Purification factor = (16.7)
EPC
where:
EPP is the Extract purity
EPC is the Extract purity of crude extract
Further information and more detailed protocols can be found using the
references cited for each process.
16.6 Notes
Note 1: PBPs content could significantly vary according to environmen-
tal factors. Be aware that samples collected during hot and sunny
season (it depends of the hemisphere) will not provide as high PBPs
yields as those collected during the cold season.
Note 2: Seaweed life cycle could also interfere with the extraction yield.
For more accurate results, separate thalli.
Note 3: Depending on the extraction method chosen (especially for
enzymatic assisted ones), you can use different buffers according to
pH required. Be sure that the buffer chosen fit with the extraction
method (enzyme accuracy as example).
Note 4: When PBPs extraction is compared, ensure that the buffer cho-
sen for all the methods is the same (same pH, same ionic strength,
and same composition). If not, discuss about the interferences led by
the buffer differences.
Note 5: Anionic polysaccharides present within the cell walls of sea-
weeds make extraction of metabolites challenging and rather inef-
ficient (Denis et al. 2009). The discharge of these highly valuable
and useful PBPs from cells can be directly related to cell rupture,
but small algae such as Spirulina sp. (300–500 µm diameter) are
resistant to this in the sense that they have multilayered cell walls,
Chapter sixteen: Extraction of phycocyanin and phycoerythrin pigments 261
References
Abalde, J., Betancourt, L., Torres, E., Cid, A., Barwell, C. 1998. Purification and char-
acterisation of phycocyanin from the marine cyanobacterium Synechococcus
sp. IO9201. Plant Science 136: 109–120.
Antelo, F. S., Anschau, A., Costa, J. A. V., Kalil, S. J. 2010. Extraction and puri-
fication of C-phycocyanin from Spirulina platensis in conventional and
integrated aqueous two-phase systems. Journal of the Brazilian Chemical
Society 21: 5.
Beer, S. and Eshel, A. 1985. Determining phycoerythrin and phycocyanin concen-
trations in aqueous crude extracts of red algae. Australian Journal of Marine
and Freshwater Research 36: 785–792.
Bennett, A. and Bogorad, L. 1973. Complimentary chromatic adaption in a fila-
mentous blue-green alga. The Journal of Cell Biology 58(2): 419.
Bermejo, R., Acién, F. G., Ibanez, M. J., Fernandez, J. M., Molina, E., Alvarez-Pez,
J. M. 2003. Preparative purification of B-phycoerythrin from the micro-
alga Porphyridium cruentum by expanded-bed adsorption chromatography.
Journal of Chromatography B 790(1–2): 317–325.
Bermejo, R., Felipe, M. A., Talavera, E. M., Alvarez-Pez, J. M. 2006. Expanded bed
adsorption chromatography for recovery of phycocyanins from the micro-
alga Spirulina platensis. Chromatographia 63(1–2): 59.
Bhaskar, S. U., Gopalaswamy, G., Raghu, R. 2005. A simple method for efficient
extraction and purification of C-phycocyanin from Spirulina platensis Geitler.
Indian Journal of Experimental Biology 43: 277–279.
Chaiklahan, R., Chirasuwan, N., Loha, V., Tia, S., Bunnag, B. 2011. Separation and
purification of phycocyanin from Spirulina sp. using a membrane process.
Bioresource Technology 102(14): 7159–7164.
Cohen-Bazire, G. and Bryant, D. A. 1982. Chapter 7. Phycobilisomes: Composition
and Structure. In: Carr, N. G. and Whitton, B. A. (Eds.). The Biology of
Cyanobacteria, pp. 143–190. Oxford, UK: Blackwell.
Dawes, C. 2016. Chapter 4. Macroalgae systematics. In: Levine, I. and Fleurence,
J. (Eds.). Seaweed in Health and Disease Prevention, pp. 107–138. London, UK:
E-Book, Elsevier.
Denis, C., Morançais, M., Gaudin, P., Fleurence, J. 2009. Effect of enzymatic diges-
tion on thallus degradation and extraction of hydro soluble compounds
from Grateloupia turuturu. Botanica Marina 52: 262–267.
Doke, J. M. Jr. 2005. An improved and efficient method for the extraction of phy-
cocyanin from Spirulina sp. International Journal of Food Engineering 1(5): 2.
Chapter sixteen: Extraction of phycocyanin and phycoerythrin pigments 263
Duarte, A. W. F., Lopes, A. M., Molino, J. V. D., Pessoa, A., Sette, L. D. 2015. Liquid-
liquid extraction of lipase produced by psychrotrophic yeast Leucosporidium
scottii L117 using aqueous two-phase systems. Separation and Purification
Technology 156(2): 215–225.
Dumay, J., Clément, N., Morançais, M., Fleurence, J. 2013. Optimisation of hydro-
lysis conditions of Palmaria palmata to enhance R-phycoerythrin extraction.
Biosource Technology 131: 21–27.
Dumay, J. and Morançais, M. 2016. Chapter 9. Proteins and pigments. In: Levine, I.
and Fleurence, J. (Eds.). Seaweed in Health and Disease Prevention, pp. 275–308.
London, UK: E-book, Elsevier.
Dumay, J., Morançais, M., Munier, M., Le Guillard, C. L., Fleurence, J. 2014.
Chapter 11. Phycoerythrins: Valuable proteinic pigments in red seaweeds.
In: Jacquot, J. P., Gadal, P., and Bourgougnon, N. (Eds.). Advances in Botanical
Research, Vol. 71, pp. 321–343.
Dumay, J., Morançais, M., Nguyen, H. P. T., Fleurence, J. 2015. Chapter 5. Extraction
and purification of R-phycoerythrin from marine red algae. In: Stengel, D. B.
and Connan, S. (Eds.). Natural Products from Marine Algae (NPMA). Methods
in Molecular Biology, p. 1308.
Engelmann, T. W. 1883. Bacterium photometricum. Archive for the Whole Physiology
of Humans and Animals 30(1): 95–124.
Fisheries and Aquaculture Statistics (FAO). 2014. FAO yearbook. Rome, Italy: Food
and Agriculture Organisation of the United Nations.
Fleurence, J. 2003. R-phycoerythrin from red macroalgae: Strategies for extrac-
tion and potential application in biotechnology. Applied Biotechnology, Food
Science and Policy 1(1): 1–6.
Fleurence, J., Massiani, L., Guyader, O., Mabeau, S. 1995. Use of enzymatic cell wall
degradation for improvement of protein extraction from Chondrus crispus,
Gracilaria verrucosa, and Palmaria palmata. Journal of Applied Phycology 7: 393.
Galland-Irmouli, A. V., Pons, L., Luçon, M. et al. 2000. One-step purification of
R-phycoerythrin from the red macroalga Palmaria palmata using pre-
parative polyacrylamide gel electrophoresis. Journal of Chromatography B:
Biomedical Sciences and Applications 739(1): 117–123.
Glazer, A. N. 1984. A Macromolecular complex optimised for light energy trans-
fer. Biochimica et Biophysica Acta 768: 29–51.
Glazer, A. N., Apell, G. S., Hixson, C. S., Bryant, D. A., Rimon, S., Brown, D. M.
1976. Biliproteins of cyanobacteria and rhodophyta: Homologous family of
photosynthetic accessory pigments. Proceedings of the National Academy of
Science 73: 428–431.
Glazer, A. N. and Hixson, C. S. 1975. Characterisation of R-phycocyanin.
Chromophore content of R-phycocyanin and C-phycoerythrin. Journal of
Biological Chemistry 250: 5487–5495.
Glazer, A. N. and Hixson, C. S. 1977. Subunit structure and chromophore composi-
tion of rhodophytan phycoerythrins. Porphyridium cruentum B-phycoerythrin
and b-phycoerythrin. Journal of Biological Chemistry 252(1): 32–42.
Goja, A. M., Tang, H., Cui, M., Li, C. 2013. Aqueous two-phase extraction advances
for bioseparation. Journal of Bioprocessing and Biotechnology 4: 1–8.
264 Protocols for Macroalgae Research
Munier, M., Morançais, M., Dumay, J., Jaouen, P. 2015. One-step purification of
R-phycoerythrin from the red edible seaweed Grateloupia turuturu. Journal of
Chromatography 992: 23–29.
Nguyen, H. P. T., Morançais, M., Fleurence, J., Dumay, J. 2016. Mastocarpus stella-
tus as a source of R-phycoerythrin: Optimisation of enzyme assisted extrac-
tion using response surface methodology. Journal of Applied Phycology 29(3):
1563–1570.
Niu, J. F., Wang, G. C., Lin, X. Z., Zhou, B. C. 2007. Large-scale recovery of
C-phycocyanin from Spirulina platensis using expanded bed adsorption
chromatography. Journal of Chromatography B 850: 267–276.
Niu, J. F., Wang, G. C., Tseng, C. K. 2006. Method for large-scale isolation and
purification of R-phycoerythrin from red alga Polysiphonia urceolata. Protein
Expression and Purification 49: 23–31.
Oliveira, E. G., Rosa, G. S., Moraes, M. A., Pinto, L. A. A. 2009. Characterisation
of thin layer drying of Spirulina platensis utilising perpendicular air flow.
Bioresource Technology 100: 1297–1303.
Patel, A., Mishra, S., Pawar, R., Ghosh, P. K. 2005. Purification and characterisation
of C-phycocyanin from cyanobacterial species of marine and freshwater
habitat. Protein Expression and Purification 40(2): 248–255.
Patil, G. and Raghavaroa, K. S. M. S. 2007. Aqueous two phase extraction for puri-
fication of C-phycocyanin. Biochemical Engineering Journal 34(2): 156–164.
Prabakaran, P. and Ravindran, A. D. 2013. Efficacy of different extraction methods
of phycocyanin from Spirulina platensis. International Journal of Research in
Pharmacy and Life Science 1(1): 15–20.
Ratanapongleka, R. 2010. Recovery of biological products in aqueous two phase
systems. International Journal of Chemical Engineering and Applications 1:
191–198.
Reise, A., Mendes, A., Lobo-Fernandes, L., Empis, J. A., Novais, J. M. 1998.
Production, extraction and purification of phycobiliproteins from Nostoc sp.
Bioresource Technology 66: 181–187.
Rito-Palomares, M. 2004. Practical application of aqueous two-phase partition
to process development for the recovery of biological products. Journal of
Chromatography B 807(1): 3–11.
Rito-Palomares, M., Nunez, L., Amador, D. 2001. Practical application of aque-
ous two-phase systems for the development of a prototype process for
C-phycocyanin recovery from Spirulina maxima. Journal of Chemical Technology
and Biotechnology 76: 1273–1280.
Sarada, R., Pillai, M. G., Ravishankar, G. A. 1999. Phycocyanin from Spirulina sp.
Influence of processing of biomass on phycocyanin yield, analysis of effi-
cacy of extraction methods and stability studies on phycocyanin. Process
Biochemistry 34: 795–801.
Silveira, S. T., Burkert, J. F. M., Costa, J. A. V., Burkert, C. A. V., Kalil, S. J. 2007.
Optimisation of phycocyanin extraction from Spirulina platensis using facto-
rial design. Bioresource Technology 98(8): 1629.
Su, C. H., Liu, C. S., Yang, P. C., Syu, K. S., Chiuh, C. C. 2014. Solid-liquid extraction
of phycocyanin from Spriulina platensis: Kinetic modelling of influential fac-
tors. Separation and Purification Technology 123: 64–68.
266 Protocols for Macroalgae Research
Sun, L., Wang, S., Gong, X., Zhao, M., Fu, X., Wang, L. 2009. Isolation, purifica-
tion and characteristics of R-phycoerythrin from a marine macroalga
Heterosiphonia japonica. Protein Expression and Purification 64: 146–154.
Wang, G. 2002. Isolation and purification of phycoerythrin from red alga Gracilaria
verrucosa by expanded-bed-adsorption and ion-exchange chromatography.
Chromatographia 56(7–8): 1–5.
Zhang, Y. M. and Chen, F. 1999. A simple method for efficient separation and
purification of C-phycocyanin and allophycocyanin from Spirulina platensis.
Biotechnology Techniques 13: 601–603.
Zhu, L. P., Gan, S. G., Lv, A. J. 2015. Chapter 51. Efficient purification and active
configuration investigation of R-phycocyanin from Polysiphonia urceolata.
In: Zhang, T. C. and Nakajima, M. (Eds.). Advances in Applied Biotechnology,
Vol. 332, pp. 489–496.
chapter seventeen
Contents
17.1 Introduction ......................................................................................... 268
17.2 State of the art ...................................................................................... 269
17.2.1 ROS detection reaction mechanism using
dihydroethidium and dichlorodihydrofluorescein
diacetate.................................................................................. 269
17.2.2 Histochemical localization mechanism using nitro
blue tetrazolium and DAB staining ................................... 271
17.3 Material ................................................................................................ 273
17.3.1 Reactive oxygen species visualization by microscopy .... 273
17.3.2 Reactive oxygen species quantification using
dihydroethidium and dichlorodihydrofluorescein
diacetate.................................................................................. 273
17.3.3 ROS histochemical localization .......................................... 273
17.4 Experimental procedures ...................................................................274
17.4.1 Reactive oxygen species visualization by microscopy .... 274
17.4.2 ROS quantification using DHE and DCFH–DA............... 274
17.4.3 Histochemical localization for reactive oxygen
species detection ....................................................................274
17.5 Tips and tricks ..................................................................................... 275
17.5.1 Reactive oxygen species visualization ............................... 275
17.5.2 Reactive oxygen species quantification ............................. 275
17.5.3 Reactive oxygen species histochemical localization ........ 276
Acknowledgment ........................................................................................... 276
References........................................................................................................ 276
267
268 Protocols for Macroalgae Research
17.1 Introduction
Marine macrophytes (including seaweeds and seagrasses) are the assem-
blage of diverse groups of phototrophic marine plants and are considered
marine ecological engineers. Both provide a range of ecologically and
economically valuable biological services; however, they are under con-
stant threat from a wide range of anthropogenic stressors, climate varia-
tion, invasive species, and pathogens and are often exposed to low- and
high-tidal cycle on daily bases (Contreras-Porcia et al. 2011; Kumar et al.
2011a, b). The relative abundance, survivability, and distribution of marine
macrophytes in such environments are principally determined by their
tolerance abilities to diverse stresses. Any adverse effect on marine mac-
rophytes because of fluctuating and stressful environmental conditions
can directly or indirectly lead to perturbations at higher trophic levels and
can eventually affect the integrity and sustainability of marine ecosys-
tems. The climatic variations and anthropogenic stressors in their ecologi-
cal niche trigger a series of physiological processes and accumulation of
reactive oxygen species (ROS), including singlet oxygen (1O2), superoxide
radicals (O2·−), hydrogen peroxide (H2O2), or hydroxyl radicals (OH·).
The cell organelles such as chloroplast, mitochondria, and peroxisomes
are vital organelles involved in highly oxidizing metabolic activities with
intense electron flow rate and are the major source of ROS production.
Furthermore, microsomes (e.g., cytochrome P450 and diamine oxidase),
peroxisomes (e.g., enzymes involved in fatty acid oxidation), and some
enzymes in the plasma membrane (e.g., NADPH oxidase and lipooxygenase)
have been identified as a potential source of ROS generator (Kumar et al.
2011b, 2014; Wojtala et al. 2014). ROS are produced directly by excitation
of O2 and subsequent formation of singlet oxygen, or by the transfer of
one, two, or three electrons to O2 or by protonation, which results in the
formation of O2·−, H2O2, or OH· radicals (Bischof and Rautenberger 2012).
Furthermore, O2·−, if protonated, can form the perhydroxyl radical (HO2·)
and can also react with reactive nitrogen species (RNS), that is, nitric
oxide radical (NO·, signaling free radical) to form more toxic peroxynitrite
(OON−) radical. Under physiological conditions, both ROS and RNS can
act as signal molecules to regulate cellular metabolism. They also affect
the transmission of signals across cells and can be considered second mes-
sengers during growth, differentiation, and cell death (Wojtala et al. 2014).
Although many ROS and RNS, while acting as a signal molecule, modify
gene expression and modulate the activity of specific defense proteins
(Kumar et al. 2014), high concentrations of ROS and/or RNS can disrupt
the cellular redox homeostasis, and can lead to oxidative burst exacer-
bating cellular damages. In this context, marine macrophytes express a
battery of antioxidant enzymes and nonenzymatic antioxidants such as
ascorbate, glutathione, tocopherol, carotenoids, phenolics, and flavonoids,
Chapter seventeen: Quantification and localization of reactive oxygen species 269
AcO Cl HO Cl O Cl O Cl
AcO Cl HO Cl HO Cl HO Cl
DCFH–DA DCFH DCFH DCF
DHE DCFH–DA
Figure 17.3 ROS burst visualized in Lessonia spicata under copper stress (100 µg L−1)
by fluorescence microscopy using DHE and DCFH–DA probes. (Adapted from
Contreras-Porcia, L. et al., Aquat. Toxicol., 94, 94–102, 2009.)
− −
N +Cl Cl N
N 4O2 + 2H+ NH HN
N N + N N N
N N
N N 4O2 N N
O O O O
NBT Formazan
(Soluble) (Insoluble–purple precipitate)
(a)
+ +
H2N NH2 HRP + H O NH NH HN NH
2 2
Figure 17.4 (a) NBT and (b) DAB oxidative reaction mechanism for ROS detection.
DAB
a b a b
NBT
O2 −
Seaweed Seagrass
(Ulva lactuca) (Zostera muelleri)
17.3 Material
17.3.1 Reactive oxygen species visualization by microscopy
• 0.2 µm-filtered natural seawater (NSW)
• 10 µM DCFH–DA (Calbiochem, UK) prepared in filtered seawater
• 100 µM DHE (Molecular Probes, Invitrogen, Australia) prepared in
filtered seawater
• Fluorescence microscopy coupled with camera
• Razor blade
• Glass slide
• Cover slips
• 50% glycerol
• Razor blade
• Glass slide
• Coverslips
• Stereomicroscope coupled with camera
• The DHE standard curve should be recorded along with the samples.
• Will be using the extinction coefficient of 2–OH–E+ for quantifica-
tion depending on the excitation wavelength.
• The excitation and emission wavelengths must be adjusted accord-
ing to the samples. Use additional controls as the tissue without
probes.
Acknowledgment
Author MK is grateful to the Australian Research Council for award-
ing him Discovery Early Career Research Award (DECRA Fellowship,
DE150100461-2015). Author LC-P is grateful to FONDECYT 1170881 and
DI-1245-16/R (Universidad Andrés Bello).
References
Bischof, K. and Rautenberger, R. 2012. Seaweed responses to environmental
stress: Reactive oxygen and antioxidative strategies. In C. Wiencke, and
K. Bischof (Eds.), Seaweed Biology. Novel Insights into Ecophysiology, Ecology
and Utilization Ecological Studies (Vol. 219, pp. 109–132). Heidelberg, Germany:
Springer.
Contreras-Porcia, L., Mella, D., Moenne, A., and Correa, J.A. 2009. Differential
responses to copper-induced oxidative stress in the marine macroalga
Lessonia nigrescens and Scytosiphon lomentaria (Phaeophyceae). Aquat. Toxicol.
94: 94–102.
Contreras-Porcia, L., Thomas, D., Flores, V., and Correa, J.A. 2011. Tolerance to
oxidative stress induced by desiccation in Porphyra columbina (Bangiales,
Rhodophyta). J. Exp. Bot. 62: 1815–1829.
Dikalov, S.I. and Harrison, D.G. 2014. Methods for detection of mitochondrial and
cellular reactive oxygen species. Antioxid. Redox Signal. 20: 372–382.
Chapter seventeen: Quantification and localization of reactive oxygen species 277
Gardner, P.R. 2002. Aconitase: Sensitive target and measure of superoxide. Meth.
Enzymol. 349: 9–23.
Kalyanaraman, B., Darley-Usmar, V., Davies, K.J., Dennery, P.A., Forman,
H.J., Grisham, M.B., Mann, G.E., Moore, K., Roberts, L.J., and Ischiropoulos,
H. 2012. Measuring reactive oxygen and nitrogen species with fluorescent
probes: challenges and limitations. Free Radic. Biol. Med. 52(1): 1–6.
Kumar, M., Bijo, A.J., Baghel, R.S., Reddy, C.R.K., and Jha, B. 2012. Selenium and
spermine alleviate cadmium induced toxicity in the red seaweed Gracilaria
dura by regulating antioxidants and DNA methylation. Plant Physiol.
Biochem. 51: 129–138.
Kumar, M., Gupta, V., Trivedi, N., Kumari, P., Bijo, A.J., Reddy, C.R.K., and Jha, B.
2011a. Desiccation induced oxidative stress and its biochemical responses in
intertidal red alga Gracilaria corticata (Gracilariales, Rhodophyta). Environ.
Exp. Bot. 72:194–201.
Kumar, M., Kumari, P., Reddy, C.R.K., and Jha, B. 2014. Salinity and desiccation
induced oxidative stress acclimation in seaweeds. In N. Bourgougnon (Ed.),
Sea Plants (Vol. 71, pp. 91–123). San Diego, CA: Academic Press Publisher,
Elsevier.
Kumar, M., Trivedi, N., Reddy, C.R.K., and Jha, B. 2011b. Toxic effects of imidazo-
lium ionic liquids on the green seaweed Ulva lactuca: Oxidative stress and
DNA damage. Chem. Res. Toxicol. 24(11): 1882–1890.
Steffen-Heins, A. and Steffens, B. 2015. EPR spectroscopy and its use in planta—
a promising technique to disentangle the origin of specific ROS. Front.
Environ. Sci. 3: 15.
Strittmatter, M., Grenville-Briggs, L.J., Breithut, L., Van-West, P., Gachon, C.M.M.,
and Küpper, F.C. 2016. Infection of the brown alga Ectocarpus siliculosus by
the oomycete Eurychasma dicksonii induces oxidative stress and halogen
metabolism. Plant Cell Environ. 39(2): 259–271.
Winterbourn, C.C. 2014. The challenges of using fluorescent probes to detect and
quantify specific reactive oxygen species in living cells. Biochim. Biophys.
Acta: General Subjects 1840(2): 730–738.
Wojtala, A., Bonora, M., Malinska, D., Pinton P., Duszynski, J., and Wieckowski, M.R.
2014. Methods to monitor ROS production by fluorescence microscopy and
fluorometry. Meth. Enzymol. 542: 243–262.
chapter eighteen
Contents
18.1 Introduction ......................................................................................... 280
18.2 State of the art ...................................................................................... 281
18.3 Materials............................................................................................... 283
18.3.1 Solvents................................................................................... 283
18.3.2 Equipment .............................................................................. 283
18.3.3 Consumables ......................................................................... 285
18.3.4 Solution recipes ..................................................................... 285
18.3.5 Instrumental setup ............................................................... 286
18.4 Experimental procedures .................................................................. 287
18.4.1 Sampling and metabolic quenching .................................. 288
18.4.1.1 Filtration of planktonic single-celled algae ...... 288
18.4.1.2 Collection of algal gametes (of Ulva spp.) ........ 288
18.4.1.3 Collection of algal thalli...................................... 289
18.4.2 Solid-phase extraction of extracellular metabolites ......... 289
18.4.3 Extraction of intracellular metabolites............................... 290
18.4.3.1 Cell disruption by ultrasound treatment ......... 290
18.4.3.2 Cell disruption with a bead mill ....................... 290
18.4.4 Two-step-derivatization for GC–MS analysis ................... 291
18.4.5 GC–MS analysis .................................................................... 292
18.4.6 Data analysis for GC–MS data ............................................ 292
18.5 Notes ..................................................................................................... 295
Acknowledgments ......................................................................................... 297
References........................................................................................................ 297
279
280 Protocols for Macroalgae Research
18.1 Introduction
Metabolomics techniques aim to comprehensively extract, quantify, and
evaluate metabolites from a given organism or community and have
developed into an indispensable tool in life sciences (Allwood et al.
2011; Aldridge and Rhee 2014) (for more information, refer to a recent
special issue in Current Opinion in Chemical Biology) (Schroeder and
Pohnert 2017). Different approaches have been brought forward that
allow us to answer a multitude of questions about the physiology of an
organism and to generate new hypotheses about its response to stress
(Goulitquer et al. 2012; Krug and Müller 2014). Among others, metabo-
lomics allows us to map changes in primary metabolism that reflect
the regulation of biochemical pathways to categorize samples by using
metabolic fingerprinting techniques or to identify metabolites that are
regulated in stress or interaction situations by comparative metabolic
profiling. Especially, comparative metabolomics is suitable for the
generation of hypotheses about the role of primary and secondary
metabolites (Lee and Fiehn 2008; Kuhlisch and Pohnert 2015). Most
commonly, mass spectrometric techniques are used for the recording
of metabolomics data. With complex samples, mass spectrometry
(MS) can be coupled to efficient separation techniques such as ultra
performance liquid chromatography (UPLC) and gas chromatography
(GC). By using such hyphenated analytical techniques, the profiling of
hundreds of metabolites within minutes is feasible. Fundamental pre-
requirements for successful metabolomics investigations are the exper-
iment planning, sample preparation, extraction, data recording, and
the statistical evaluation of the results. In this contribution, we describe
a very robust and validated protocol for the generation of metabolic
data from marine organisms. Initially developed for the investiga-
tion of the diatom Skeletonema marinoi, the protocol was successfully
adapted to the investigation of different algal taxa (Nylund et al. 2011;
Vidoudez and Pohnert 2012; Mausz and Pohnert 2015), including flag-
ellated gametes and thalli of the green macroalga Ulva mutabilis and
water samples for exometabolomic profiling (Alsufyani et al. 2017).
Even profiling of entire plankton communities is feasible without the
need for further alterations (Kuhlisch et al. submitted), and it can be
predicted that only minor adaptations to the protocol are required
to make it suitable for a broad range of other (marine) organisms. By
introducing a few additional experimental steps, the method that was
initially developed for the investigation of the endometabolome (i.e.,
the intracellular metabolites) can also be used to monitor the exome-
tabolome (i.e., the metabolites released into the surrounding seawater;
see Barofsky et al. 2009 and Alsufyani et al. 2017). For high-quality
Chapter eighteen: Metabolomics of intra- and extracellular metabolites 281
18.3 Materials
18.3.1 Solvents
• Acetone (certified ACS, Fisher Chemical)
• Chloroform (for HPLC, HiPerSolv CHROMANORM® [≥99.8%, fil-
tered through 0.2 µm, packed under N2, stabilized with 0.6% etha-
nol], VWR)
• Ethanol (gradient grade for LC, LiChrosolv®; ≥99.9%, filtered through
0.2 µm, Merck)
• Hexane (for GC, SupraSolv® [≥98%], Merck; stored over 4 Å molecu-
lar sieve)
• Methanol (for HPLC, Chromasolv®Plus [≥99.9%], Sigma-Aldrich)
• Pyridine (for HPLC, Chromasolv®Plus [≥99.9%], Sigma-Aldrich; stored
over 4 Å molecular sieve under argon)
• Tetrahydrofuran (THF; for HPLC, HiPerSolv CHROMANORM®
[≥99.7%, filtered through 0.2 µm, packed under N2, not stabilized],
VWR; stored under argon)
• Water (for HPLC, Chromasolv®Plus [filtered through 0.2 µm],
Sigma-Aldrich)
• Extraction solution (see Table 18.1 in Section 18.3.4)
• Column elution solution (see Table 18.2 in Section 18.3.4)
• Internal standard (IS) solution (see Table 18.3 in Section 18.3.4)
• Methoxyamine hydrochloride (98%, Sigma-Aldrich; hygroscopic
and thus stored in a desiccator under argon and vacuum-dried
before use)
• Retention time index (RI) solution (see Table 18.4 in Section 18.3.4)
• MSTFA (1 mL vials, Macherey–Nagel)
18.3.2 Equipment
• Fume hood
• Lab coat
• Safety goggles
• Chemical-resistant gloves (nitril or latex, dependent on solvent)
• Eppendorf® pipettes (1000, 100, and 10 µL, recently checked and
calibrated)
284 Protocols for Macroalgae Research
Top manifold
Nalgene® tubing
(a) (b)
Figure 18.1 Setup of filtration system (a) and filtrate extraction system (b).
Chapter eighteen: Metabolomics of intra- and extracellular metabolites 285
18.3.3 Consumables
• Eppendorf® pipette tips (blue, yellow, and white)
• Eppendorf® centrifuge tubes (1.5 mL, 2 mL)
• Whatman® glass microfiber filters (ø 47 mm, grade GF/C)
• SPE columns CHROMABOND® Easy (3 mL, 200 mg, Macherey–Nagel)
• Metal beads (ø 3 mm, stainless steel)
• Centrifuge tubes (15 mL)
• Glass screw neck vials both 1.5 and 4 mL, the 1.5 mL vials have to fit
GC–MS or LC–MS autosamplers
• Glass inserts for 1.5 mL vials (200 µL) with metal springs compatible
with autosamplers
• Screw caps for 1.5 and 4 mL vials with septa (PTFE coated)
• Liquid nitrogen
1) Research question
Control group, Treatment group(s), n = 5 for each
2) Experimental design Blank samples, n = 3
3.2.1
100 mg 3.2.2–3
Freeze in
liquid N2, Add 5 μL IS sol.,
Lyophilize, Add 1 mL 3.2.4–5 3.2.6 3.2.7
Homogenize extraction sol. 10 min ultrasonic bath Transfer Dry with
15 min at 30,000 g/4°C supernatant to desiccator
1.1.4–6 3.1.1 1.5 mL glass vial
Transfer aliquot
Add 1 mL to centrifuge tube
extraction sol.,
Add 5 μL IS sol. Vacuum
Vacuum
Vacuum
Figure 18.2 Metabolomics workflow for the analysis of intra- and extracellu-
lar metabolites of micro- and macroalgae by GC–MS. All numbers refer to steps
within the protocol.
288 Protocols for Macroalgae Research
10. Add 5 µL IS solution, close the vial, and vortex for 10 s.
11. Optional: Samples can be stored at −20°C for a few days and at −80°C
for several weeks.
12. Transfer for each sample an aliquot of 1.5 mL into a 1.5 mL glass vial.
For LC–MS see Note 4.
13. Evaporate to dryness under vacuum using a desiccator (Note 5).
Reduce the pressure from ambient pressure to 0 mbar considering
solvent boiling points (Note 6).
14. Vent the desiccator slowly with dry air or argon (Note 7) and imme-
diately close all vials.
15. Samples are ready for derivatization (see Section 18.4.4).
23. Centrifuge all samples at 8000 × g for 5 min using 15 mL centrifuge
tubes padded with a cloth as vial mounting. In the case of precipitate
formation, transfer the supernatant into a new glass insert.
24. Analyze the batch of samples immediately (Note 9) by GC–MS
(see Section 18.4.5.).
20
1000
750
500
PC 2 (15.8%)
10
250
0
m/z
−250
0 −500
−750
−1000
−10 0 5 10 15 20 25 30
Retention time (min)
−20 −10 0 10 20 30
PC 1 (50.7%)
(b) CAP DA—score plot (e) Heatmap plot
20 78 1
0.5
0
−0.5
10
−1
Component 2
599
0
−10
−20
−10 −5 0 5 10 15
Component 1
0.8
Correlation with component 2
0.6
78 599
0.4 1.0 1.0
Peak intensity
0.5 0.5
0.2 0 0
−0.5
0 −0.5
−1.0
−1.0
−0.2
A B A B
−0.4
−0.6
−1.5 −1.0 −0.5 0 0.5 1.0 1.5
Correlation with component 1
Figure 18.3 (Continued) All plots were generated from a dataset that was
obtained by GC–MS analysis of the macroalga Ulva mutabilis with either three
(a–c) or two (d–f) treatment groups (n = 4). Score plots of (a) principal coordinate
analysis and (b) CAP show the separation of sample groups. The vector plot of
the CAP (c) and the t-test-based cloud plot (d, from XCMS Online) indicate char-
acteristic metabolites. These selected metabolites can be visualized semiquan-
titatively with a heat map (e, from MetaboAnalyst) or box plots (f, from XCMS
Online).
(Figure 18.3b) for correct sample groups. Use the correlation values
of the original variables with the CAP axes to generate a vector plot
(Figure 18.3c) and select highly correlated variables. Describe these
variables by their m/z and tR values (see Section 18.4.6, step 4).
10. Compare the mass spectra of all highly correlated variables as
extracted by AMDIS (see Section 18.4.6, step 4) with spectral libraries
using MS Search 2.0 (nist, http://chemdata.nist.gov/, 2005). A com-
mon commercial database is the NIST library. Other, noncommercial
alternatives are available which often focus on specific metabolites,
for example, the Golm-library for plant metabolites. Document the
quality of spectral comparison with the R-match value.
11. Compare the retention times of all highly correlated variables with
externally measured reference compounds by calculating nonlinear
retention indices (van den Dool and Kratz 1963). Follow Fiehn et al.
(2008) for metabolomics standards initiative-compliant identification.
18.5 Notes
For a successful untargeted metabolomic approach, the personal secu-
rity and sample protection against contaminants and chemical reactions
with H2O and O2 are essential. It is thus recommended to wear personal
protective equipment and work under fume hoods whenever necessary.
Moreover, read the material safety data sheets (MSDS) of all reagents and
follow the recommendations for correct disposal. As biological samples
are rather complex, avoid any contamination by humans or lab equipment
(e.g., fatty acids, plasticizers). It is thus advisable to wear gloves, use glass
or PTFE whenever possible, and rinse lab equipment with solvents before
use. For the same reason, blanks should be carried through the whole pro-
cedure. As the derivatization is moisture sensitive, all involved chemicals
and samples have to be dry, which is especially challenging for salt water
samples. Samples should be exposed to air as rarely as possible. Several
metabolites are labile. Therefore, work should be carried out without lon-
ger interruptions.
Acknowledgments
We gratefully thank Alina Hera for her help in the Ulva mutabilis analysis.
Furthermore, we acknowledge Franziska Speck for testing protocol com-
prehensibility in the lab and Kathleen Thume for proofreading the manu-
script. The current work was supported by the Jena School of Microbial
Communication (CK, TW), by the Collaborative Research Center 1127
“Chemical Mediators in complex Biosystems” and CRC 1067 “AquaDiva”
(TW, GP) and by the European Union’s Horizon 2020 research and inno-
vation program under the Marie Sklodowska-Curie grant agreement No.
642575 (GC, TW, and GP).
References
Aldridge, B. B., and K. Y. Rhee. 2014. Microbial metabolomics: Innovation, applica-
tion, insight. Curr. Opin. Microbiol. 19:90–96.
Allen, A. E., J. Laroche, U. Maheswari et al. 2008. Whole-cell response of the pen-
nate diatom Phaeodactylum tricornutum to iron starvation. Proc. Nat. Acad. Sci.
105 (30):10438–10443.
Allwood, J. W., R. C. De Vos, A. Moing et al. 2011. Plant metabolomics and its
potential for systems biology research background concepts, technology,
and methodology. Methods Enzymol. 500:299–336.
Alsufyani, T., A. Weiss, and T. Wichard. 2017. Time course exo-metabolomic profiling
in the green marine macroalga Ulva (Chlorophyta) for identification of growth
phase-dependent biomarkers. Mar. Drugs 15 (1):14. doi: 10.3390/md15010014.
Álvarez-Sánchez, B., F. Priego-Capote, and M. D. Luque de Castro. 2010. Meta-
bolomics analysis II. Preparation of biological samples prior to detection.
TrAC, Trends Anal. Chem. 29 (2):120–127.
Anderson, M. J., and T. J. Willis. 2003. Canonical analysis of principal coordi-
nates: A useful method of constrained ordination for ecology. Ecology 84 (2):
511–525.
Barofsky, A., P. Simonelli, C. Vidoudez et al. 2010. Growth phase of the diatom
Skeletonema marinoi influences the metabolic profile of the cells and the selec-
tive feeding of the copepod Calanus spp. J. Plankton Res. 32 (3):263–272.
Barofsky, A., C. Vidoudez, and G. Pohnert. 2009. Metabolic profiling reveals
growth stage variability in diatom exudates. Limnol. Oceanogr.: Methods
7:382–390.
Becker, J., P. Berube, C. Follett et al. 2014. Closely related phytoplankton species
produce similar suites of dissolved organic matter. Front. Microbiol. 5 (111).
doi: 10.3389/fmicb.2014.00111.
Bieleski, R. L. 1982. Sugar alcohols. In Plant Carbohydrates I: Intracellular
Carbohydrates, (Eds.) F. A. Loewus and W. Tanner, pp. 158–192. Berlin,
Germany: Springer.
Bölling, C., and O. Fiehn. 2005. Metabolite profiling of Chlamydomonas reinhardtii
under nutrient deprivation. Plant Physiol. 139 (4):1995–2005.
Bromke, M. A., P. Giavalisco, L. Willmitzer, and H. Hesse. 2013. Metabolic analy-
sis of adaptation to short-term changes in culture conditions of the marine
diatom Thalassiosira pseudonana. PLoS One 8 (6):e67340.
298 Protocols for Macroalgae Research
Lee, D. Y., and O. Fiehn. 2008. High quality metabolomic data for Chlamydomonas
reinhardtii. Plant Methods 4. doi: 10.1186/1746-4811-4-7.
Lisec, J., N. Schauer, J. Kopka, L. Willmitzer, and A. R. Fernie. 2006. Gas chro-
matography mass spectrometry-based metabolite profiling in plants. Nat.
Protocols 1 (1):387–396.
Longnecker, K., M. C. K. Soule, and E. B. Kujawinski. 2015. Dissolved organic mat-
ter produced by Thalassiosira pseudonana. Mar. Chem. 168:114–123.
Mausz, M. A., and G. Pohnert. 2015. Phenotypic diversity of diploid and haploid
Emiliania huxleyi cells and of cells in different growth phases revealed by
comparative metabolomics. J. Plant Physiol. 172:137–148.
McCune, B., J. B. Grace, and D. L. Urban. 2002. Analysis of Ecological Communities.
2 ed., Vol. 28. Gleneden Beach, Oregon: MjM software design.
Menden-Deuer, S., and E. J. Lessard. 2000. Carbon to volume relationships for
dinoflagellates, diatoms, and other protist plankton. Limnol. Oceanogr.
45 (3):569–579.
Minor, E. C., M. M. Swenson, B. M. Mattson, and A. R. Oyler. 2014. Structural char-
acterization of dissolved organic matter: A review of current techniques for
isolation and analysis. Environ. Sci. Process Impacts 16 (9):2064–2079.
Nylund, G. M., F. Weinberger, M. Rempt, and G. Pohnert. 2011. Metabolomic
assessment of induced and activated chemical defence in the invasive red
alga Gracilaria vermiculophylla. PLoS One 6 (12):e29359.
Ritter, A., L. Cabioch, L. Brillet-Guéguen et al. 2017. Herbivore-induced chemical
and molecular responses of the kelps Laminaria digitata and Lessonia spicata.
PLoS One 12 (3):e0173315.
Ritter, A., S. M. Dittami, S. Goulitquer et al. 2014. Transcriptomic and metabolomic
analysis of copper stress acclimation in Ectocarpus siliculosus highlights sig-
naling and tolerance mechanisms in brown algae. BMC Plant Biol. 14 (1):116.
Roessner, U., C. Wagner, J. Kopka, R. N. Trethewey, and L. Willmitzer. 2000.
Technical advance: Simultaneous analysis of metabolites in potato tuber by
gas chromatography-mass spectrometry. Plant J. 23 (1):131–142.
Saccenti, E., H. C. J. Hoefsloot, A. K. Smilde, J. A. Westerhuis, and M. M. W. B.
Hendriks. 2014. Reflections on univariate and multivariate analysis of
metabolomics data. Metabolomics 10 (3):361–374.
Schroeder, F., and G. Pohnert. 2017. Editorial overview: Omics techniques to map
the chemistry of life. Curr. Opin. Chem. Biol. 36:v–vi.
Tautenhahn, R., G. J. Patti, D. Rinehart, and G. Siuzdak. 2012. XCMS Online:
A web-based platform to process untargeted metabolomic data. Anal. Chem.
84 (11):5035–5039.
van den Berg, R. A., H. C. Hoefsloot, J. A. Westerhuis, A. K. Smilde, and
M. J. van der Werf. 2006. Centering, scaling, and transformations: Improving
the biological information content of metabolomics data. BMC Genomics
7:142. doi: 10.1186/1471-2164-7-142.
van den Dool, H., and P. D. Kratz. 1963. A generalization of retention index system
including linear temperature programmed gas-liquid partition chromatog-
raphy. J. Chromatogr. 11 (4):463–471.
Vidoudez, C., and G. Pohnert. 2012. Comparative metabolomics of the diatom
Skeletonema marinoi in different growth phases. Metabolomics 8 (4):654–669.
Wagner, C., M. Sefkow, and J. Kopka. 2003. Construction and application of a
mass spectral and retention time index database generated from plant
GC-EI-TOF-MS metabolite profiles. Phytochemistry 62 (6):887–900.
300 Protocols for Macroalgae Research
Preparative extraction
of exometabolites from
seaweed surfaces
Florian Weinberger
Contents
19.1 Introduction ......................................................................................... 301
19.2 State of the art ...................................................................................... 302
19.3 Materials ............................................................................................... 304
19.4 Experimental procedures ................................................................... 304
19.4.1 Development of extraction protocol ................................... 304
19.4.2 Determination of algal surface/mass ratio........................ 305
19.4.3 Extraction ............................................................................... 306
19.5 Notes ..................................................................................................... 307
References........................................................................................................ 307
19.1 Introduction
The surface chemistry of seaweeds and other marine organisms is of
increasing interest in biotechnology and ecology. The molecular compo-
nents that are released, taken up, or perceived by an alga reflect all its
interactions with the biotic and abiotic environment. For this reason, these
compounds are often carriers of ecologically or physiologically important
functions. In particular, the relationships between algal hosts and com-
patible and noncompatible epibionts are often (if not always) driven by
molecular communication, and they typically involve chemical deterrents
or cues. Marine biotechnology increasingly targets such compounds for
antifouling applications. At the same time, ecologists studying seaweed–
epibiont interactions also aim to identify compounds that are bioactive
when they are present on macroalgal surfaces at physiological concentra-
tion. In particular, the second goal requires extraction methods that yield
surface metabolites and exclude tissue metabolites. Solid-phase extrac-
tion (SPE) is available to prepare relatively small samples for sensitive
301
302 Protocols for Macroalgae Research
Table 19.1 Exposure times to different organic solvents that are tolerated
before epidermal cells of seaweeds are disrupted
Species Solvent Time (s) Reference
(A) Rhodophyta
Laurencia obtusa Hexane 60 or less de Nys et al. (1998)
DCM, DE, EA, and <10 de Nys et al. (1998)
MeOH
Laurencia rigida 1% or less DCM in 30 Nylund et al. (2007)
hexane
1% DCM in hexane 30 or less Nylund et al. (2007)
Delisea pulchra Hexane 40 or less Nylund et al. (2007)
DCM, DE, EA, and <10 de Nys et al. (1998)
MeOH
Pterocladia capillacea Hexane 40 or less Nylund et al. (2007)
Solieria robusta Hexane 40 or less Nylund et al. (2007)
Gracilaria 25% DCM in hexane 5 or less Wang et al. (2016)
vermiculophylla
(B) Phaeophyta
Dictyopteris 4% or less DCM in 30 Nylund et al. (2007)
acrostichoides hexane
4% DCM 40 or less Nylund et al. (2007)
Dilophus marginatus 6% or less DCM in 30 Nylund et al. (2007)
hexane
6% DCM in hexane 40 or less Nylund et al. (2007)
Taonia atomaria MeOH 15 or less Othmani et al. (2016)
50% MeOH in DCM 20 or less Othmani et al. (2016)
Fucus vesiculosus 4% DCM in hexane 40 or less Brock et al. (2007)
50% MeOH in hexane 5 or less Lachnit et al. (2010)
50% MeOH in hexane 10 or less Saha et al. (2011)
Fucus serratus 50% MeOH in hexane 5 or less Rickert et al. (2016)
Note: DCM = Dichloromethane, DE = Diethyl ether, EA = Ethyl acetate, MeOH = Methanol.
19.3 Materials
• Algal samples to be extracted should be generally collected and
maintained under low-stress conditions. Desiccation, temperature
shock, or salinity shock have to be prevented for obvious reasons.
If different age cohorts, sexes, or populations are to be studied with
the final protocol (see 19.4.3), make sure that they are all investigated
during the preparatory steps (19.4.1 and 19.4.2), as they could differ
in their sensitivities.
• Sufficient amounts of cooled natural seawater to allow for repeated
rinsing and interim maintenance of sample seaweeds (Note 1).
• Salad spinner.
• Balance.
• Vent hood.
• Magnetic stirrer.
• Glass beakers (size adapted to the size of seaweeds to be extracted).
• Organic solvent.
• Natural seawater containing 0.05 % w/v of Evans Blue (20 mL per
sample).
• Rack with test tubes for incubation during staining (20 mL).
• Large- and medium-sized forceps.
• Stopwatch.
• Microscope with glass slides.
• Flatbed scanner.
• Imaging software (e.g., Fiji, a free software package available for
Windows, MacOS, Linux, and other systems [Schindelin et al. 2012]).
• Ruler.
• Filtration devise with glass microfiber filters.
• Evaporator.
• Lyophilizer.
5. Scanning: Dry the algae with paper, and spread them on a flatbed
scanner. Individuals that are too large or exhibit nonflat morphol-
ogies can be cut into pieces, which need to be distributed on the
scanner without overlap. A scale is placed on the scanner as well.
To obtain a good background contrast, cover the scanner with spot-
lessly clean and bright white material (e.g., an isopor panel). Scan at
high resolution and save the obtained image in a standard graphics
format.
6. Image analysis: Use imaging software to determine the total projec-
tion area of each alga.
7. Determine SA by using either Equation 19.1 or 19.4. Most seaweeds
exhibit either flat or filamentous morphologies. For thalli with flat
morphology, SA can be calculated from TP as
SA = 2 × TP (19.1)
and, because
SA = π × TP (19.4)
19.4.3 Extraction
All extractions should be carried out in a vented hood with clean equip-
ment. Prior to the extraction, examined algae need to be maintained at
low-stress conditions.
19.5 Notes
Note 1: To prevent stress, use seawater with a similar salinity and tem-
perature as in the collection site of the algal material.
Note 2: In most cases, nonpolar solvents such as hexane are tolerated
best (Table 19.1) and should be tested first. The solvent polarity may
then be increased stepwise until exposure times of less than 5 s
result in cell damage.
References
Brock, E., G. M. Nylund, and H. Pavia. 2007. Chemical inhibition of barnacle
larval settlement by the brown alga Fucus vesiculosus. Mar. Ecol. Progr. Ser.
337:165–174.
Cirri, E. and G. Pohnert. 2017. Disruption-free solid phase extraction of surface
metabolites from macroalgae. In Protocols for Macroalgae Research, B. Charrier,
T. Wichard, and C.R.K. Reddy, (Eds.) Chapter 20, 311–321. Boca Raton, FL:
CRC Press, Taylor & Francis Group.
de Nys, R., S. A. Dworjanyn, and P. D. Steinberg. 1998. A new method for deter-
mining surface concentrations of marine natural products on seaweeds.
Mar. Ecol. Progr. Ser. 162:79–87.
Lachnit, T., M. Wahl, and T. Harder. 2010. Isolated thallus-associated compounds
from the macroalga Fucus vesiculosus mediate bacterial surface colonization
in the field similar to that on the natural alga. Biofouling 26 (3):247–255.
Nylund, G. M., P. E. Gribben, R. de Nys, P. D. Steinberg, and H. Pavia. 2007. Surface
chemistry versus whole-cell extracts: antifouling tests with seaweed metab-
olites. Mar. Ecol. Progr. Ser. 329:73–84.
Othmani, A., J. F. Briand, M. Aye, M. Molmeret, and G. Culioli. 2016. Surface
metabolites of the brown alga Taonia atomaria have the ability to regulate
epibiosis. Biofouling 32 (7):801–813.
Rickert, E., M. Lenz, F. R. Barboza, S. N. Gorb, and M. Wahl. 2016. Seasonally fluc-
tuating chemical microfouling control in Fucus vesiculosus and Fucus serratus
from the Baltic Sea. Mar. Biol. 163 (10):203.
Saha, M., M. Rempt, B. Gebser, J. Grueneberg, G. Pohnert, and F. Weinberger.
2012. Dimethylsulphopropionate (DMSP) and proline from the surface of
the brown alga Fucus vesiculosus inhibit bacterial attachment. Biofouling
28 (6):593–604.
308 Protocols for Macroalgae Research
Disruption-free solid-phase
extraction of surface
metabolites from macroalgae
Emilio Cirri and Georg Pohnert
Contents
20.1 Introduction ......................................................................................... 309
20.2 State of the art .......................................................................................310
20.3 Materials................................................................................................311
20.3.1 Fucus vesiculosus .....................................................................311
20.3.2 Solid-phase surface extraction method ............................. 312
20.4 Experimental procedures .................................................................. 312
20.4.1 Extraction procedure ............................................................ 313
20.4.2 Data evaluation ..................................................................... 315
20.5 Notes ..................................................................................................... 315
Acknowledgments ......................................................................................... 317
References........................................................................................................ 317
20.1 Introduction
Surface metabolites play a fundamental role in the mediation of interac-
tions on biotic surfaces of, for example, macroalgae, corals, or sponges.
Such compounds control settling processes, regulate predator–prey rela-
tionships, and mediate infection processes (Wahl 2009, Dobretsov et al.
2013, da Gama et al. 2014). A hallmark of such interactions is the locally
much focused action of the compounds in question (Dworjanyn et al.
1999, 2006). The surface concentration of such metabolites often exceeds
the concentration in the medium, but it can even exceed concentrations
within the tissue as exuded compounds can accumulate in a diffusion-
limited laminar boundary layer. Despite their ecological importance,
309
310 Protocols for Macroalgae Research
until now, only a few methods allow the estimation of local concentra-
tions of surface metabolites. As a consequence, many investigations on
the effect of surface metabolites were based on bioassays with extracts of
whole organisms (Hellio et al. 2000, Nylund et al. 2011). Such experiments
do not reflect the real ecological relevance of surface-active substances
because only metabolites at the surface or near a producer should be con-
sidered (Nylund et al. 2007). The determination of metabolites within the
laminar boundary layer around an aquatic organism, a thin film of about
100–200 µm that determines the transition between the surface and the
surrounding water is thus crucial for experiment planning and evalua-
tion. Thus far, the laminar boundary layer has been studied to determine
uptake rates of nutrients (Wheeler 1980) or to model them (Hadley 2014),
or it has been investigated in correlation with climate changes and oceans’
acidification (Cornwall 2014), but the study of metabolites identity and
concentration at macroalgal surface is crucial both from an ecological and
an industrial point of view, especially in the framework of antifouling
substances research (Bhadury and Wright 2004, Rajan 2016).
5s 60 s 600 s
1.3
Control
1.2
Red/Green ratio
C18
Treatment
1.1 Control
C18
Hexan
Hexan/Methanol
1.0
20.3 Materials
20.3.1 Fucus vesiculosus
• Fucus vesiculosus samples can be collected independently of the sea-
son. The alga can be extracted directly after collection in the field or
can be kept in aquaria as described below.
312 Protocols for Macroalgae Research
1. 2.
3.
C18 - Material
4.
5.
Extract
Figure 20.2 Schematic workflow of the C18 method. (1) Algal fronds are removed
from the water and left for 2 min to remove excess water by dripping; (2) fronds
are transferred to Petri dishes and covered with absorption material; (3) the C18
material is washed off with excess seawater and collected in an empty solid-phase
extraction cartridge equipped with a frit; (4) the material is washed with deionized
water to remove salts; (5) elution with organic solvents finalizes sample preparation.
(Reprinted with permission from Cirri et al. 2016.)
6. Leave the alga in the Petri dish for 60 s (Note 5).
7. Take the alga out of the Petri dish using forceps, shaking it gently to
remove excess SPE material.
8. Hold the alga with forceps over the funnel connected with the empty
polypropylene column and with the Büchner flask (Figure 20.3).
9. Wash the algal surface with ASW from a spray bottle to remove the
most of SPE material. The SPE material is collected in an empty 6 mL
polypropylene column (Note 6).
10. Wash the funnel and the cartridge with the SPE material with excess
(ca. 20 mL) MilliQ water to remove salt from the seawater/medium,
by taking care that all SPE material is going into the cartridge. While
washing, you can apply a gentle vacuum (≈550 mbar) to make the
powder settle in the cartridge (0.5–1 mL bed volume) (Note 7).
11. Add an appropriate internal standard for recovery calculations.
12. Elute the compounds with methanol (three times 0.5 mL) into a 4 mL
glass vial under ambient pressure. Optimal flow rate should be
maximum one drop per second (Note 8).
13. Remove the solvent under a stream of nitrogen and redissolve the
sample in 100 µL of methanol (Notes 9 and 10). Transfer the sample
into a 250 µL glass insert placed in a 1.5 mL glass vial (or other vials
suitable for the autosampler of your own GC or HPLC).
Chapter twenty: Extraction of surface metabolites from macroalgae 315
14. Store the sample at −20°C or proceed to measure it with the desired
technique (GC–MS or LC–MS, as in, e.g., Cirri et al. 2016, Vidoudez
and Pohnert 2011) (Chapter 18 by Kuhlisch et al. 2018).
15. Place the extracted alga on a white, plane surface equipped with
a scale bar and take a picture. Calculate the surface of the alga as
shown in Chapter 19 by Weinberger (2017) with an image-processing
software (in our case, we used the open source software ImageJ,
http://imagejnihgov/ij/.) (Note 11). For thalli with flat morphology,
surface areas (SA) can be calculated as
SA = 2 × TP
SA = π × TP.
After the C18 material is completely dry, weight the cartridge to cal-
culate the amount of extraction powder used for the experiment.
20.5 Notes
Security advice: Care should be taken that the dust of the SPE material is
not inhaled as aerosols are problematic under prolonged exposure. MeOH
is flammable and toxic, thus handling under a fume hood or a good ven-
tilated area is advised.
316 Protocols for Macroalgae Research
Note 1: Take care of where you are cutting your alga: During the
following step of the method, you have to avoid washing and
removing the SPE material that is in contact with damaged tis-
sue in cutting sites. Otherwise, you would risk extracting internal
metabolites.
Note 2: Our method development was limited to test a different type of
C18 SPE material, but the flexibility of this approach allows extend-
ing the range of absorbing material that can be used. For example,
material contained in prepackaged SPE cartridges (HLB, StrataX,
and Ionic Exchanger) can be used as well. The choice depends on the
polarity of the metabolites under consideration. It is not necessary
to purchase new empty cartridges: You can use old used cartridges;
empty them and clean them before reusing them.
Note 3: The Petri dish dimensions can be changed as desired, and
smaller or bigger samples can be used, depending on the availability
of biological material and the concentration of surface metabolites.
The Petri dish method was preferred to other methods because it is
faster and assures a uniform and simultaneous covering of the algal
surface. A drawback of the method is the loss of a huge percentage
of SPE material as only about one-fifth of the powder spread in the
Petri dish attached to the algal surface. However, this powder can
be reused as it does not get contaminated by exometabolites of the
alga. An alternative to the Petri dish method could be to use a small
mesh sieve to spread the SPE material over the algal surface. Either
plastic or glass funnels can be used. Glass guarantees less chemi-
cal contaminations. However, the plastic material allows for stirring
and collecting the C18-powder easily, as the silica material tends to
stick to the glass surfaces.
Note 4: It is essential that the surface is not getting completely dry: The
thin layer of water that remains on the surface (laminar bound-
ary layer) is the environment from which metabolites shall be
determined.
Note 5: Different extraction times were tested, and 60 s was chosen as the
best compromise between a good interaction and absorption of metabo-
lites with C18 material and an easy recovery of the powder from the alga.
Note 6: The amount of water used for the washing steps is not a crucial
parameter, but for more precise absolute quantification of metabo-
lites, this step can also be easily standardized, always by using the
same amount of water.
Note 7: Different vacuum setups could be used, as a vacuum manifold
(Visiprep™, Supelco, USA) for multiple, simultaneous extractions.
However, the use of a vacuum pump is not necessary. In case the
vacuum is not applicable, the settlement of the SPE material will
Chapter twenty: Extraction of surface metabolites from macroalgae 317
require longer time and a larger amount of water to make sure that
the absorbing material is properly packed.
Note 8: Our method was optimized for 1.5 mL of methanol as an extrac-
tion solvent, but as in all SPE methods, the amount and the polarity
of the solvent (or mixture solvents) can be adapted. Other solvents
can be applied depending on the used solid phase and the nature
of the metabolites which should be extracted. C18 material does not
need to be dry before elution; however, each material has its specific
treatment to be fully effective. For all these information, also check
the recommendations of the manufacturer.
Note 9: If needed or desired, the sample can be stored at this point
at −20°C, and the drying step could be performed afterward.
Note 10: The volume of redissolution should be adapted to some surface
metabolites present and the sensitivity of the analytical instrument
to obtain a response in the linear range. For GC–MS analysis of polar
compounds, a derivatization protocol can be used starting from this
step. For more information about derivatization, see chapter 18 by
Kuhlisch et al. (2017) and Vidoudez and Pohnert (2011).
Note 11: The measurement of the surface is an essential step of this
method, but sometimes difficult to achieve, because of the texture
and morphology of the alga. Try to get your sample as flat as pos-
sible, by using, for example, a glass slide to cover your alga and some
weights to keep the alga plane.
Acknowledgments
The authors acknowledge funding the Jena School for Microbial
Communication and the International Max Planck Research School on
the Exploration of Ecological Interactions with Molecular and Chemical
Techniques and the EU Marie Sklodowska-Curie Initial Training Network
(ITN) program Algal Microbiome: Friends or Foes (ALFF) for funding.
References
Andras, T.D., Alexander, T.S., Gahlena, A., Parry, R.M., Fernandez, F.M., Kubanek, J.,
Wang, M.D., and Hay, M.E. 2012. Seaweed allelopathy against coral: Surface
distribution of a seaweed secondary metabolite by imaging mass spectrom-
etry. J. Chem. Ecol. 38:1203–1214.
Bhadury, P. and Wright, P.C. 2004. Exploitation of marine algae: Biogenic com-
pounds for potential antifouling applications. Planta 219 (4):561–578.
Cirri, E. 2014. Development and optimization of an extraction method for the
analysis of algal, surface metabolites, Master Thesis, University of Florence.
Cirri, E., Grosser, K., and Pohnert, G. 2016. A solid phase extraction based non-
disruptive sampling technique to investigate the surface chemistry of mac-
roalgae. Biofuling 32(2):145–153.
318 Protocols for Macroalgae Research
Cornwall, E.C., Boyd, P., McGraw, C.M., Hepburn, C.D., Pilditch, C.A., Morris, J.N.,
Smith, A.M., and Hurd, C.L. 2014. Diffusion boundary layers ameliorate the
negative effects of ocean acidification on the temperate coralline macroalga
Arthrocardia corymbosa. PLoS One 9 (9):e109468.
da Gama, B.A.P., Plouguerne, E., and Pereira, R.C. 2014. The antifouling defence
mechanisms of marine macroalgae. In: Advances in Botanical Research—Sea
Plants. Bourgounon, N., Jacquoud, J.P., and Gadal, P. (Eds.), pp. 413–440.
Amsterdam, the Netherlands: Elsevier.
de Nys, R., Dworjanyn, S.A., and Steinberg, P.D. 1998. A new method for deter-
mining surface concentrations of marine natural products on seaweeds.
Mar. Ecol. Prog. Ser. 162:79–87.
Dobretsov, S., Abed, R.M.M., and Teplitski, M. 2013. Mini-review: Inhibition of
biofouling by marine microorganisms. Biofouling 29:423–441.
Dworjanyn, S.A., de Nys, R., and Steinberg, P.D. 1999. Localisation and surface
quantification of secondary metabolites in the red alga Delisea pulchra. Mar.
Biol. 133:727–736.
Dworjanyn, S.A., Wright, J.T., Paul, N.A., de Nys, R., and Steinberg, P.D. 2006. Cost
of chemical defence in the red alga Delisea pulchra. Oikos 113:13–22.
Grosser, K., Zedler, L., Schmitt, M., Dietzek, B., Popp, J., and Pohnert, G. 2012.
Disruption-free imaging by Raman spectroscopy reveals a chemical sphere
with antifouling metabolites around macroalgae. Biofouling 28:687–696.
Hadley, S., Wild-Allen, K., Johnson, C., and Macleod, C. 2014. Modeling macroal-
gae growth and nutrient dynamics for integrated multi-trophic aquaculture.
J. Appl. Phycol., 27 (2):901–916.
Hall, S., Wild-Allen, K., Johnson, C., and Macleod, C. 2015. Modeling macroalgae
growth and nutrient dynamics for integrated multi-trophic aquaculture.
J. Appl. Phycol. 27 (2):901–916.
Hellio, C., Bremer, G., Pons, A.M., Le Gal, Y., and Bourgougnon, N. 2000. Inhibition
of the development of microorganisms (bacteria and fungi) by extracts of
marine algae from Brittany, France. Appl. Microbiol. Biotechnol. 54:543–549.
Kuhlisch, C., Califano, G., Wichard, T., and Pohnert, G. 2018. Metabolomics of intra-
and extracellular metabolites from micro- and macroalgae using GC-MS
and LC-MS. In Biotechnology Protocols for Macroalgae Research, Charrier, B.,
Wichard, T., Reddy, C.R.K. (Eds.), Chapter 18, 281–302. Boca Raton, FL: CRC
Press, Taylor & Francis Group.
Lachnit, T., Fischer, M., Kuenzel, S., Baines, J.F., and Harder, T. 2013. Compounds
associated with algal surfaces mediate epiphytic colonization of the marine
macroalga Fucus vesiculosus. FEMS Microbiol. Ecol. 84:411–420.
Lachnit, T., Wahl, M., and Harder, T. 2010. Isolated thallus-associated compounds
from the macroalga Fucus vesiculosus mediate bacterial surface colonization
in the field similar to that on the natural alga. Biofouling 26:247–255.
Nylund, G.M., Gribben, P.E., de Nys, R., Steinberg, P.D., and Pavia, H. 2007. Surface
chemistry versus whole-cell extracts: Antifouling tests with seaweed metab-
olites. Mar. Ecol. Prog. Ser. 329:73–84.
Nylund, G.M., Weinberger, F., Rempt, M., and Pohnert, G. 2011. Metabolomic
assessment of induced and activated chemical defence in the invasive red
alga Gracilaria vermiculophylla. PLoS One 6:e29359.
Chapter twenty: Extraction of surface metabolites from macroalgae 319
Rajan, R., Selvaraj, M., Palraj, S., and Subramanian, G. 2016. Studies on the anticor-
rosive & antifouling properties of the Gracilaria edulis extract incorporated
epoxy paint in the Gulf of Mannar Coast, Mandapam, India. Prog. Org. Coat,
90:448-454.
Rasband, W. 1994–2014. ImageJ, US National Institutes of Health, Bethesda, MD.
Available from: Accessed December 22, 2017, https://imagej.nih.gov/ij/
Saha, M., Rempt, M., Gebser, B., Grueneberg, J., Pohnert, G., and Weinberger, F.
2012. Dimethylsulphopropionate (DMSP) and proline from the surface of
the brown alga Fucus vesiculosus inhibit bacterial attachment. Biofouling
28:593–604.
Saha, M., Rempt, M., Grosser, K., Pohnert, G., and Weinberger, F. 2011. Surface-
associated fucoxanthin mediates settlement of bacterial epiphytes on the
rockweed Fucus vesiculosus. Biofouling 27:423–433.
Slaveykova, V.I., Guignard, C., Eybe, T., Migeon, H.-N., and Hoffmann, L. 2009.
Dynamic NanoSIMS ion imaging of unicellular freshwater algae exposed to
copper. Anal. Bioanal. Chem. 393:583–589.
Vidoudez, C. and Pohnert, G. 2011. Comparative metabolomics of the diatom
Skeletonema marinoi in different growth phases. Metabolomics 8(4):654–669.
Wahl, M., Molis, M., Ringelhan, F., Rohnstock, L., and Lenz, M. 2009. Effects of
grazing pressure on the induction of anti-herbivory defences in bladder-
wrack (Fucus vesiculosus) in different seasons. 8th International Temperate
Reefs Symposium. Accessed January 12, 2016. Adelaide, Australia.
Weinberger, F. 2018. Preparative extraction of exometabolites from seaweed
surfaces. In Biotechnology Protocols for Macroalgae Research, Charrier, B.,
Wichard, T., Reddy, C.R.K. (Eds.), Chapter 19, 303–310. Boca Raton, FL: CRC
Press, Taylor & Francis Group.
Wheeler, W.N. 1980. Effect of boundary layer transport on the fixation of carbon
by the giant kelp Macrocystis pyrifera. Mar. Bio. 56(2):103–110.
section three
Contents
21.1 Introduction ......................................................................................... 323
21.2 State of the art: Imaging cell walls in brown algae ........................ 324
21.3 Materials ............................................................................................... 325
21.4 Experimental procedures................................................................... 327
21.4.1 Preparation of algal material and fixation procedures .......327
21.4.2 Embedding protocol for London resins White resin ....... 328
21.4.3 Embedding protocol for Lowicryl resin ............................ 329
21.4.4 Sectioning of resin-embedded samples ............................. 330
21.4.5 Imaging algal cell walls using general stains ................... 330
21.4.6 Immunolabeling for fluorescence microscopy ................. 330
21.4.7 Immunolabeling for electron microscopy ......................... 331
21.4.8 Pretreatments before immunolabeling .............................. 331
21.5 Notes ..................................................................................................... 332
Acknowledgments ......................................................................................... 333
References........................................................................................................ 333
21.1 Introduction
Cell walls in brown algae are major cell compartments involved in impor-
tant biological roles such as algal growth and development, osmotic regu-
lation of cell surfaces, and defense responses. Such a variety of functional
properties is supported by dynamic and complex cell-wall architectures.
Brown algal cell walls are composed predominantly of polysaccharides
together with lower amounts of phenols, proteins, and halide compounds
(Deniaud-Bouët et al. 2014). The fucose-containing sulfated polysaccharides
and the alginates are the major sets of matrix polysaccharides, which are by
323
324 Protocols for Macroalgae Research
are interlinked. Equally important is the fact that the variations are not
just between lineages and species, but might also occur between organs,
tissues, and microdomains of the wall. Most of the chemical knowledge
gained on brown algal cell walls used analyses that require homogeniza-
tion of large amounts of material, generally including a diversity of cell
types. The information of each cell type is lost on analysis. In land plants,
the development of immunolocalization techniques has made significant
contributions to the exploration of many aspects of the biology of cell-wall
polysaccharides (Lee et al. 2011). These methods generally use well-defined
monoclonal antibodies that have been developed from immunization
procedures with cell-wall–derived materials. This technology has been
used for decades in plant biology and is now extending to brown algal cell
walls. Recently, specific monoclonal antibodies toward cell-wall polysac-
charides of brown algae were generated and characterized. They include
the brown algal monoclonal (BAM) antibodies that bind structural vari-
ants of FCSPs (BAM1–4) and alginates (BAM6–11) (Torode et al. 2015, 2016).
Although the structural features recognized by these antibodies have not
always been fully defined, they allow detailed cytological analysis to be
performed. They are likely to significantly increase our understanding of
the organization and developmental dynamics of the polysaccharides in
brown algal cell walls. In optical microscopy, these antibodies can be used
in combination to general stains, which are useful for the detection of all
cell walls in tissues.
Studies on plant cell-wall polysaccharides indicate that some epitopes
might not be readily accessible in all cell walls, because of other polymers
masking them (Marcus et al. 2008; Hervé et al. 2009; Xue et al. 2013). The
use of dedicated enzymes to remove populations of cell-wall polysac-
charides was instrumental in these study processes to reveal epitopes.
The situation is likely to be similar in brown algae and the use of specific
enzymatic treatments can be a valuable addition to the immunolabeling
techniques.
The current chapter discusses the use of specific antibodies in combi-
nation with histochemical techniques for the detection of cell-wall poly-
saccharides in tissues or at cell surfaces in brown algae. A set of protocols
is provided for both optical and electron microscopy. Additional guidance
is given for the general staining of all cell walls and the enzymatic pre-
treatment with alginate lyases to remove alginates.
21.3 Materials
• Seawater and fresh brown algae.
• Razor blades or Leitz 1320 freezing microtome (Ernst LeitzWetzlar
GmbH, Wetzlar, Germany) coupled with Tissue Freezing Medium®
326 Protocols for Macroalgae Research
100 μm
Figure 21.1 Micrographs showing toluidine blue O (TBO) staining of cell walls
and indirect immunofluorescence labeling of sulfated fucan and alginate epit-
opes in equivalent sections of Fucus serratus male conceptacles.
Chapter twenty one: In situ imaging of cell-wall polysaccharides 327
1. Fix samples.
2. Rinse the samples three times for 10 min with 0.5 M ammonium
chloride in 0.1 M phosphate buffer pH 7.4 to quench the free alde-
hyde groups.
Chapter twenty one: In situ imaging of cell-wall polysaccharides 329
3. Dehydrate for 10 min with 30% ethanol followed by 10 min with
50% ethanol, then 3 × 15 min with 70% ethanol, 3 × 15 min with
90% ethanol, and finally 3 × 15 min with absolute ethanol.
4. Infiltrate the specimens with a 3:1 mixture of ethanol and LR White
resin for 1 h.
5. Infiltrate the specimens with a 1:1 mixture of ethanol and LR White
resin for 1 h.
6. Infiltrate the specimens with a 1:3 mixture of ethanol and LR White
resin overnight.
7. Infiltrate the specimens with pure LR White resin. Repeat three
times every 3 h and incubate overnight.
9. Transfer the samples to gelatin capsules. Fill the capsules with 100%
LR White resin and place the caps. Make sure to exclude any oxygen
as this will inhibit polymerization.
10. Allow polymerization either at 37°C for five days (to minimize the
heat effect on a reduction of the antigenicity) or at 60°C for two days.
Alginates (LM7)
Detail
2 μm 1 μm
21.5 Notes
Note 1: Compared to plant or animal cells, cells from marine species are
hyperosmotic and particular care should therefore be taken to main-
tain the fixative in iso-osmolarity with those cells, that is, 1100 mOsm.
Note 2: The TBO staining method can be used as a postlabeling
procedure during which it can be very useful in quenching the auto-
fluorescence emitted by the algal cells.
Note 3: Controls are essential to validate the results—omitting the pri-
mary antibody is a good control to determine any nonspecific binding.
Note 4: Specific care should be taken to avoid photobleaching of the
fluorochromes. The labeling procedure is performed in darkness
and the light exposure under the microscope should be kept to a
minimum.
Chapter twenty one: In situ imaging of cell-wall polysaccharides 333
Note 5: The upper side of the grids (i.e., without sections) must be kept
clean and dry during all steps.
Note 6: Application of polysaccharide-degrading enzymes to materials
not fixed to a glass slide is likely to result in separation of cells and
may cause degradation of samples.
Acknowledgments
We acknowledge funding from the French National Research Agency
with regard to the IDEALG program with reference ANR-10-BTBR-04.
References
Acétarin, J.D., Carlemalm, E., and Villiger, W., 1986. Developments of new
Lowicryl resins for embedding biological specimen of even lower tempera-
tures. J. Micros. 143, 81.
Campa, C., Holtan, S., Nilsen, N., Bjerkan, T.M., Stokke, B.T., and Skjak-Braek, G.,
2004. Biochemical analysis of the processive mechanism for epimeriza-
tion of alginate by mannuronan C-5 epimerase AlgE4. Biochem. J. 381 (1),
155–164.
Carlemalm, E., Garavito, R.M., and Villiger, W., 1982. Resin development for elec-
tron microscopy and an analysis of embedding at low temperature. J. Micros.
126, 123.
Deniaud-Bouët, E., Kervarec, N., Michel, G., Tonon, T., Kloareg, B., and Hervé, C.,
2014. Chemical and enzymatic fractionation of cell walls from fucales:
Insights into the structure of the extracellular matrix of brown algae. Ann.
Bot. 114 (6), 1203–1216.
Hervé, C., Rogowski, A., Gilbert, H.J., and Knox, J.P., 2009. Enzymatic treatments
reveal differential capacities for xylan recognition and degradation in pri-
mary and secondary plant cell walls. Plant J. 58 (3), 413–422.
Hervé, C., Siméon, A., Jam, M., Cassin, A., Johnson, K.L., Salmeán, A.A., Willats,
W.G.T., Doblin, M.S., Bacic, A., and Kloareg, B., 2016. Arabinogalactan pro-
teins have deep roots in eukaryotes: Identification of genes and epitopes
in brown algae and their role in fucus serratus embryo development. New
Phytologist 209 (4), 1428–1441.
Kellenberg, E., Carlemalm, E., Villiger, W., Roth, J., and Garavito, R.M., 1980. Low
Denaturation Embedding for Electron Microscopy of Thin Sections. Chemische
Werke Lowi GmbH, Waldkraiburg, Germany, F.R.G, pp. 1–59.
Lee, K.J.D., Marcus, S.E., and Knox, J.P., 2011. Cell wall biology: Perspectives from
cell wall imaging. Mole. Plant 4 (2), 212–219.
Marcus, S.E., Verhertbruggen, Y., Hervé, C., Ordaz-Ortiz, J.J., Farkas, V., Pedersen,
H.L., Willats, W.G.T., and Knox, J.P., 2008. Pectichomogalacturonan
masks abundant sets of xyloglucan epitopes in plant cell walls. BMC Plant
Biol. 8, 60.
Torode, T.A., Marcus, S.E., Jam, M., Tonon, T., Blackburn, R.S., Hervé, C., and Knox,
J.P., 2015. Monoclonal antibodies directed to fucoidan preparations from
brown algae. PLoS One 10 (2), e0118366.
334 Protocols for Macroalgae Research
Torode, T.A., Siméon, A., Marcus, S.E., Jam, M., LeMoigne, A.M., Duffieux, D.,
Knox J.P., and Hervé, C., 2016. Dissection of cell wall assembly dynamics
during early embryogenesis in the brown alga Fucus using monoclonal
antibodies. J. Exp. Bot. 67 (21), 6089–6100.
Xue, J., Bosch, M., and Knox, J.P., 2013. Heterogeneity and glycan masking of cell
wall microstructures in the stems of Miscanthus × giganteus, and its parents
M. sinensis and M. sacchariflorus. PLoS One 8 (11), e82114.
chapter twenty two
Contents
22.1 Introduction ......................................................................................... 335
22.1.1 Biomechanical characterization of macroalgal
cell walls ............................................................................. 335
22.2 State of the art ...................................................................................... 336
22.2.1 Mechanical characterization of macroalgal cell walls ......336
22.3 Materials ............................................................................................... 338
22.4 Experimental procedures................................................................... 338
22.4.1 Preparation of algal material for atomic force
microscopy analysis.............................................................. 338
22.4.2 Choosing a force for indentation ........................................ 339
22.4.3 Influence of osmoticum upon macroalgal mechanics ..... 341
22.4.4 Selection of tip size ............................................................... 342
22.4.5 Performing atomic force microscopy based
elasticity measurements ....................................................... 343
22.5 Notes ..................................................................................................... 345
Acknowledgments ......................................................................................... 346
References........................................................................................................ 346
22.1 Introduction
22.1.1 Biomechanical characterization of macroalgal cell walls
The three independent multicellular macroalgae lineages (red, green, and
brown) show conserved adaptations to reducing drag, improving buoyancy,
and displaying remarkable developmental plasticity in their body forms.
As walled organisms, the structure, shape, and mechanical properties of
335
336 Protocols for Macroalgae Research
(a) (b)
(c) + Position −
22.3 Materials
• Seawater (artificial or natural)
• Fresh algae
• Razor blades/scalpel
• Forceps
• Petri dishes
• Microscope slides
• Gene Frame Seals (Thermo Scientific, 11570294)
• 1% (w/v) low-melt agarose in seawater
• AFM Cantilevers: 0.5 and 3 N m–1 stiffness; 10 nm, 1 and 10 µm tips
(Windsor Scientific, UK)
• Two-component epoxy resin
• Silica beads of various diameters (e.g., 5, 10, 50 µm; www.
microspheres-nanospheres.com)
• Atomic force microscope with top-view optics (JPK NanoWizard3,
JPK AG, Berlin)
3. Melt the agarose (1.5% [w/v] with seawater) and place ~250 µL (adjust
volume depending on sample number and size) onto the center of
the slide (within the Gene Frame), allow to partially set, and then
settle the excised algal piece on top of the agar (Note 1). Partial set-
ting allows the gel to have structure enough to hold the sample but
still enough pliability to form a good connection and fit the sample
in place.
4. Place the algae sample near to top of the agarose mound to ensure
relative flatness of the sample and minimize the chance of agarose
covering the sample.
5. Flood the reservoir with seawater.
6. Early stages of embryogenesis and sporophyte germination can be
studied without additional mounting support due to the natural
adhesive properties of the developing alga. To do this, settle fer-
tilized eggs/spores upon desired substratum (microscope slides)
that can be loaded into the AFM. When ready to start the AFM-
indentations, place a Gene Frame around the samples which forms a
reservoir of seawater to maintain hydration.
0.7
0.6
0.5
Depth (μm)
0.4
0.3
0.2
(a) 0.1
45
35
IM (MPa)
25
15
5
100 300 500 700 900 1100 1300 1500
(b) Force (nN)
Figure 22.2 Influence of indentation force and media upon indentation depth and
modulus in three macroalgal species. Fucus, Ulva, and Chondrus samples were
subjected to a set of force spectroscopy experiments at a range of forces in 1X
(solid) and 4X (dashed) artificial seawater (ASW). Resulting indentation depths
(a) allow the experimenter to determine an appropriate force for further experi-
ments and to examine the linearity of the material response. Resulting indenta-
tion (b) moduli (IM) allow the experimenter to assess the linearity of the material
response as well, which should be considered alongside knowledge of cell-wall
geometry. Examining sensitivity of both results to increased ASW concentration
allows for the consideration of sample sensitivity to evaporation of indentation
media during the experiment; Fucus is relatively insensitive, whereas Ulva is very
sensitive.
6. Compare the resultant indentation depths and IMs for the two con-
centrations of media.
7. Assess whether your sample is sensitive to changes in salinity. If it
is, we recommend that the solution be exchanged often during the
experiment. This may be achieved by making an initial waterline
mark, and topping up to this during the experiment with distilled
water. The changes seen here may be due to complex phenomenon:
turgor pressure may be affected (as in plants) resulting in a lower
measured IM. In the case of Ulva, in which the cell wall is over 20 µm
thick (Messyasz et al. 2013) and a maximum indentation of 0.5 µm is
applied, it is unlikely that turgor was influencing the measurements
in 1X seawater; the material properties of the wall, and its hydrogel
matrix, may change due to both changes in salt concentration and
dehydration. It is likely that this is the dominant phenomenon seen
in Figure 22.2 for Ulva and Chondrus.
40 20 40 20
IM (MPa)
(a) 0 0 0 0
100
90 Mean
80 Median
70
60
IM (MPa)
50
40
30
20
10
0
S. muticum; S. muticum; S. muticum; F. serratus; F. serratus; F. serratus;
10 nm 1 μm 10 μm 10 nm 1 μm 10 μm
(b) Species; tip diameter
Figure 22.3 The influence of AFM tip size on spatial resolution and indentation
modulus. Two species of brown alga, F. serratus and S. muticum, were tested using
three tip diameters but constant indentation force (10 nm, 1 µm, 10 µm; 500 nN).
Maps of indentation modulus (IM; 10 × 10 µm grids) are shown in (a). Smaller tip
sizes yield better XY resolution of surface structures and larger tip sizes give spa-
tially averaged values. (b) IM values vary with tip size and this may alter compa-
rability of datasets between experiments. The experimenter should be consistent
and clear on the tip size chosen.
grid with 8 × 8–128 × 128 points along that grid. The larger and
denser your grid, the longer the experiment but the better the reso-
lution. This decision requires some optimization by the user.
4. After utilizing the force you have experimentally determined, start
acquiring data in your force-map grid. Pay attention to the force–
position curves produced by the AFM during the run (most bio-
AFMs allow real-time observation of each curve as it is produced);
macroalgal samples can suffer from adhesion due to their coatings
of extracellular mucilage. When strong adhesion is present, as visu-
alized by a negative force at the start of the indentation curve and
end of the retraction curve (Figure 22.4), it is best to use a stiffness fit
for elasticity determination. When adhesion is only present on the
retraction curve, one can safely fit a Hertz-type model to the inden-
tation curve to obtain an IM. Figure 22.5 shows the topography of a
Fucus zygote rhizoid alongside the IM and stiffness maps obtained
through analysis.
5. Occasionally, some samples will appear very soft, indenting much
more than expected. In our experience, this results from one of two
Adhesion
Force
No adhesion
retract only
(a) (b)
Adhesion
indent and
retract
(c) Position
Figure 22.4 (a) The effect of sample adhesion on AFM-based indentation experi-
ments. In an ideal situation, there is no adhesion between the sample and the
tip. Neither the approach (dark gray) nor retraction (gray) curves result in values
that dip below the baseline noncontact force (dashed line). (b) Biological samples
often do show adhesion that may result in the tip being stuck on the sample dur-
ing retraction or (c) additionally being pulled down onto the surface early. When
adhesion is present, stiffness analysis is more appropriate.
Chapter twenty two: AFM analysis of macroalgae 345
30 2
Height (μm)
IM (MPa)
0 0
(a) (b)
Stiffness (N m−1)
(c)
scenarios: (1) either the sample has become unmounted in the aga-
rose or is covered in agarose, or (2) there is an inordinate amount of
mucilage on the sample. The latter can be helped by thoroughly rins-
ing your samples before mounting, the former requires remounting
of the sample.
6. When extracting data from AFM maps (i.e., for comparison of
regions), we recommend plotting values on the basis of height of
the sample and extracting the IM values as a secondary value. This
removes unconscious bias from selection of data. In Figure 22.5a, the
lowest part of the topographic map is the glass slide (also the high-
est IM and stiffness values), and this could be used as a height filter
threshold.
22.5 Notes
Note 1: When orientating samples for AFM analysis, consider the topo-
graphical structure of the algae in relation to angle and approach
of the cantilever. The cantilever requires access to the sample—
disrupting this access through ridges/obstacles will limit the capac-
ity to perform the experiment. Obtaining an unobstructed sampling
will require familiarization with the AFM you are using.
346 Protocols for Macroalgae Research
Acknowledgments
Funding for the work presented here was provided by the Gatsby
Charitable Foundation (GAT3396/PR4; T.A.T. and S.A.B). M.L. was sup-
ported by the R. Lewin and F.E. Fritsch Prize Studentship in Phycology.
J.L.K. was supported by the George and Lillian Schiff Studentship in
Engineering. We thank our colleagues in the Phycomorph Network for
their support and collaboration (COST Action FA1406; http://www.
phycomorph.org/).
References
Braybrook, S.A. 2015. Chapter 13—Measuring the elasticity of plant cells with
atomic force microscopy. Methods in Cell Biology 125: 237–254.
Braybrook, S.A., and Peaucelle, A. 2013. Mechano-chemical aspects of organ for-
mation in arabidopsis thaliana: The relationship between auxin and pectin.
PLoS One 8(3): e57813.
Fernandes, A.N., Chen, X.Y., Scotchford, C.A., Walker, J., Wells, D.M., Roberts, C.J.,
Everitt, N.M. 2012. Mechanical properties of epidermal cells of whole liv-
ing roots of Arabidopsis thaliana: An atomic force microscopy study. Physical
Review E 85: 1–8.
Messyasz, B., Czerwik-marcinkowska, J., Massalski, A., Uher, B., Rybak, A.,
Szendzina, L., Pikosz, M. 2013. Morphological and ultrastructural studies
on Ulva flexuosa subsp. Pilifera (Chlorophyta) from Poland. Acta Societatis
Botanicorum Poloniae 82: 157–163.
Milani, P., Gholamirad, M., Trass, J., Arnéodo, A., Boudaoud, A., Argoul, F.,
Hamant, O. 2011. In vivo analysis of local wall stiffness at the shoot apical
meristem in Arabidopsis using atomic force microscopy. The Plant Journal
67: 1116–1123.
Peaucelle, A., Braybrook, S.A., Le Guillou, L., Bron, E., Kuhlemeier, C., Höfte, H.
2011. Pectin-induced changes in cell wall mechanics underlie organ initia-
tion in Arabidopsis. Current Biology 21: 1720–1726.
Sampathkumar, A., Krupinski, P., Wightman, R., Milani, P., Berquand, A.,
Boudaoud, A., Hamant, O., Jönsson, H., Meyerowitz, E.M. 2014. Subcellular
and supracellular mechanical stress prescribes cytoskeleton behavior in
Arabidopsis cotyledon pavement cells. eLife 3: e01967.
Chapter twenty two: AFM analysis of macroalgae 347
Tesson, B., and Charrier, B. 2014. Brown algal morphogenesis: Atomic force micros-
copy as a tool to study the role of mechanical forces. Frontier in Plant Science
5: 1563.
Torode, T.A., Marcus, S.E., Jam, M., Tonon, T., Blackburn, R.S., Hervé, C., Knox, J.P.
2015. Monoclonal antibodies directed to fucoidan preparations from brown
algae. PLoS One 10(2): e0118366.
chapter twenty three
Contents
23.1 Introduction ......................................................................................... 350
23.2 State of the art ...................................................................................... 351
23.3 Materials ............................................................................................... 353
23.3.1 Reagents.................................................................................. 353
23.3.2 Ectocarpus culture .................................................................. 353
23.3.3 Equipment and software...................................................... 353
23.4 Experimental procedures................................................................... 354
23.4.1 Preparation of living algal material for time-lapse
microscopy ............................................................................. 354
23.4.1.1 Gametes release on glass-bottom Petri dish,
multiwell plate or free coverslips ...................... 354
23.4.1.2 Preparation of “homemade” glass bottom
Petri dishes............................................................ 354
23.4.2 Cell-surface deformation monitoring using
time-lapse microscopy ......................................................... 355
23.4.2.1 Prior preparation of the equipment .................. 356
23.4.2.2 Cell-surface labeling with fluorescent
microspheres ........................................................ 356
23.4.2.3 Measuring irreversible deformations
during growth (plastic strain) ............................ 356
23.4.2.4 Measuring deformations induced by
hypo- or hypertonic shock (mainly elastic
deformations) ....................................................... 357
23.4.3 Picture analysis: Quantification of cell-surface
marker displacement ............................................................ 359
23.5 Notes ..................................................................................................... 360
References........................................................................................................ 362
349
350 Protocols for Macroalgae Research
23.1 Introduction
Macroalgal cells are surrounded by a stiff cell wall that prevents any
relative cell displacement inside the multicellular thallus (Mirabet et al.
2011; Charrier et al. 2012). As a consequence, their development and
morphogenesis rely mainly on the pattern of cell division and cell elon-
gation, which impacts the proper final shape (Kuchen et al. 2012; Bassel
et al. 2014). Cell growth and morphogenesis imply mechanical deforma-
tions, which depends on molecular signaling pathways impacting the
mechanical properties of the cellular components and/or act through
force generation (Boudaoud 2010; Uyttewaal et al. 2010; Robinson et al.
2013). Before getting an integrated understanding of morphogenetic
mechanisms, geometrical deformations of cells and tissues must be char-
acterized and quantified (Mirabet et al. 2011). Moreover, characterization of
cell or tissue mechanical properties (i.e., deformability) is needed to assess
the physical and structural basis of deformations and to understand how
such deformations are regulated (Geitmann 2006; Routier-Kierzkowska
and Smith 2013). Deformations can be measured in response to external or
internal force variations (i.e., turgor) and allow one to get insight into the
possible biological mechanisms regulating the mechanical deformations
(Peters and Bernstein 1997). Deformation can be linked to the localization
of cellular factors such as cell-wall components, delivered vesicles, or ele-
ments of the cytoskeleton.
Many studies monitored the displacement of natural or artificial
surface markers regularly or randomly scattered at the cell surface dur-
ing growth and morphogenesis. Such studies were mainly conducted in
green land plants (Richards and Kavanagh 1943; Erickson and Sax 1956;
Hejnowicz and Brodzki 1960; Schnyder et al. 1987; Kutschera and Briggs
1988; Shaw et al. 2000; Menand et al. 2007; Armour et al. 2015; Yanagisawa
et al. 2015), green algae (Green 1965b; Chen 1973; Métraux et al. 1980;
Hejnowicz et al. 1977; Serikawa and Mandoli 1998; Von Dassow et al. 2001),
and fungi (Castle 1958; Soll and Herman 1983; Soll et al. 1985; Staebell
and Soll 1985; Bartnicki-Garcia et al. 2000; Abenza et al. 2015) but were
much less attempted on other groups of macroalgae (Waaland et al. 1972;
Kataoka 1982).
To our knowledge, no kinetic studies of cell or tissue growth pattern
were conducted in brown algae (Phaeophyceae), whereas this class of mac-
roalgae displays interesting specific features, among which are its great
morphological diversity and its phylogenetic position (Charrier et al.
2012; Bogaert et al. 2013). In the current chapter, we describe a protocol
to observe the fine pattern of cell-surface deformations during growth or
in response to turgor variations (osmotic shocks) in the sporophytic fila-
ments of Ectocarpus siliculosus, a model species for the brown algae. Cell
surface was marked with commercial fluorescent microspheres that get
Chapter twenty three: Dynamic and microscale mapping of cell growth 351
ionic, weak chemical interactions with the cell wall. First studies using
giant-celled characean algae such as Nitella or Acetabularia used anionic
resin particles (Green 1965a, b; Chen 1973; Hejnowicz et al. 1977;
Métraux et al. 1980; Kataoka 1982) or carbon particles (Waaland et al.
1972; Serikawa and Mandoli 1998; Von Dassow et al. 2001), whereas
Castle (1958) used starch grain on the tip-growing sporangiophore of
Phycomyces. In other eumycetes, polystyrenes beads (Soll et al. 1985;
Staebell and Soll 1985) or carbon particles (Bartnicki-Garcia et al. 2000)
were used. In all these studies, the displacements of surface mark-
ers were followed by time-lapse optical microscopy, sometimes with
special devices using horizontal objectives to observe cells growing
vertically (Castle 1958; Green 1965b; Chen 1973; Métraux et al. 1980).
Most recent studies on fungal or land plant tip-growing cells used fluo-
rescent surface markers such as commercial fluorescent microspheres
(Shaw et al. 2000; Dumais et al. 2004; Menand et al. 2007; Abenza et al.
2015; Yanagisawa et al. 2015) or droplets of fluorescent paint (Armour
et al. 2015), and deformation was monitored using epifluorescence or
confocal scanning microscopy.
After the time-lapse acquisition, images must be extracted, and
individual particle trajectories are mapped to quantify cell-surface
deformations. In the first studies, pictures were imprinted, and mor-
phometric measurements were done manually on imprinted images
(Castle 1958; Green 1965b; Kataoka 1982). Use of image analysis soft-
ware allowed more rapid image normalization (for size and growth
axis orientation) and analysis (Soll et al. 1985; Staebell and Soll 1985;
Bartnicki-Garcia et al. 2000; Shaw et al. 2000; Mine and Okuda 2003;
Menand et al. 2007; Abenza et al. 2015; Armour et al. 2015; Yanagisawa
et al. 2015).
Several methods for the calculation of the deformation were employed.
The simplest studies used surface-marking experiments only to locate
and measure the extent of the growth site in individual cells (Soll et al.
1985; Serikawa and Mandoli 1998; Menand et al. 2007). Bartnicki-Garcia
et al. (2000) finely traced the trajectory of markers on the growing tip and
compared them with theoretical trajectories predicted by various growth
models. However, most studies, especially those dedicated to tip-growing
cells, calculated the relative elemental growth rate as described in Peters
and Bernstein (1997). This approaches the irreversible and ideal extension
rate of an infinitely small elemental piece of cell wall in two perpendicu-
lar directions (and thus the growth anisotropy) as a function of its posi-
tion along the cells (generally relatively to a point of reference) (Castle
1958; Green 1965b; Chen 1973; Hejnowicz et al. 1977; Shaw et al. 2000; Von
Dassow et al. 2001; Dumais et al. 2004; Abenza et al. 2015). Some more
complex, computational approaches also exist (see, e.g., Armour et al.
2015) but are out of the scope of this chapter.
Chapter twenty three: Dynamic and microscale mapping of cell growth 353
23.3 Materials
23.3.1 Reagents
• Seawater.
• Ethanol 70% (v:v).
3. Under a sterile laminar flow hood, spray the Petri dishes (and their
lid) copiously with 70% ethanol and let them dry overnight under
the UV light of the hood.
4. The next day, keep the dishes under the laminar flow hood. Once
they are completely dry, cut 22 × 22 mm square pieces of double-
faced adhesive tape (i.e., of the size of a coverslip). Stick each piece
below the bottom of a drilled dish (Note 4), centering it on the hole of
the dish.
5. By using a fine sterilized scalpel or cutter, cut the central part of each
piece of the adhesive tape facing the hole of the dish.
6. Open the culture dishes bearing the inoculated coverslips. Remove
most of the filaments on the surface of the coverslip with fine for-
ceps, except a few at the very center.
7. Remove the coverslip from its Petri dish using forceps and a fine
needle. Gently dry the coverslips with a paper towel, except at the
center of the top face, where a few number of filaments must be kept
(Note 5). Be sure to let a small drop of seawater at this place to protect
those filaments from desiccation. This drop must have a diameter
lower than that of the central hole of the drilled dish.
8. Put the coverslip on the lid of a 50 mL Falcon tube. Then stick it
right below a drilled dish with a piece of double-faced adhesive tape.
Make sure that the square-shaped piece of double-faced adhesive
tape is right on the coverslip, and that the central filaments within
their drop of seawater end up in the hole of the dish. Press gently for
about 10 s on the internal face of the dish, around the central hole
(Note 6).
9. Immediately fill the dish with seawater and seal it with Parafilm®.
10. Paste the border of the coverslip (i.e., at the junction with the bottom
of the dish) with silicone glue using the tip of a pipettor cone (Note 7).
11. Place the dish under standard culture conditions for one night. Make
the dish straddle two inverted empty dishes or Falcon tube lids to
have the bottom face (with silicone glue) not in contact with the shelf
of the culture cabinet.
12. One day later, when the silicone glue is dry, the homemade glass-
bottom Petri dishes are ready to be used for time-lapse experiments.
c b a
d
e f g
c b a
d
(a) e f g
b c
a
e f
d
b c
a
e f
d
(b)
not selecting more than 4–5 positions, and no more than 1–2 focus
planes per position (Note 17).
4. Acquire the first series of pictures corresponding to the initial state
of the cells (t0 = normal osmolarity) (Note 18).
5. By using a pipette, take off most of the seawater from the dish.
6. Immediately after, fill the dish with a large volume (from 2 to 15 mL,
according to the volume available) of hypotonic or hypertonic sea-
water (Note 19). Then, correct each position that will probably be
modified in the z-axis because of the change of medium (Note 20).
7. After a short time (elastic stretching of Ectocarpus cell wall takes
about 1 min), acquire a second series of pictures at the same posi-
tions. This second series will correspond to the stable, inflated (after
hypotonic shocks), or shrunk (after hypertonic shocks) state of the
cells.
8. Export the pictures. Figure 23.1 shows a cell from an Ectocarpus fila-
ment, surface deformation of which was monitored after a hypotonic
shock in half-strength seawater.
1. For each time point and each position, export one bright-field pic-
ture of the median plane of the cell and one or several fluorescent
pictures of one or several planes of the cell showing fluospheres.
2. Process images using dedicated software such as GIMP (Smith and
Joost 2012; available at https://www.gimp.org). First, if time-lapse
facilities do not allow one to faithfully superpose successive images,
this should be done manually by using translations and/or rota-
tions, taking static markers outside the cell as landmarks. On each
image, draw a 1-pixel large line onto the cell wall, making the cell
contour. For convenience, this line should be drawn in color if the
microscope picture is black-and-white, and/or drawn on a separate
layer. Similarly, denote each marker by a pixel in another color.
3. From each cell contour, compute a cubic spline, thus creating one spline
per image. Smoothing should be carefully adjusted to avoid effects at
pixel-size scale, yet keeping local contour curvature. Similarly, build
a trajectory for each marker, by joining all marker pixels of the same
color in the time series, leading to one spline per marker. The degree
of each spline should be three, but if less than four-time steps are
available, then the degree should be n − 1 for n images.
360 Protocols for Macroalgae Research
23.5 Notes
Note 1: In most cases, we observed a bloom of bacteria invading the
whole glass bottom, rapidly killing and consuming all the early
Ectocarpus filaments.
Note 2: This step is dangerous and must be carried out in adequate
safety conditions. The metal perforator must be held at the extrem-
ity of a handle made of an isolating material (e.g., wood) to prevent
any burning injury.
Note 3: The hole must be much lower in size compared with the size of
the coverslip (22 × 22 mm).
Note 4: To efficiently stick the double-faced adhesive tape on the exte-
rior face of the dish bottom, turn the dish upside-down without its
lid, on the lid of a 50 mL Falcon tube (the top face of the Falcon lid
must be in contact with the internal face of the bottom of the dish).
Then press the adhesive tape on the exterior face of the dish bottom.
Note 5: Clean and dry the peripheral border of the coverslip to ensure a
good sticking of the coverslip under the double-faced adhesive tape
(cf the next step). Indeed, Ectocarpus and bacteria growing on the
surface of the coverslip seem to deposit some substances that make
the surface more hydrophilic. Cleaning the surface with a paper
towel will thus prevent the central drop of seawater from spreading
on the coverslip.
Note 6: You can press on the internal face of the Petri dish around the
central hole using a 50 mL Falcon tube without its lid and turned
upside-down.
Note 7: Be careful to keep the center of the coverslip clean of any trace
of glue, especially the area facing the central hole. This area must be
perfectly clean when high-magnification observations are planned.
Make sure that the silicone glue forms a thin bulge all around the
periphery of the coverslip, which does not diffuse to the center of
the coverslip. It would otherwise impede the movement of the thin
high-magnification objective.
Chapter twenty three: Dynamic and microscale mapping of cell growth 361
Note 20: The addition of several milliliters of medium in the dish will
increase the total weight on the object-holder and the stage. This can
lower all the positions by several µm and thereby require correction
of the z-position before the acquisition.
References
Abenza, J. F., E. Couturier, J. Dodgson, J. Dickmann, A. Chessel, J. Dumais,
and R. E. C. Salas. 2015. Wall mechanics and exocytosis define the shape
of growth domains in fission yeast. Nature Commun 6. Nature Publishing
Group: 8400. doi:10.1038/ncomms9400.
Armour, W. J., D. A. Barton, A. M. Law, and R. L. Overall. 2015. Differential
growth in periclinal and anticlinal walls during lobe formation in arabi-
dopsis cotyledon pavement cells. Plant Cell 27 (9): 2484–2500. doi:10.1105/
tpc.114.126664.
Bartnicki-Garcia, S., C. E. Bracker, G. Gierz, R. López-Franco, and H. Lu. 2000.
Mapping the growth of fungal hyphae: Orthogonal cell wall expansion dur-
ing tip growth and the role of turgor. Biophys J 79 (5): 2382–2390. doi:10.1016/
S0006-3495(00)76483-6.
Bassel, G. W., P. Stamm, G. Mosca, P. Barbier de Reuille, D. J. Gibbs, R. Winter,
A. Janka, M. J. Holdsworth, and R. S. Smith. 2014. Mechanical constraints
imposed by 3D cellular geometry and arrangement modulate growth pat-
terns in the arabidopsis embryo. Proc Natl Acad Sci USA 111 (23): 8685–8690.
doi:10.1073/pnas.1404616111.
Bogaert, K. A., A. Arun, S. M. Coelho, and O. De Clerck. 2013. Brown algae as a
model for plant organogenesis. In Plant Organogenesis: Methods and Protocols,
I. De Smet (Ed.), Vol. 959, pp. 97–125. New York: Springer Science+Business
Media. doi:10.1007/978-1-62703-221-6.
Boudaoud, A. 2010. An introduction to the mechanics of morphogenesis for plant
biologists. Trends Plant Sci 15 (6): 353–360. doi:10.1016/j.tplants.2010.04.002.
Castle, E. S. 1958. The topography of tip growth in a plant cell. J Gen Physiol 41:
377–383.
Charrier, B., A. Le Bail, B. de Reviers, A. L. Bail, and B. D. Reviers. 2012. Plant pro-
teus: Brown algal morphological plasticity and underlying developmental
mechanisms. Trends Plant Sci 17 (8): 468–477. doi:10.1016/j.tplants.2012.03.003.
Chen, J. C. W. 1973. The kinetics of tip growth in the nitella rhizoid. Plant Cell
Physiol 14 (4): 631–640.
Cox, R. W., and M. A. Peacock. 1978. The velocity field of growing ear cartilage.
J Anat 126 (Pt 3): 555.
Dumais, J., S. R. Long, and S. L. Shaw. 2004. The mechanics of surface expansion
anisotropy in Medicago Truncatula root hairs. Plant Physiol 136 (2): 3266–3275.
doi:10.1104/pp.104.043752.
Erickson, R. O., and K. B. Sax. 1956. Elemental growth rate of the primary root of
zea mays. Proc Natl Acad Sci USA 100 (5): 487–498.
Geitmann, A. 2006. Experimental approaches used to quantify physical parame-
ters at cellular and subcellular levels. Am J Bot 93 (10): 1380–1390. doi:10.3732/
ajb.93.10.1380.
Green, P. B. 1965a. Anion-exchange resin spheres as marking material for wet cell
surfaces. Exp Cell Res 40: 195–196.
Chapter twenty three: Dynamic and microscale mapping of cell growth 363
Contents
24.1 Introduction ......................................................................................... 365
24.2 State of the art ...................................................................................... 367
24.3 Materials ............................................................................................... 369
24.3.1 Chemicals ............................................................................... 369
24.3.2 Solutions and recipes ............................................................ 369
24.3.2.1 Preparation of Rhodamine–Phalloidin
(R415) or AlexaFluor568–Phalloidin
(A12380, life technology) ..................................... 369
24.3.2.2 Preparation of buffers .......................................... 370
24.3.2.3 Other solutions ..................................................... 372
24.3.3 Equipment .............................................................................. 373
24.4 Experimental procedures................................................................... 373
24.4.1 Preparation of poly-l-lysine-coated coverslips ................. 373
24.4.2 Preparation of Ectocarpus samples ...................................... 373
24.4.2.1 Filaments grown on coverslips .......................... 373
24.4.2.2 Free-floating filaments .........................................374
24.4.3 Actin staining procedure ......................................................374
24.5 Notes ..................................................................................................... 375
Acknowledgments ......................................................................................... 376
References........................................................................................................ 376
24.1 Introduction
Brown algae (Phaeophyceae) are a very interesting group of macroalgae
that acquired a complex multicellularity independently of land plants or
animals (Bogaert et al. 2013). In the past 10 years, genetic and molecular
365
366 Protocols for Macroalgae Research
24.3 Materials
24.3.1 Chemicals
• DMSO 100%.
• Glycerol 100%.
• Cell-wall digesting enzymes:
• Cellulase (Onozuka R-10, Yakult).
• Hemicellulase (H2125, Sigma).
• Driselase (D8037-1G, Sigma).
• Macerozyme (Onozuka R-10, Yakult).
• Pectinase (P2401, Sigma).
• Alginate lyase (A1603, Sigma).
• m-maleimido benzoic acid N-hydroxy succinimide ester (MBS,
M2786, Sigma).
• Natural or artificial seawater (ASW) (to wash free-floating filaments
before prefixation. Must be autoclaved and kept sterile, if possible).
• Paraformaldehyde (PFA, powder).
• p-phenylenediamine (PDA).
• Poly-l-lysine (1 mg mL−1).
• Rhodamine–Phalloidin (Sigma or Biotium) or AlexaFluor568–
Phalloidin (in general, AlexaFluor568 is more suited for plant mate-
rial than Rhodamine–Phalloidin).
• Triton X-100.
The instructions for the preparation of buffers and other solutions are
indicated in Tables 24.1 through 24.10.
24.3.3 Equipment
1.54 mL Eppendorf® tubes.
15- and 50 mL plastic tubes.
Fine forceps.
Glass coverslips (sterilized, if possible).
Microscope slides (sterilized, if possible).
Parafilm®.
Pipettes and tips (10/100/1000 µL range).
Plastic or glass Petri dishes.
Plastic Pasteur pipette.
(a) (b)
11. Stain the nucleus in 10 µg mL−1 Hoechst 33258 in PBS for 10 min, in
the dark at RT.
12. Wash 3 × 10 min in PBS, in the dark at RT.
13. Mount in 0.2% phenylenediamine in glycerol:PBS 2:1. The slides are
now ready to be observed under the fluorescent microscope. Two
examples of actin-staining pattern observed in Ectocarpus sporo-
phyte vegetative cells are shown in Figure 24.1.
24.5 Notes
Note 1: Alternatively, slides can be used instead of coverslips.
Note 2: By turning over the Petri dish on its lid, you create a moist cham-
ber in which the coverslips will be incubated. The moisture avoids
the evaporation of the medium deposited on the surface of the cov-
erslips, which is important especially for long incubation times. The
Parafilm® is hydrophobic and prevents the medium to flow over the
surface of the coverslips.
Note 3: Avoid sucking filaments into the pipette tip when removing the
medium from the tube while manipulating as gently as possible. If
needed, slowly centrifuge the tube to take the filaments at the bottom
of the tube. Avoid centrifuging filaments until they have been fixed.
376 Protocols for Macroalgae Research
Note 4: Do not pressure the filaments with the forceps, just spread them
around very gently, because Ectocarpus filaments are easily dam-
ageable. Do not leave large piles of entangled filaments, which pre-
vent proper staining of the filament, subsequently interfering with
the observation. If needed, spread the filaments under a dissecting
microscope using a fine needle.
Note 5: Incubation duration might require adjustments, depending on
the amount and nature of the treated material.
Note 6: The first washing step can be omitted. Alternatively, remove
half quantity of the first solution above and complete with the solu-
tion of PFA (8%) without washing step.
Note 7: Spread free-floating filaments on poly-l-lysine-coated coverslips
(or slides) as described in Section 24.4.1. The rest of the protocol is
identical for both types of sample.
Acknowledgments
This research was cofinanced by the National and Kapodistrian University
of Athens (program “Kapodistrias”), the Roscoff Biology Station (Centre
National de la Recherche Scientifique et Université Pierre et Marie Curie)
and the COST Action Phycomorph FA 1406.
References
Alessa, L., and D. L. Kropf. 1999. F-Actin marks the rhizoid pole in living Pelvetia
compressa zygotes. Development 126 (1): 201–209.
Barden, J. A., M. Miki, B. D. Hambly, and C. G. dos Remedios. 1987. Localization
of the phalloidin and nucleotide-binding sites on actin. Eur J Biochem 162 (3):
583–588.
Bisgrove, S. R., and D. L. Kropf. 2001. Cell wall deposition during morphogenesis
in fucoid algae. Planta 212 (5–6): 648–658.
Bisgrove, S. R., and D. L. Kropf. 2004. Cytokinesis in brown algae: Studies of asym-
metric division in fucoid zygotes. Protoplasma 223 (2–4): 163–173.
Bogaert, K. A., A. Arun, S. M. Coelho, and O. De Clerck. 2013. Brown algae as a
model for plant organogenesis. In Plant Organogenesis: Methods and Protocols,
I. De Smet (Ed.), 959: 97–125. New York: Springer Science+Business Media.
Braun, M., and G. O. Wasteneys. 1998. Distribution and dynamics of the cyto-
skeleton in graviresponding protonemata and rhizoids of characean algae:
Exclusion of microtubules and a convergence of actin filaments in the apex
suggest an actin-mediated gravitropism. Planta 205 (1): 39–50.
Brawley, S. H., and K. R. Robinson. 1985. Cytochalasin treatment disrupts the
endogenous currents associated with cell polarization in fucoid zygotes:
Studies of the role of F-actin in embryogenesis. J Cell Biol 100 (4): 1173–1184.
Charrier, B., S. M. Coelho, A. Le Bail, T. Tonon, G. Michel, P. Potin, B. Kloareg,
C. Boyen, A. F. Peters, and J. M. Cock. 2008. Development and physiology of
the brown alga Ectocarpus siliculosus: Two centuries of research. New Phytol
177 (2): 319–332.
Chapter twenty four: Actin staining in Ectocarpus 377
Chebli, Y., J. Kroeger, and A. Geitmann. 2013. Transport logistics in pollen tubes.
Molecular Plant 6 (4): 1037–1052.
Coelho, S. M., D. Scornet, S. Rousvoal, N. Peters, L. Dartevelle, A. F. Peters, and
J. M. Cock. 2012a. Immunostaining of Ectocarpus cells. Cold Spring Harb
Protoc 2012 (3): 369–372.
Coelho, S. M., D. Scornet, S. Rousvoal, N. Peters, L. Dartevelle, A. F. Peters, and
J. M. Cock. 2012b. Isolation and regeneration of protoplasts from Ectocarpus.
Cold Spring Harb Protoc 2012 (3): 361–364.
Deniaud-Bouët, E., N. Kervarec, G. Michel, T. Tonon, B. Kloareg, and C. Hervé.
2014. Chemical and enzymatic fractionation of cell walls from fucales:
Insights into the structure of the extracellular matrix of brown algae. Ann
Bot 114 (6): 1203–1216.
Doris, F. P., and M. W. Steer. 1996. Effects of fixatives and permeabilisation buffers
on pollen tubes: Implications for localisation of actin microfilaments using
phalloidin staining. Protoplasma 195: 25–36.
Dyachok, J., C.-M. Yoo, K. Palanichelvam, and E. B. Blancaflor. 2010. Sample prepa-
ration for fluorescence imaging of the cytoskeleton in fixed and living plants
roots. In Cytoskeleton - Methods and Protocols, R.H. Gavin (Ed.), 54: 157–169.
New York: Springer Science+Business Media New York.
Garbary, D. J., A. R. McDonald, and J. G. Duckett. 1992. Visualization of the cyto-
skeleton in red algae using fluorescent labelling. New Phytol 120: 435–444.
Garbary, D. J., and A. R. McDonald. 1996. Fluorescent labelling of the cytoskeleton
in Ceramium strictum (Rhodophyta). J Phycol 32 (1): 85–93.
Hable, W. E., and D. L. Kropf. 2005. The Arp2/3 complex nucleates actin arrays dur-
ing zygote polarity establishment and growth. Cell Motil Cytoskel 61 (1): 9–20.
Hable, W. E., N. R. Miller, and D. L. Kropf. 2003. Polarity establishment requires
dynamic actin in fucoid zygotes. Protoplasma 221 (3–4): 193–204.
Harper, J., D. McCurdy, M. Sanders, and J. Salisbury. 1992. Actin dynamics during
the cell cycle in Chlamydomonas reinhardtii. Cell Motil Cytoskel 126: 117–126.
Hawkins, E. K., J. J. Lee, and M. Correia. 2003. Polar localization of filamentous actin
in cells of the scale-forming alga Pleurochrysis sp. Protoplasma 220 (3–4): 233–236.
Heath, I. B., G. Gupta, and S. Bai. 2000. Plasma membrane-adjacent actin fila-
ments, but not microtubules, are essential for both polarization and hyphal
tip morphogenesis in Saprolegnia ferax and Neurospora crassa. Fungal Genet
Biol 30 (1): 45–62.
Hervé, C., A. Siméon, M. Jam, A. Cassin, K. L. Johnson, A. A. Salmeán, W. G. T.
Willats et al. 2016. Arabinogalactan proteins have deep roots in eukaryotes:
Identification of genes and epitopes in brown algae and their role in Fucus
serratus embryo development. New Phytol 209 (4): 1428–1441.
Heslop-Harrison, J., and Y. Heslop-Harrison. 1991. The actin cytoskeleton
in unfixed pollen tubes following microwave-accelerated DMSO-
permeabilisation and TRITC-phalloidin staining. Sex Plant Reprod 4: 6–11.
Kakimoto, T., and H. Shibaoka. 1987a. A new method for preservation of actin fila-
ments in higher plant cells. Plant Cell Physiol 28 (8): 1581–1585.
Kakimoto, T., and H. Shibaoka. 1987b. Actin filaments and microtubules in the pre-
prophase band and phragmoplast of tobacco cells. Protoplasma 140 (2–3): 151–156.
Karyophyllis, D., C. Katsaros, and B. Galatis. 2000. F-Actin Involvement in api-
cal cell morphogenesis of Sphacelaria rigidula (Phaeophyceae): Mutual align-
ment between cortical actin filaments and cellulose microfibrils. Eur J Phycol
35 (2): 195–203.
378 Protocols for Macroalgae Research
Contents
25.1 Introduction ......................................................................................... 381
25.2 State of the art ...................................................................................... 383
25.3 Materials ............................................................................................... 384
25.3.1 Reagents.................................................................................. 384
25.3.2 Equipment .............................................................................. 384
25.4 Experimental procedures................................................................... 384
25.4.1 Preparation of material......................................................... 384
25.4.2 Cryogen and substitution fixative ...................................... 385
25.4.3 Cryofixation ........................................................................... 386
25.4.4 Freeze substitution ................................................................ 386
25.4.5 Infiltration and embedding of samples into resin............ 386
25.5 Notes ..................................................................................................... 387
25.6 Concluding remarks ........................................................................... 387
References........................................................................................................ 388
25.1 Introduction
Chemical fixation involves the immersion of samples in a solution that
inactivates and stabilizes cellular contents. Although the general protocol
is the same, modifications are required to give optimal results (Huang
and Yeung 2015). The chemical fixation procedure is complicated, but
it has been a well-accepted method, giving good-quality images. The
main steps in chemical fixation are as follows: (a) fixation using aldehyde
fixatives (glutaraldehyde, paraformaldehyde) to cross-link proteins and
osmium tetroxide (OsO4) to preserve lipids in membranes and lipopro-
teins; (b) dehydration using an organic solvent (ethanol, acetone); and
(c) embedding samples in resins (Epon, Spurr, LR White, Lowcryl, etc.).
381
382 Protocols for Macroalgae Research
In most cases, the chemical fixation protocols used for land plants and ani-
mals are inadequate to apply to brown algae. Therefore, special modifica-
tions should be developed for this group (Clayton and Beaks 1983, Hallam
and Luff 1988). One of the characteristics of brown algae that should be
considered during preparation of transmission electron microscopy
(TEM) samples is that the cell wall contains alginates and fucoidans in
addition to cellulose (Kloareg and Quatrano 1988). These polysaccharides
flow out into the sample preparation buffers in conventional chemical
fixation methods. For example, alginate becomes soluble under low con-
centrations of calcium in the buffer. Swollen cell walls caused by low cal-
cium buffer induce detachment of the plasma membrane from the cell
wall. Therefore, phosphate buffer is inadequate for preparation of brown
algal cells, because calcium binds to phosphate. Thus, many researchers
use cacodylate buffer with 0.1%–1% calcium or seawater for the fixation
buffer. On the other hand, mucilaginous polysaccharides in the cell wall
of brown algae disturb penetration of fixatives and resins into the tissue,
and as a result, it is difficult to make ultrathin sections with satisfactory
preservation of the cell structures. Phenolic compounds in physodes,
which are membrane-bounded structures with osmiophilic or electron-
dense material, also cause a problem for chemical fixation in brown algae.
Leakage of the phenolics into the cytoplasm affects fixation of samples for
TEM, usually resulting in negative contrast images of the cell structure.
To trap the phenolics from physodes, caffeine is usually added to the alde-
hyde fixative and washing buffer (Clayton and Beaks 1983). Components
of the fixative and selection of the buffer become crucial factors affecting
TEM images of brown algae.
Physical fixation is achieved by cryofixation, composed of rapid
freezing (104 –105 K s−1) to the temperature of liquid nitrogen (−196°C)
or below and subsequent freeze substitution. All metabolic activities
in the cell are instantly stopped by cryofixation, which preserves the
native status of cells. However, cryofixation is not always successful in
all samples. In cryofixation, the most important thing is to prevent for-
mation of ice crystal in cells. One problem with cryofixation is that there
is a limitation in the area or depth of sample preserved, as it is difficult
to achieve high cooling to produce amorphous ice instead of ice crystals.
This depth-limit is usually a few micrometers. After cryofixation, fro-
zen cellular water should be substituted with an organic solvent under
low temperature (−180°C) (Gilkey and Staehelin 1986). There are three
methods in cryofixation: immersion, metal contact, and high-pressure
freezing. In immersion freezing, specimens are plunged into liquid
propane. This method is simple and handy, but freezing depth is less
than 10–20 µm. In the metal contact and high-pressure freezing meth-
ods, rapid freezing is performed by liquid helium or liquid nitrogen
using expensive instrumentation. The metal contact method permits a
Chapter twenty five: Cryofixation of brown algae for TEM 383
M
Ch
P P M
Ch
G
N
Ch
N N
Ch G
Es
C
P
M
(a) (b)
25.3 Materials
25.3.1 Reagents
Liquid nitrogen
Liquid propane
Acetone
Dry ice
2% Osmium tetraoxide (OsO4) in acetone
Spurr Low Viscosity Embedding Kit (Polysicences, Inc., Warrington,
PA, USA)
25.3.2 Equipment
Filter paper
Thin, acetone-resistant support film
Gold wire loop (5–10 mm diam.) with formvar film
Metal caps (appropriate for liquid propane and liquid nitrogen, for
example, small metal milk jug around 10 mL)
Polypropylene tube (50 mL)
Cryogenic tubes (2 mL)
Glass vial (2 or 4 mL)
Small styrene containers (appropriate for holding metal caps, polypro-
pylene tubes, and cryogenic vials)
(a) (b)
(c) (d)
Figure 25.2 Preparation for immersion freezing. (a) Sample carriers. A formvar
film on gold wire loops and a thin support film with acetone tolerance; (b) mak-
ing liquid propane in the fume hood. Place a hose connected to a propane gas
cylinder (right) into a precooled 50 mL polypropylene tube; (c) liquid propane and
liquid nitrogen poured into separate metal caps on the liquid nitrogen bath; and
(d) dry-ice acetone to keep cryogenic vials under –80°C.
been used as cryogens (Ryan 1992). However, ethane has not been used as
a cryogen for brown algae.
The substitution fixative is adjusted by adding OsO4 to acetone to a
final concentration of 2%. It is then transferred into a 2 mL cryogenic vial
and cooled in a liquid nitrogen bath.
25.4.3 Cryofixation
Excess culture medium is quickly drained with filter paper before immer-
sion into liquid propane, taking care to avoid plasmolysis. The specimen’s
carrier is held with forceps and rapidly plunged into the metal cap filled
with liquid propane. Quick immersion of the sample into liquid propane
is a key step for obtaining quality images in cryofixation, as it prevents
the growth of ice crystals. After 10 s, the sample is transferred into liquid
nitrogen and then preserved into precooled (<−80°C) 2% osmium–acetone
substitution fixative.
the resin is first diluted to 10% with acetone. The resin concentration is
then increased 10% every half day until it reaches 100%. A rotary shaker is
used to facilitate the infiltration of resin into cells. Samples in the vials are
exchanged with fresh 100% resin two times, and they are then infiltrated
with 100% resin overnight. New resin containing DMAE is added to the
samples and is kept for half a day. Finally, samples are embedded in the
resin on dishes of aluminum foil, and polymerization is carried out in an
oven at 60°C overnight.
In the case of film with adhering germlings, the resin pedestal must
be prepared beforehand with the same resin using aluminum-foil dishes.
The film is placed upside down with a little resin on the polymerized
resin pedestal. After polymerization, excess resin surrounding the film is
trimmed, the film removed with forceps, and the samples remain on the
flat resin disc (Note 3).
25.5 Notes
Note 1: Liquid propane is explosive when mixed with oxygen for a long
time, and, therefore, each of the above steps should be carried out in
a fume hood. After finishing these steps, liquid propane is immedi-
ately discarded outdoors. The filter paper is torn off by hand before
use to allow the promotion of absorption of water from the small
sample. Cool the cryogenic vials containing substitution medium
before use. Leave the cryogenic vials on liquid nitrogen until half of
the liquid freezes.
Note 2: Osmium tetroxide is highly hazardous. When handling osmium
tetroxide, wear gloves and carry out all steps in the fume hood. To
avoid contamination of freezers and refrigerators with OsO4, put the
cryogenic vials in a metallic can with a cover and seal it with plastic
tape.
Note 3: Vials containing samples immersed in more than 90% resin
should be put in a plastic container with silica gel to control humidity.
depresses the melting point of water and limits the formation of destruc-
tive ice crystals. This method preserves cell structures up to 600 µm in
depth. In brown algae, tissue from Fucus distichus and huge apical cells
from Halopteris congesta have been studied using this method (Nagasato
et al. 2015, Nagasato et al. 2017). High-quality ultrastructural images were
impossible to obtain using immersion-freezing methods. The disadvan-
tages of high-pressure freezing methods are that the instrument is expen-
sive, and a lot of liquid nitrogen is used. However, almost all samples were
well preserved and gave informative images using this technique.
Cryofixation is also more suitable for immunofluorescence and
immunoelectron microscopy than chemical fixation, because antigen-
recognition regions against the target molecules are preserved in cryofix-
ation but are not with chemical fixation (Guo and Huang 2015). In brown
algae, cryofixation has been applied using both procedures (Fu et al. 2014,
Fu et al. 2016). To obtain structures close to the native state of the cells,
cryofixation should be employed in cytological procedures.
References
Clayton, M. N., and Beakes, G. W. 1983. Effects of fixatives on the ultrastructure
of physodes in vegetative cells of Scytosiphon lomentaria (Scytosiphonaceae,
Phaeophyta). Journal of Phycology 19: 4–16.
Fu, G., Nagasato, C., Ito, T., Müller, D. G., and Motomura, T. 2013. Ultrastructural
analysis of flagellar development in plurilocular sporangia of Ectocarpus
siliculosus (Phaeophyceae). Protoplasma 250: 261–272.
Fu, G., Nagasato, C., Oka, S., Cock, J. M., and Motomura, T. 2014. Proteomics
analysis of heterogeneous flagella in brown algae (stramenopiles). Protist 165:
662–675.
Fu, G., Nagasato, C., Yamagishi, T., Kawai, H., Okuda, K., Takao, Y., Horiguchi, T.,
and Motomura, T. 2016. Ubiquitous distribution of helmchrome in phototac-
tic swarmers of the stramenopiles. Protoplasma 253: 929–941.
Gilkey, J. C., and Staehelin, L. A. 1986. Advances in ultrarapid freezing for the
preservation of cellular ultrastructure. Journal of Electron Microscopy Technique
3: 177–210.
Guo, F., and Huang, B. Q. 2015. Immunogold labeling for electron microscopy:
Strategy and problem solving. In Plant Microtechniques and Protocols,
E. C. T. Yeung, C. Stasolla, C. Sumner, and B. Q. Huang (Eds.), pp. 225–249.
Springer International Publishing, Switzerland.
Hallam, N. D., and Luff, S. E. 1988. The fixation and infiltration of larger brown
algae (Phaeophyta) for electron microscopy. British Phycological Journal 23:
337–346.
Huang, B. Q., and Yeung, E. C. 2015. Chemical and physical fixation of cells and
tissues: An overview. In Plant Microtechniques and Protocols, E. C. T. Yeung,
C. Stasolla, C. Sumner, and B. Q. Huang (Eds.), pp. 23–43. Springer International
Publishing, Switzerland.
Katsaros, C., Motomura, T., Nagasato, C., and Galatis, B. 2009. Diaphragm develop-
ment in cytokinetic vegetative cells of brown algae. Botanica Marina 52: 150–161.
Chapter twenty five: Cryofixation of brown algae for TEM 389
Kloareg, B., and Quatrano, R. S. 1988. Structure of the cell walls of marine algae
and ecophysiological functions of the matrix polysaccharides. Oceanogry
and Marine Biology, An Annual Review 26: 259–315.
McDonald, K. L., Morphew, M., Verkade, P., and Müller-Reichert, T. 2007. Recent
advances in high-pressure freezing: Equipment and specimen loading meth-
ods. In Methods in Molecular Biolology, Vol. 369, Electron Microscopy: Methods
and Protocols, 2nd Ed., J. Kuo (Ed.), pp. 143–173. Humana Press, Totowa, NJ.
Nagasato, C., Inoue, A., Mizuno, M., Kanazawa, K., Ojima, T., Okuda, K., and
Motomura, T. 2010. Membrane fusion process and assembly of cell wall dur-
ing cytokinesis in the brown alga, Silvetia babingtonii (Fucales, Phaeophyceae).
Planta 23: 287–298.
Nagasato, C., and Motomura, T. 2002. Ultrastructural study on mitosis and cytoki-
nesis in Scytosiphon lomentaria zygotes (Scytosiphonales, Phaeophyceae) by
freeze-substitution. Protoplasma 219: 140–149.
Nagasato, C., Tanaka, A., Ito, T., Katsaros, C., and Motomura, T. 2017. Intercellular
translocation of molecules via plasmodesmata in the multiseriate filamen-
tous brown alga, Halopteris congesta (Sphacelariales, Phaeophyceae). Journal
of Phycology 53: 333–341.
Nagasato, C., Terauchi, M., Tanaka, A., and Motomura, T. 2015. Development and
function of plasmodesmata in zygotes of Fucus distichus. Botanica Marina 58:
229–238.
Ryan, K. P. 1992. Cryofixation of tissues for electron microscopy: A review of
plunge cooling methods. Scanning Microscopy International 6: 715–743.
Schoenwaelder, M. E., and Clayton, M. N. 2000. Physode formation in embryos of
Phyllospora comosa and Hormosira banksii (Phaeophyceae). Phycologia 39: 1–9.
Seguí-Simarro, J. M. 2015. High-pressure freezing and freeze substitution of in vivo
and in vitro cultured plant samples. In Plant Microtechniques and Protocols,
E. C. T. Yeung, C. Stasolla, C. Sumner, and B. Q. Huang (Eds.), pp. 117–134.
Springer International Publishing, Switzerland.
Terauchi, M., Nagasato, C., Kajimura, N., Mineyuki, Y., Okuda, K., Katsaros, C., and
Motomura, T. 2012. Ultrastructural study of plasmodesmata in the brown
alga Dictyota dichotoma (Dictyotales, Phaeophyceae). Planta 236: 1013–1026.
chapter twenty six
Contents
26.1 Introduction ......................................................................................... 392
26.2 State of the art ...................................................................................... 393
26.2.1 General considerations ......................................................... 393
26.2.2 Protocol considerations ........................................................ 394
26.3 Materials ............................................................................................... 395
26.3.1 Fixation, dehydration, and embedding ............................. 395
26.3.2 Polymerization....................................................................... 397
26.3.3 Sectioning ............................................................................... 397
26.3.4 Uranyl acetate/Lead citrate staining .................................. 397
26.3.5 Immunogold labeling ........................................................... 398
26.4 Experimental procedures................................................................... 399
26.4.1 Fixation ................................................................................... 399
26.4.2 Dehydration ........................................................................... 399
26.4.3 Infiltration .............................................................................. 400
26.4.4 Polymerization....................................................................... 400
26.4.5 Sectioning ............................................................................... 401
26.4.6 Uranyl acetate/Lead citrate staining .................................. 403
26.4.7 Immunogold labeling ........................................................... 404
26.4.8 Considerations for light microscopy and correlative
studies ..................................................................................... 405
26.5 Notes ..................................................................................................... 407
References........................................................................................................ 409
391
392 Protocols for Macroalgae Research
26.1 Introduction
Macroscopic marine algae or seaweeds comprise a diverse range of mul-
ticellular photosynthetic eukaryotes that are classified as brown-, red-,
and green algae. Taxonomically, green seaweeds (Chlorophyta) belong
to the green lineage in the eukaryotic tree of life (Viridiplantae), and
are evolutionarily closer to land plants than other seaweed taxa (Becker
and Marin 2009, Leliaert et al. 2012); red seaweeds (Rhodophyta) are a
distinct and ancient eukaryotic algal lineage (Yoon et al. 2004), whereas
brown seaweeds (Phaeophyceae) form an independent lineage, which
is neither related to the other algal groups nor to land plants (Patterson
1999). Marine algae exhibit a wide array of morphological forms ranging
from giant kelps with distinctive thallus parts, such as holdfast, stipe, and
blade, to small filamentous or sheet-like phenotypes. Marine seaweeds are
important primary producers and some are keystone species in particular
habitats where they constitute the base of nutrient cycles and ultimately
define biotic diversity. Many seaweeds form extensive beds in intertidal
and subtidal zones of rocky shores, providing the physical infrastructure
for distinct ecosystems (Bold and Wynne 1978, Lobban and Harrison 1994,
Graham and Wilcox 2000). Although humans have historically used many
of these algae for food, fertilizers, and gelling agents, seaweeds are now
taking on even greater economic significance in the biofuel, pharmaceu-
tical, and bioremediation industries (Cardozo et al. 2007, Wargacki et al.
2012, Enquist-Newman et al. 2014, Cunha and Grenha 2016). Yet, the basic
biology of most seaweeds, including fundamental physiological processes
related to growth, development, and reproduction is poorly resolved and
lags significantly behind what is known about land plants. At the cell
biology level, though some seaweed species have been studied as model
organisms for specific biological phenomena, for example, adhesion in
Ulva sp. (Callow and Callow 2006) and polarity establishment in fertilized
eggs in Fucus sp. (Hervé et al. 2016, Torode et al. 2016), little is known about
the vast majority of taxa. Recently, new and valuable molecular studies
have emerged and are providing keen insight into the evolution, phyloge-
netic relationships, and physiological events of various marine algae (Cock
et al. 2010, Collén et al. 2013). Similar endeavors focused at the cellular level
will be required to help translate this wealth of new molecular data. The
technological foundation of many cell biology studies is microscopy. This
includes light microscopy (LM)-based imaging such as those employing
immunofluorescence labeling and fluorescent protein transformants, and
transmission electron microscopy (TEM)-based analyses. LM and TEM
imaging may also be coupled in correlative microscopy studies to provide
complementary data and different topological perspectives. For exam-
ple, in immunocytochemical investigations, immunofluorescence-based
imaging yields overview maps of specific epitope distribution in various
Chapter twenty six: Probing the subcellular topography of seaweeds 393
the fixation process. Third, the choice of the embedding chemical (e.g.,
plastic) is also critical, depending on the ultimate goal of the microscopy
study. Some plastics are polymerized with heat (e.g., epoxy plastics such
as Spurr’s Low-Viscosity Embedding Mixture) that may potentially cause
alterations to the antigens recognized by mAbs. Other agents may be
polymerized with UV at a wide range of temperatures. Many laboratories
employ LR White as it can be either UV-polymerized (room temperature
[RT], 4°C or −20°C) or heat polymerized (60°C). Other plastics can be UV
polymerized at very low temperature (e.g., −40°C or lower in the case of
Lowicryl® embedding media) but require special equipment and are often
a source of noxious vapors.
26.3 Materials
The following materials represent some of the resources required for pro-
cessing seaweeds for microscopic studies.
Aclar film
Mold
(d) (e) (f )
(g) (h)
26.3.2 Polymerization
• BEEM® embedding capsules, gelatin capsules, or molds for flat
embedding
• Aclar® 33c embedding film (if using LR White)
• Tweezers
• Laboratory UV lamp (for LR White medium)
• Laboratory oven (for Spurr’s medium)
26.3.3 Sectioning
• Ultramicrotome such as the Leica EM UC6 ultramicrotome
• Histo-diamond knife (e.g., DiATOME, Hatfield, PA, USA)
• Single-edge blades of carbon steel
• High precision and ultrafine tweezers
• Microscope slides
• Formvar-coated copper, nickel, or gold grids
• Chloroform
• Squeeze lab bottle with dH2O
• Glass pipette with a plug of cotton inside
• Filter paper ripped in pieces
• Eyelash tool: Use glue or nail polish to attach an eyelash or a fine hair
of an artist’s fine brush to a toothpick handle
• Grid storage box
• Disposable pipettes.
• Uranyl acetate 1% (w/v) solution.
• Lead citrate 1% (w/v) solution.
• High precision and ultrafine tweezers.
• 100 mL glass beakers.
• Glass Petri dish (100 mm diameter).
• Filter paper ripped in pieces.
• Silicone staining plates.
• Grid storage box.
26.4.2 Dehydration
Most embedding agents are hydrophobic. Therefore, water must be
removed from the specimen. Therefore, samples are slowly dehydrated
in a series of ethanol or acetone before adding the embedding medium.
Using a pipette to add or remove the solutions, samples are dehy-
drated in the graded ethanol or acetone series (10% [v/v], 30% [v/v], 50%
400 Protocols for Macroalgae Research
[v/v], 70% [v/v], 90% [v/v], and 100% [v/v]) during 30–60 min for each
grade, with gentle shaking, either at RT, at 4°C (ice bath or refrigerator) or
at −20°C (commercial freezer). For embedding media that may be polym-
erized with UV light (e.g., LR White), the ethanol dehydration should be
carried on at 4°C or −20°C; for epoxy plastics especially of osmicated spec-
imens, RT dehydration is satisfactory. The 70% (v/v) dehydrating stage is
an appropriate timing for an overnight incubation (Note 6).
26.4.3 Infiltration
After dehydration, the tissue segments are slowly infiltrated in a series of
embedding medium dissolved in the dehydrating agent (Note 7).
For LR White: Incubate the samples in a series of LR White:ethanol of
1:2, 1:1, 2:1, and 1:0. The first three infiltration steps should be 2–4 h long
each, at RT, 4°C or −20°C. The seaweed parts should be lightly shaken
during infiltration, placed in pure LR White at the desired temperature,
and lightly shaken for a period of up to two days.
For Spurr’s medium, prepare the resin beforehand in a plastic cup
according to the manufacturer’s instructions. Use a similar infiltration
series using acetone as the solvent. The final incubation with 100% Spurr’s
medium is performed overnight, or for one day, at RT.
26.4.4 Polymerization
A variety of specimen molds used for polymerization is commercially
available, such as plastic BEEM® capsules, gelatin capsules (Figure 26.1e),
and molds for flat embedding made of rubber, Teflon, or flexible plastic
(Figure 26.1f).
1. Seaweed parts may be trimmed with a razor blade to fit in the appro-
priate mold.
2. Add a small amount of the resin of choice (if using LR White and has
to be UV polymerized, an accelerator should be added in advance
following the manufacturer’s instructions). Using forceps, carefully
place a piece at the bottom of a BEEM® capsule or at the end of a
flat embedding mold in the desired orientation/position, and fill the
mold with resin. A dissecting light microscope is often helpful to
properly position the seaweed part in the mold.
3. For LR White polymerization, the BEEM® capsule is capped care-
fully. For flat embedding molds, a piece of Aclar® embedding film
should be cut and carefully placed over the molds, making sure that
there are no air bubbles between the resin and the film (Figure 26.1f)
(Note 8). Position a laboratory UV lamp 10–20 cm above the mold and
polymerize the sample for 24 h at the desired temperature (Note 9).
Chapter twenty six: Probing the subcellular topography of seaweeds 401
26.4.5 Sectioning
An ultramicrotome is the most convenient instrument to obtain sections
ranging in thickness from 500 nm for LM to 50–70 nm for TEM purposes.
Depending on the embedding medium chosen, the same block can be
conveniently used for correlative studies at the LM and TEM levels. Thick
sections may be immunolabeled and imaged with a fluorescence LM, and
labeled zones identified and documented. Thin sections are obtained
from selected zones and processed for immunogold labeling studies to
elucidate a more precise epitope location through electron microscopy.
Specific protocols include the following:
Chloroform pipette
(c)
Grid box
UA LC
NaOH
(d)
(e)
1 2 3 4 5
10 9 8 7 6
(f ) (g)
Figure 26.2 (a) Gold, nickel, or copper (first to third column, respectively) grids
are used to hold sections for TEM observations. (b) Useful tools for sectioning:
chloroform pipette, and eyelash attached to a toothpick. (c) Trimming the block
with a razor blade is the first and essential step for a successful sectioning.
(d) During the sectioning procedure, sections can be observed floating on the
water (arrow). (e) Apparatus used for uranyl acetate/lead citrate staining proce-
dure. A CO2-free chamber is essential to avoid the formation of lead carbonate
deposits. UA, Uranyl acetate drops; LC, Lead citrate chamber; NaOH, Sodium
hydroxide pellets. (f) 10 Multiwelled immunoslide is useful when performing
immunolabeling with different antibodies. (g) Microbiology loop used to pick
sections.
Chapter twenty six: Probing the subcellular topography of seaweeds 403
1. Keep the solutions in 15 mL Falcon™ tubes at 4°C for regular use,
and before using them make sure to centrifuge for 1 min at 1000 rpm
to precipitate unwanted particles.
2. Prepare a CO2-free chamber: Put a silicone plate in a glass Petri dish,
place sodium hydroxide pellets around it, put the number of drops
404 Protocols for Macroalgae Research
of lead citrate needed on the wells (one per grid), and close the cham-
ber with the lid (Figure 26.2e, Note 11).
3. Put a drop of uranyl acetate on a silicone plate, one drop for each
grid, and incubate for 5–10 min (section facing the stain).
4. Wash the grids with a gentle continuous stream of dH2O, dry with a
ripped filter paper piece.
5. Place each grid on a drop of lead citrate, close the chamber, and let it
incubate for 1–2 min.
6. Dip the grids in a beaker with the CO2-free dH2O, gently, for a few
seconds, wash again with a gentle continuous stream of dH2O, dry
with ripped filter paper, and store indefinitely in the grid box until
observation.
(a) (b)
(c) (d)
(e) Pd (f)
IS IS
CW
IS
CW
Figure 26.3 (a) Natural habitat of the brown seaweed Laminaria digitata. (b) Cross
section of L. digitata blade stained with toluidine blue. (c) Negative control where
no primary mAb is used. (d) Cross section of L. digitata blade immunolabeled
with LM7 mAb. LM7 is an excellent probe for alginate-containing extracellular
matrix in brown seaweed tissues. (e) Transmission electron micrograph of L. digitata
blade cross section immunogold labeled with LM7. (f) Detail of the gold particles
showing the labeling of the cell wall. CW, cell wall; IS, intracellular space;
Pd, plasmodesmata. Scale bar: (b–d) = 20 µm, (e) = 1 µm, (f) = 200 nm.
Chapter twenty six: Probing the subcellular topography of seaweeds 407
26.5 Notes
Variations and refinements in reagents, equipment, and protocols are
often necessary for the accommodation of the aforementioned protocols
in particular laboratories. The following list includes some common areas
where changes may be performed, and comments on details that should
be kept in mind while performing the experiments.
References
Becker, B., and B. Marin. 2009. Streptophyte algae and the origin of embryophytes.
Annals of Botany 103 (7):999–1004.
Bold, H. C., and M. J. Wynne. 1978. Introduction to the Algae: Structure and
Reproduction. Englewood, NJ: Prentice Hall.
Callow, J. A., and M. E. Callow. 2006. The Ulva spore adhesive system. In Biological
Adhesives, A. M. Smith and J. A. Callow (Eds.), pp. 63–78. Berlin, Germany:
Springer.
Cardozo, K. H. M., T. Guaratini, M. P. Barros, V. R. Falcão, A. P. Tonon, N. P. Lopes,
S. Campos et al. 2007. Metabolites from algae with economical impact.
Comparative Biochemistry and Physiology, Part C: Toxicology & Pharmacology
146 (1–2):60–78.
Cock, J. M., L. Sterck, P. Rouzé, D. Scornet, A. E. Allen, G. Amoutzias, V. Anthouard
et al. 2010. The Ectocarpus genome and the independent evolution of multi-
cellularity in brown algae. Nature 465 (7298):617–621.
Collén, J., B. Porcel, W. Carré, S. G. Ball, C. Chaparro, T. Tonon, T. Barbeyron
et al. 2013. Genome structure and metabolic features in the red sea-
weed Chondrus crispus shed light on evolution of the Archaeplastida.
Proceedings of the National Academy of Sciences of the United States of America
110 (13):1–6.
Cunha, L., and A. Grenha. 2016. Sulfated seaweed polysaccharides as multifunc-
tional materials in drug delivery applications. Marine Drugs 14 (3):42.
Enquist-Newman, M., A. M. Faust, D. D. Bravo, C. N. Santos, R. M. Raisner, A. Hanel,
P. Sarvabhowman et al. 2014. Efficient ethanol production from brown mac-
roalgae sugars by a specific yeast platform. Nature 505 (7482):239–243.
Graham, L. E., and L. W. Wilcox. 2000. Algae. New York: Prentice Hall.
Hallam, N. D., and S. E. Luff. 1988. The fixation and infiltration of larger brown
algae (Phaeophyta) for electron microscopy. British Phycological Journal
23 (4):337–346.
Hayat, M. A. 1981. Principles and Techniques of Electron Microscopy—Biological
Applications. 2nd ed. Vol. 1. Baltimore, MD: University Park Press.
Hervé, C., A. Siméon, M. Jam, A. Cassin, K. L. Johnson, A. A. Salmeán, W. G. T.
Willats, M. S. Doblin, A. Bacic, and B. Kloareg. 2016. Arabinogalactan pro-
teins have deep roots in eukaryotes: Identification of genes and epitopes
in brown algae and their role in Fucus serratus embryo development. New
Phytologist 209 (4):1428–1441.
Leliaert, F., D. R. Smith, H. Moreau, M. D. Herron, H. Verbruggen, C. F. Delwiche,
and O. De Clerck. 2012. Phylogeny and molecular evolution of the green
algae. Critical Reviews in Plant Sciences 31:1–46.
Lobban, C. S., and P. J. Harrison. 1994. Seaweed Ecology and Physiology. Cambridge,
UK: Cambridge University Press.
410 Protocols for Macroalgae Research
Login, G. R., and A. M. Dvorak. 1988. Microwave fixation provides excellent pres-
ervation of tissue, cells and antigens for light and electron microscopy. The
Histochemical Journal 20 (6):373–387.
Nagasato, C., A. Inoue, M. Mizuno, K. Kanazawa, T. Ojima, K. Okuda, and
T. Motomura. 2010. Membrane fusion process and assembly of cell wall dur-
ing cytokinesis in the brown alga, Silvetia babingtonii (Fucales, Phaeophyceae).
Planta 232 (2):287–298.
Nagasato, C., and T. Motomura. 2002. Ultrastructural study on mitosis and cytoki-
nesis in Scytosiphon lomentaria zygotes (Scytosiphonales, Phaeophyceae) by
freeze-substitution. Protoplasma 219 (3):140–149.
Nagasato, C., N. Kajimura, M. Terauchi, Y. Mineyuki, and T. Motomura. 2014.
Electron tomographic analysis of cytokinesis in the brown alga Silvetia
babingtonii (Fucales, Phaeophyceae). Protoplasma 251 (6):1347–1357.
Patterson, D. J. 1999. The diversity of eukaryotes. The American Naturalist 154
(S4):S96–S124.
Raimundo, S. C., U. Avci, C. Hopper, S. Pattathil, M. G. Hahn, and Z. A. Popper.
2016. Immunolocalization of cell wall carbohydrate epitopes in seaweeds:
Presence of land plant epitopes in Fucus vesiculosus L. (Phaeophyceae). Planta
243 (2):337–354.
Raimundo, S. C., S. Pattathil, S. Eberhard, M. G. Hahn, and Z. A. Popper. 2017.
β-1,3-Glucans are components of brown seaweed (Phaeophyceae) cell walls.
Protoplasma 254 (2):997–1016.
Torode, T. A., A. Siméon, S. E. Marcus, M. Jam, M.-A. Le Moigne, D. Duffieux,
J. P. Knox, and C. Hervé. 2016. Dynamics of cell wall assembly during early
embryogenesis in the brown alga Fucus. Journal of Experimental Botany
67:6089–6100.
Venable, J. H., and R. Coggeshall. 1965. A simplified lead citrate stain for use in
electron microscopy. The Journal of Cell Biology 25 (2):407–408.
Wargacki, A. J., E. Leonard, M. N. Win, D. D. Regitsky, C. N. S. Santos, P. B. Kim,
S. R. Cooper et al. 2012. An engineered microbial platform for direct biofuel
production from brown macroalgae. Science 335 (6066):308–313.
Webster, P. 2007. Microwave-assisted processing and embedding for transmission
electron microscopy. In Electron Microscopy: Methods and Protocols, J. Kuo
(Ed.), pp. 47–65. Totowa, NJ: Humana Press.
Yoon, H. S., J. D. Hackett, C. Ciniglia, G. Pinto, and D. Bhattacharya. 2004.
A molecular timeline for the origin of photosynthetic eukaryotes. Molecular
Biology and Evolution 21 (5):809–818.
chapter twenty seven
Contents
27.1 Introduction ..........................................................................................411
27.2 State of the art ...................................................................................... 412
27.3 Materials ............................................................................................... 412
27.3.1 Materials for optical microscopy ........................................ 412
27.3.2 Materials for scanning electron microscopy ..................... 413
27.4 Experimental procedures....................................................................414
27.4.1 Choice of the portion to cut and orientation of the
sample ......................................................................................414
27.4.2 Optical microscopy ............................................................... 420
27.4.3 Scanning electron microscopy ............................................ 423
27.5 Notes ..................................................................................................... 425
Acknowledgments ......................................................................................... 427
References........................................................................................................ 427
27.1 Introduction
Coralline algae (CA) are distributed from tropical regions to the poles,
thriving from the intertidal to the lower boundaries of the euphotic zone.
Because of the calcium carbonate present in their cell walls, they fill a
paramount role as bioconstructors of coral reefs, coralligenous, rhodolith
beds, providing three-dimensional habitats and contributing to carbon-
ate deposition in marine ecosystems. CA are object of studies related to
global change and are also used in paleoenvironmental reconstruction
as biological datalogger (McCoy and Kamenos 2015; Kamenos et al. 2016).
The Marine Strategy Framework Directive (MSFD; European Parliament
and Council of the European Union 2008; European Commission 2010)
has included the assessment of CA distribution and biodiversity within
411
412 Protocols for Macroalgae Research
27.3 Materials
27.3.1 Materials for optical microscopy
For the decalcification and stain of CA, different solutions can be used
(Woelkerling 1988; Bressan and Babbini 2003).
Chapter twenty seven: Coralline algae preparation for SEM and OM 413
(a)
(b)
(c)
(d)
Figure 27.1 Material required for the cut of samples (a–d): Single-edged razor
blade, chisels with fine tip, and hammer.
414 Protocols for Macroalgae Research
T T T T
T T
T
Figure 27.2 Section of the thallus showing solitary and paired trichocytes (T).
Note the neck (arrowheads) of trichocyte megacells and the normal vegetative
cells (arrows) between the solitary and paired trichocytes.
Chapter twenty seven: Coralline algae preparation for SEM and OM 415
Vegetative features
P
CC
EF
NC
(a) (b) (c)
S
S
(d) (e) S (f )
Reproductive features
PP
RC
(d) (e) (f )
F
F
Figure 27.5 Transverse section of Sporolithon sp. Cells with both fusions (F) and
secondary pit connection (arrowhead). Primary pit connections (arrow) join cells
of the same filament. Scale bar = 10 µm.
L
L
(a) (c)
T T
(b) (d)
Figure 27.6 (a) Section of crustose thallus in a longitudinal plane of view. (b) Section of crustose thallus in transverse plane of view.
Scale bar = 50 µm (a), 30 µm (b). (c) Section of protuberance in longitudinal plane of view. (d) Section of protuberance in the trans-
Chapter twenty seven: Coralline algae preparation for SEM and OM
(a) (b)
Figure 27.8 Longitudinal section (black line) of protuberance directed from its
apex (arrow) to the base.
(a) (b)
Figure 27.9 (a) Crustose thallus: transverse section parallel to the growth margin
(arrow) and perpendicular to the radial section. (b) Lamellate thallus: Transverse sec-
tion parallel to the growth margin (arrow) and perpendicular to the radial section.
(a) (b)
Figure 27.12 Surface view of a thin crust. Note the four-celled element on the
original germination disc. Scale bar = 100 µm.
Chapter twenty seven: Coralline algae preparation for SEM and OM 421
(a) (b)
Figure 27.15 CA sample placed in a sealed box for dehydration in a series of 30%,
60%, 90%, and 100% ethanol.
10. Soften the resin blocks under an infrared lamp and trim them with
a razor blade to remove surplus resin.
11. Place the resin block on the sledge microtome. Make sections
5–15 µm thick, and place them serially placed on glass slides. Flatten
the sections with a drop of histoclear (clearing agent) and mount
them on permanent slides using some drops of Eukitt.
Figure 27.16 Fragments of samples oriented and embedded in the clay. (a) Line of
fracture directed from the apex (arrowhead) to the base of protuberance to obtain
longitudinal section. (b) Line of fracture perpendicular to the growth axis of pro-
tuberance to get a transverse plane of view. (c) Longitudinal section of encrust-
ing thallus. The line of fracture is perpendicular to the margin (arrowhead).
(d) Transverse section of lamella. The line of fracture is parallel to the growth
margin (arrowhead).
424 Protocols for Macroalgae Research
(a) (b)
Figure 27.17 Samples mounted on stubs. (a) Protuberance. (b) Lamella. Longitu-
dinal sections are indicated with arrows; note that in the lamellate fragment the
longitudinal section is perpendicular to the margin (M). Transverse sections
are shown with arrowheads.
Figure 27.18 Stubs with samples collocated in the sputter coater for coating with
gold/palladium mixture.
Chapter twenty seven: Coralline algae preparation for SEM and OM 425
13. Samples are thus ready for SEM observations (Figure 27.19). If dif-
ferent planes of a sample need to be examined, the stub should be
placed individually into the SEM chamber to allow the tilting of it.
Otherwise, if only the upper planes of the samples require observa-
tion, several stubs can be placed together in the chamber and viewed
consecutively.
27.5 Notes
Note 1: Use the mixture immediately after having added accelerator to
the resin. It is very important to seal the box, to exclude any contacts
with oxygen and to prevent surface ripples in hardened resin blocks.
Note 2: Usually, the selected portions of CA should include one or more
of the following: margins, lamellae, protuberance, and areas with
reproductive structures, as shown in Figure 27.20a–d. For taxa with
protuberances, the sections must be done on both protuberances
and crustose parts. In case of thin crusts, better is to cut them along
with the substratum (Figure 27.20a) to avoid breaking of the sample.
Note 3: Orient sample in the clay to obtain the desired section
(Figure 27.16). Prepare the clay blocks just before embedding the
samples into it. The thickness of the clay should not exceed 5 mm
otherwise it could be too hard to be cut.
426 Protocols for Macroalgae Research
(a) (b)
(c) (d)
Figure 27.20 (a) Margin of the encrusting thallus (arrow). (b) Thallus with lamel-
lae (arrows). (c) Unattached thallus (rhodolith) with protuberances (arrows).
(d) Multiporate conceptacles of Mesophyllum sp. (arrows).
Note 4: The razor can be used several times, paying attention that it is
not damaged. If damaged razor is used the fracture can be compro-
mised and not useful for observation.
Note 5: As the glue rapidly hardens, determine prior how to orient frag-
ments on the stub. Place the samples with the plane to observe facing
up, as shown in Figure 27.17a. If the sample is oriented with the plane
to observe is placed lateral (Figure 27.17b), during SEM observations
the stub can be tilted also allowing these planes to be viewed. It is
a good practice to put several fragments of the same sample on a
single stub to have sufficient material to examine. The labeling of
the stubs must be done on its bottom side to prevent labels being
covered during coating phase.
Chapter twenty seven: Coralline algae preparation for SEM and OM 427
Acknowledgments
Figure 27.2 has been previously published in Wolf et al., 2015 (Phytotaxa
224(1): 59–71).
References
Alexandersson T. 1974. Carbonate cementation in coralline algal nodules in the
Skagerrak, North Sea; biochemical precipitation in undersaturated waters.
J. Sediment. Petrol. 44: 7–26.
Bailey A., Bisalputra T. 1970. A preliminary account of the application of thin-
sectioning, freeze-etching, and scanning electron microscopy to the study
of coralline algae. Phycologia 9: 83–101.
Basso D., Babbini L., Kaleb S., Bracchi V.A., Falace A. 2016. Monitoring deep
Mediterranean rhodolith beds. Aquatic Conserv: Mar. Freshw. Ecosyst. 26:
549–561.
Borowitzka M.A., Larkum A.W.D., Nockolds C.E. 1974. A scanning elctron micro-
scope study of the structure and organization of the calcium carbonate
deposits of algae. Phycologia 13: 195–203.
Bressan G., Babbini L. 2003. Biodiversità marina delle coste Italiane: Corallinales
del Mar Mediterraneo: Guida all deteminazione. Biologia Marina Mediterranea
10(Suppl. 2): 1–237.
Chamberlain Y.M. 1983. Studies in the Corallinaceae with special reference to
Fosliella and Pneophyllum in the British Isles. Bulletin of the British Museum
(Natural History) Botany 11: 291–463.
European Commission. 2010. Commission decision of 1 September 2010 on crite-
ria and methodological standards on good environmental status of marine
waters. Official Journal of the European Union L 232/14 .
European Parliament and Council of the European Union. 2008. Directive
2008/56/EC of the European Parliament and of the Council of 17 June 2008
establishing a framework for Community action in the field of marine envi-
ronmental policy (Marine Strategy Framework Directive). Official Journal of
the European Union L 164/19.
Ghirardelli L.A. 2002. Endolithic microorganisms in live and dead thalli of coral-
line red algae (Corallinales, Rhodophyta) in the Northern Adriatic Sea. Acta
Geologica Hispanica 37(1): 53–60.
Harvey A., Woelkerling W., Farr T., Neill K., Nelson W. 2005. Coralline Algae
of Central New Zealand: An Identification Guide to Common ‘‘Crustose’’ Species.
National Institute of Water & Atmospheric Research Information Series
No. 57, Wellington, 145 p.
Irvine L.M., Chamberlain Y.M. 1994. Seaweeds of the British Isles, Vol. 1, Rhodophyta
Part 2B Corallinales, Hildenbrandiales. HMSO, London. p. 276.
Kamenos N.A., Burdett H.L., Darrenogue N. 2016. Coralline algae as palaeo-
climatic proxies. In Riosmena-Rodríguez R., Nelson W., Aguire J. (Eds.),
Rhodolith/Maerl Beds: A Global Perspective. Switzerland: Springer.
Littler M.M. 1976. Calcification and its role among macroalgae. Micronesica 12:
27–41.
428 Protocols for Macroalgae Research
Contents
28.1 Introduction ......................................................................................... 429
28.2 State of the art ...................................................................................... 430
28.3 Materials ............................................................................................... 432
28.3.1 Algal material ........................................................................ 432
28.3.2 Sample homogenization ....................................................... 432
28.3.3 RNA extraction ...................................................................... 432
28.3.3.1 Reagents ................................................................ 432
28.3.3.2 Equipment............................................................. 432
28.3.4 RNA quality control ............................................................. 433
28.4 Experimental procedures................................................................... 433
28.4.1 Sample homogenization ....................................................... 433
28.4.2 RNA extraction ...................................................................... 433
28.4.3 RNA quality control ............................................................. 435
28.4.3.1 Measuring RNA concentration and purity ...... 435
28.4.3.2 RNA integrity control ......................................... 436
28.5 Notes ..................................................................................................... 436
Acknowledgments ......................................................................................... 437
References........................................................................................................ 437
28.1 Introduction
Environmental genomics and transcriptomics are powerful approaches
to gain novel insights into evolutionary ecology, organism–environment
interactions, and ecosystem processes (Vandenkoornhuyse et al. 2010).
Main aims of transcriptomics are to catalog all expressed RNA species,
to determine the transcriptional structure of genes, and to quantify
429
430 Protocols for Macroalgae Research
RNA in low ionic strength buffers (Falcão et al. 2008). Furthermore, RNA
extracted from macroalgae is often contaminated with acidic polysaccha-
rides because of almost identical densities between those molecules (Kim
et al. 1997). Commercially available kits using guanidinium salts (e.g.,
the RNeasy Mini Kit; Qiagen, and Hildesheim) usually fail for brown
algae. A more promising approach consists of the utilization of a cet-
yltrimethylammonium bromide (CTAB)-based buffer and subsequent
phenol–chloroform purification followed by selective precipitation with
LiCl (Pearson et al. 2006, Dittami et al. 2009). One disadvantage of this
method is that lithium salts, even in minute amounts, often interfere with
downstream enzymatic applications (e.g., reverse transcription) (Bilgin
et al. 2009). Here, a fast and effective approach to extract high-quality
RNA from brown algae for transcriptomic analysis is presented. The
developed protocol consists of a CTAB extraction followed by the usage
of the RNeasy Plant Mini Kit (Qiagen, Hildesheim, Germany), applying
a modified protocol based on the company’s user manual (Heinrich et al.
2012a). The extraction protocol was tested successfully in several brown
algae (e.g., S. latissima, Desmarestia anceps, and Fucus serratus) and in the
green alga Ulva sp.; thus it might also be effective in the red algae. The
isolated RNA was successfully used for downstream applications such as
RT-PCR, Microarray hybridizations, 454 sequencing, and Illumina high-
throughput RNA sequencing (Heinrich et al. 2012a, b, Heinrich et al. 2016,
Iñiguez et al. 2017). A comparison of different RNA extraction methods
for brown algae regarding RNA yield, purity, and integrity can be found
in Table 28.1.
28.3 Materials
28.3.1 Algal material
Laboratory cultures, and field materials, can be used for RNA extrac-
tion. RNA isolation from culture material proves to be easier as it typi-
cally shows a low degree of contamination with endo- and epiphytes
and often contains lower amounts of polysaccharides and polyphenolic
compounds. When using materials collected in the field, collection sites
should be well considered. For example, material collected from sites
with strong grazing pressure exhibits higher contents of polyphenolic
compounds; sporophytes from strong wave-exposed sampling sides usu-
ally contain higher amounts of polysaccharides. Both substances might
hamper the RNA-extraction process. Furthermore, field material should
be carefully cleaned before extracting RNA to minimize contamination.
To prevent RNA degradation, samples should be stored at −80°C and pro-
cessed within four weeks.
28.3.3.2 Equipment
• 2 mL microcentrifuge tubes (PCR-clean)
• Microcentrifuge, for example, Eppendorf 5418R (Eppendorf GmbH,
Hamburg, Germany)
• Thermoblock
• Vortexer with a microcentrifuge adapter
• Pipettes 1000, 100, and 10 µL and corresponding tips.
Chapter twenty eight: Extraction of high quality RNA from brown algae 433
1. Precool the mortar by pouring liquid nitrogen into it, precool the
pestle, and microcentrifuge tubes and chemical spoon by placing
them into a liquid nitrogen dewar.
2. Add the sample (at least 500 mg) into the mortar still containing liq-
uid nitrogen and grind with the pestle until a fine powder is pro-
duced (Note 1).
3. Transfer the homogenized sample into a precooled 2 mL microcen-
trifuge tube using the precooled chemical spoon. At most 1/4 of the
cup should be filled with the homogenized sample. Store the sample
at −80°C until further use to avoid RNA degradation.
65
60
55
50
45
Time (s)
40
35
30
25
20
15
L 1 2 3 4 5 6 7 8 9 10 11 12
28.5 Notes
Note 1: Do not allow the sample to thaw during grinding. Add extra
liquid nitrogen to the mortar during the homogenization to avoid
thawing. This should be done very slowly to avoid splashing of the
material.
Note 2: Do not allow the homogenized material to thaw.
Note 3: Incubation time can be raised up to 30 min if only small amounts
of biomass are available or if the RNA extraction yields from previ-
ous extractions have been insufficient.
Chapter twenty eight: Extraction of high quality RNA from brown algae 437
28S 28S
18S 18S
1 2
(FU) (FU)
500
500
0 0
15 20 25 30 35 40 45 50 55 (s) 15 20 25 30 35 40 45 50 55 (s)
3 4
(FU) (FU)
40 20
20 10
0 0
15 20 25 30 35 40 45 50 55 (s) 20 25 30 35 40 45 50 55 60 65 (s)
Acknowledgments
This work was partly funded by the Deutsche Forschungsgemeinschaft
(DFG) in the framework of the priority program “Antarctic Research with
comparative investigations in Arctic ice areas” by a grant (HE 6734/1-1).
Andreas Wagner and Claudia Daniel are acknowledged for technical
assistance.
References
Becker, C., A. Hammerle-Fickinger, I. Riedmaier, and M.W. Pfaffl. 2010.
mRNA and microRNA quality control for RT-qPCR analysis. Methods
50 (4):237–243.
Bilgin, D.D., E.H. DeLucia, and S.J. Clough. 2009. A robust plant RNA isolation
method is suitable for Affymetrix GeneChip analysis and quantitative real-
time RT-PCR. Nat. Protoc. 4 (3):333–340.
Cock, J.M., L. Sterck, P. Rouze et al. 2010. The Ectocarpus genome and the indepen-
dent evolution of multicellularity in brown algae. Nature 465 (7298):617–621.
Copois, V., F. Bibeau, C. Bascoul-Mollevi et al. 2007. Impact of RNA degradation on
gene expression profiles: Assessment of different methods to reliably deter-
mine RNA quality. J. Biotechnol. 127 (4):549–559.
438 Protocols for Macroalgae Research
Deng, Y., J. Yao, X. Wang, H. Guo, and D. Duan. 2012. Transcriptome sequencing
and comparative analysis of Saccharina japonica (Laminariales, Phaeophyceae)
under blue light induction. PLoS One 7 (6):e39704.
Dittami, S.M., D. Scornet, J.L. Petit et al. 2009. Global expression analysis of the
brown alga Ectocarpus siliculosus (Phaeophyceae) reveals large-scale repro-
gramming of the transcriptome in response to abiotic stress. Genome Biol.
10 (6):51.
Ekblom, R. and J. Galindo. 2011. Applications of next generation sequencing in
molecular ecology of non-model organisms. Heredity 107 (1):1–15.
Falcão, V.D.R., A.P. Tonon, M.C. Oliveira, and P. Colepicolo. 2008. RNA Isolation
method for polysaccharide rich algae: Agar producing Gracilaria tenuistipi-
tata (Rhodophyta). J. Appl. Phycol. 20 (1):9–12.
Fleige, S. and M.W. Pfaffl. 2006. RNA integrity and the effect on the real-time qRT-
PCR performance. Mol. Asp. Med. 27 (2):126–139.
Heinrich, S., S. Frickenhaus, G. Glöckner, and K. Valentin. 2012a. A comprehensive
cDNA library of temperature stressed Saccharina latissima (Phaeophyceae).
Eur. J. Phycol. 47 (2):83–94.
Heinrich, S., K. Valentin, S. Frickenhaus, U. John, and C. Wiencke. 2012b.
Transcriptomic analysis of acclimation to temperature and light stress in
Saccharina latissima (Phaeophyceae). PLoS One 7 (8):e44342.
Heinrich, S., K. Valentin, S. Frickenhaus, and C. Wiencke. 2015. Temperature
and light interactively modulate gene expression in Saccharina latissima
(Phaeophyceae). J. Phycol. 51:93–108.
Heinrich, S., K. Valentin, S. Frickenhaus, and C. Wiencke. 2016. Origin matters—
Comparative transcriptomics in Saccharina latissima (Phaeophyceae). J. Exp.
Mar. Biol. Ecol. 476:22–30.
Iñiguez, C., S. Heinrich, L. Harms, and F.J.L. Gordillo. 2017. Increased temperature
and CO2 alleviate photoinhibition in Desmarestia anceps: From transcrip-
tomics to carbon utilization. J. Exp. Bot. 68:3971–3984.
Johnson, M.T.J., E.J. Carpenter, Z. Tian et al. 2012. Evaluating methods for isolat-
ing total RNA and predicting the success of sequencing phylogenetically
diverse plant transcriptomes. PLoS One 7 (11):e50226.
Kim, S.H., Y.K. Lee, C.B. Hong, and I.K. Lee. 1997. A simple method for RNA
extraction from marine macroalgae. Algae 12:53–56.
Lane, C.E., C. Mayes, L.D. Druehl, and G.W. Saunders. 2006. A multi-gene molec-
ular investigation of the kelp (Laminariales, Phaeophyceae) supports sub-
stantial taxonomic re-organization. J. Phycol. 42 (2):493–512.
Liang, X., X. Wang, S. Chi et al. 2014. Analysis of Saccharina japonica transcriptome
using the high-throughput DNA sequencing technique and its vanadium-
dependent haloperoxidase gene. Acta Oceanol. Sin. 33 (2):27–36.
Pearson, G., G. Hoarau, A. Lago-Leston et al. 2010. An expressed sequence tag
analysis of the intertidal brown seaweeds Fucus serratus (L.) and F. vesiculo-
sus (L.) (Heterokontophyta, Phaeophyceae) in response to abiotic stressors.
Mar. Biotechnol. 12 (2):195–213.
Pearson, G., A. Lago-Leston, M. Valente, and E. Serrao. 2006. Simple and rapid
RNA extraction from freeze-dried tissue of brown algae and seagrasses.
Europ. J. Phycol. 41 (1):97–104.
Vandenkoornhuyse, P., A. Dufresne, A. Quaiser et al. 2010. Integration of molecu-
lar functions at the ecosystemic level: Breakthroughs and future goals of
environmental genomics and post‐genomics. Ecol. Lett. 13 (6):776–791.
Chapter twenty eight: Extraction of high quality RNA from brown algae 439
Contents
29.1 Introduction ......................................................................................... 441
29.2 State of the art ...................................................................................... 443
29.3 Materials ............................................................................................... 444
29.3.1 Equipment .............................................................................. 444
29.3.1.1 Algae cultivation .................................................. 444
29.3.1.2 Laser microdissection ......................................... 444
29.3.2 Reagents.................................................................................. 445
29.4 Experimental procedures................................................................... 445
29.4.1 Cultivation of Spirogyra and induction of sexual
reproduction .......................................................................... 445
29.4.1.1 Media preparation ............................................... 445
29.4.1.2 Vegetative growth ................................................ 446
29.4.1.3 Sexual reproduction induction .......................... 446
29.4.2 Fixation of thallus before laser capture microdissection..... 447
29.4.3 Laser capture microdissection ............................................ 449
29.5 Notes ..................................................................................................... 449
Acknowledgment ........................................................................................... 451
References........................................................................................................ 451
29.1 Introduction
Zygnematophyceae belong to the paraphyletic Charophyte group, which
is composed entirely of unicellular and multicellular algae. Together
with the land plants, Charophytes form a monophyletic group called
Streptophyta. Charophytes are of particular interest for evodevo studies
441
442 Protocols for Macroalgae Research
zygotes that are suitable material for RNA extraction. These protocols have
been used to obtain gamete- and zygote-specific transcriptomes after RNA
amplification and sequencing on an Illumina platform (unpublished data).
29.3 Materials
29.3.1 Equipment
29.3.1.1 Algae cultivation
• Temperature and light-controlled growth chamber
• Laminar flowhood
• 500 µL sterile cell culture plastic flask with vented cap
• Glass Pasteur pipettes
• 1 L glass bottles
• 50 mL sterile plastic tubes
• Horizontal microscope slide holder
• Plastic sandwich boxes to contain 200–500 mL
• 0.2 µm Millipore filter and large plastic syringe
• Standard lab equipment (gloves, etc.)
• 500 µL plastic tubes with adhesive tube caps (Carl Zeiss Microscopy,
#415190-9201-000)
• Soft paint brush
29.3.2 Reagents
• Bold modified basal freshwater solution 50X (SIGMA #B5282); on
arrival, aliquot stock in 50 mL plastic tubes and freeze at −20°C
• Vitamins:
• Kao and Michayluk vitamin solution 100X (SIGMA #K3129)
Or
• Biotin powder (SIGMA #B4639)
• Cyanocobalamin (SIGMA #C3607)
• Thiamine (SIGMA #T1270)
• Pure water
• Ethanol 100%
• Acetic acid 100%
• ProtectRNA™ RNase Inhibitor 500X concentrate (SIGMA #R7397)
• RNaseZap™ RNase Decontamination solution (Invitrogen #AM9780)
• Poly-l-lysine solution 0.1% (w/v) in H2O (SIGMA #P8920)
• RNase-free water
• Silica gel
(c)
f
f
m
(d)
f
m
(b) (e)
1. Place the PEN membrane slides under UV rays for 30 min. This helps
to make the plastic surface more hydrophilic.
2. Pour ~1 mL of poly-l-lysine solution onto the surface of the PEN
membrane slides and let it dry completely.
3. Place a slide in a clean horizontal slide holder. The slide holder can
be custom made (e.g., from a PEN membrane slide plastic box). It is
important that the slide can be moved while keeping it horizontal.
4. Prepare the following three solutions:
• BSM 1/10th cultivation medium
• 25% ethanol, 10% acetic acid, and 1X SIGMA RNase inhibitor
mixed with RNase-free H2O
• 25% ethanol, 1X SIGMA RNase inhibitor mixed with RNase-free
H2O
5. Clean three plastic sandwich boxes that are larger than the slide holder.
6. Place the slide holder in a plastic box then pour the BSM 1/10th
medium up to the limit of the surface of the slide. The solution must
barely cover the slide.
7. Using a dissecting microscope, select fragments of thalli where
reproduction and gametogenesis took place. Transfer pieces of frag-
ments to the surface of the membrane slide using a clean brush and
then spread the filaments equally over the slide (Note 2).
8. Very gently transfer the slide holder into a second plastic box
(keeping the slide as horizontal as possible) and then pour the 25%
ethanol, 10% acetic acid, and 1X SIGMA RNase inhibitor solution
up to the limit of the slide. Again, make sure that the solution
barely covers the slide. Note at this step that you will lose some
pieces of thallus as they will be washed off of the slide. Incubate
for 10 min.
9. Transfer the slide holder and the slide into a third plastic box and
pour the 25% ethanol, 1X SIGMA RNase inhibitor solution as above-
mentioned. Incubate for 10 s.
Chapter twenty nine: Induction of sexual reproduction in Spirogyra 449
10. Remove and transfer the slide holder and the slide to 37°C to dry
(~30 min).
11. Once dried, the slides can be kept at room temperature in a box con-
taining silica gel for a few days before LCM.
29.5 Notes
Note 1: Gametogenesis induction is at best unreliable. We found that
having very healthy cultures was crucial, and thus it is important to
subculture the algae every 4–5 days (several times) before attempting
induction. Counterintuitively, we also found that gametogenesis works
best when only a moderate number of filaments are transferred to BSM
1/10th; do not try to saturate the culture from the onset. Spirogyra can
be easily cultured on agar plates. In this case, we found that gameto-
genesis occurred more easily, even on a full BSM. Induction can thus
be attempted on 1% agar plates containing BSM 1/10th.
Note 2: Transferring Spirogyra filaments to LCM slides is a tedious task
and it is very important not to touch the membrane with a hard
450 Protocols for Macroalgae Research
(a) (b)
Acknowledgment
This work was funded by an ERC Advanced Investigator Grant (EDIP) to
JAL. We are grateful to Julie Bull for technical support.
References
Allen, M. A. 1958. The Biology of a Species Complex in Spirogyra. Bloomington, IA:
Indiana University.
Andersen, R. A., J. A. Berges, P. J. Harrison, and M. M. Watanabe. 2005. Recipes
for freshwater and saltwater media. In Algal Culturing Techniques, edited by
R. A. Andersen, pp. 429–538. London, UK: Elsevier Academic Press.
Czurda, V. 1933. Experimentelle analyse der kopulationsauslösenden Bedingungen
bei Mikroorganismen—1. Untersuchungen an Algen (Spirogyra, Zygnema
Und Hyalotheca). Beihefte zum Botanischen Centralblatt 51: 711–761.
Drummond, C. S., J. Hall, K. G. Karol, C. F. Delwiche, and R. M. McCourt.
2005. Phylogeny of Spirogyra and Sirogonium (Zygnematophyceae)
based on rbcL sequence data. Journal of Phycology 41 (5): 1055–1064.
doi:10.1111/j.1529-8817.2005.00130.x.
Emmert-Buck, M. R., R. F. Bonner, P. D. Smith, R. F. Chuaqui, Z. Zhuang,
S. R. Goldstein, R. A. Weiss, and L. A. Liotta. 1996. Laser capture micro-
dissection. Science 274 (5289): 998–1001. doi:10.1126/science.274.5289.998.
Espina, V., J. D. Wulfkuhle, V. S. Calvert, A. VanMeter, W. Zhou, G. Coukos, D. H.
Geho, E. F. Petricoin, and L. A. Liotta. 2006. Laser-capture microdissection.
Nature Protocols 1 (2): 586–603. doi:10.1038/nprot.2006.85.
Gomez, S. K., and M. J. Harrison. 2009. Laser microdissection and its applica-
tion to analyze gene expression in Arbuscular Mycorrhizal symbiosis. Pest
Management Science 65 (5): 504–511. doi:10.1002/ps.1715.
Grote, M. 1977. Über die auslösung der generativen fortpflanzung unter kontrol-
lierten bedingungen bei der grünalge Spirogyra Majuscula. Zeitschrift Für
Pflanzenphysiologie 83 (2): 95–107. doi:10.1016/S0044-328X(77)80064-0.
Hoshaw, R. W., J.-C. Wang, R. McCourt, and H. M. Hull. 1985. Ploidal changes in
clonal cultures of Spirogyra Communis and implications for species defini-
tion. ResearchGate 72 (7): 1005–1011.
Ichimura, T. 1971. Sexual cell division and conjugation-papilla formation in sexual
reproduction of Closterium Strigosum. In Proceedings of the 7th International
Seaweed Symposium, edited by K. Nishizawa, pp. 208–214, Tokyo, Japan:
University of Tokyo Press.
Ikegaya, H., T. Nakase, K. Iwata, H. Tsuchida, S. Sonobe, and T. Shimmen. 2012.
Studies on conjugation of Spirogyra using monoclonal culture. Journal of
Plant Research 125 (3): 457. doi:10.1007/s10265-011-0457-3.
Nagata, Y. 1973. Rhizoid differentiation in Spirogyra I. Basic features of rhizoid
formation. Plant and Cell Physiology 14 (3): 531–541.
Nakazono, M., F. Qiu, L. A. Borsuk, and P. S. Schnable. 2003. Laser-capture micro-
dissection, a tool for the global analysis of gene expression in specific plant
cell types: Identification of genes expressed differentially in epidermal cells
or vascular tissues of maize. The Plant Cell Online 15 (3): 583–596. doi:10.1105/
tpc.008102.
452 Protocols for Macroalgae Research
Contents
30.1 Introduction ......................................................................................... 454
30.2 State of the art ...................................................................................... 455
30.3 Materials ............................................................................................... 456
30.3.1 Strains and vector.................................................................. 457
30.3.1.1 E. coli ....................................................................... 457
30.3.1.2 Vector ..................................................................... 457
30.3.2 General stock solutions ........................................................ 457
30.3.3 PCR, cloning and transformation procedures .................. 457
30.3.4 Protein expression and purification ................................... 458
30.3.5 Equipment .............................................................................. 458
30.3.6 Software .................................................................................. 459
30.4 Experimental procedures .................................................................. 459
30.4.1 Bioinformatics analysis ........................................................ 459
30.4.2 Cloning ................................................................................... 459
30.4.2.1 Amplification of gene of interest........................ 459
30.4.2.2 PCR fragment purification .................................. 460
30.4.2.3 PCR fragment digestion ...................................... 460
30.4.2.4 pFO4 vector preparation ..................................... 460
30.4.2.5 Ligation step .......................................................... 461
30.4.2.6 Transformation in E. coli DH5α .......................... 461
30.4.2.7 PCR screening....................................................... 461
30.4.2.8 Plasmid extraction and glycerol stock............... 462
30.4.2.9 Transformation of expression strain, PCR
screening, and glycerol stock ............................. 462
30.4.3 Small-scale test expression .................................................. 462
30.4.3.1 Cultivation was performed in two phases ....... 462
30.4.3.2 Lysis of cells and detection of proteins ............. 462
453
454 Protocols for Macroalgae Research
30.1 Introduction
The marine environment is highly complex and contains the vast major-
ity of known and unknown biodiversity. It is also the last frontier to
understand the control of the global climate and hides a wealth of bio-
logical resources still to be tapped for food, health, and energy. Marine
macroalgae are thus photosynthetic eukaryotes that provide relevant
and significant biological models to gain insight into the deep evolution
of fundamental biological processes, which are at the same time comple-
mentary to those obtained by the study of other models such as plants
or animals. Seaweeds that grow in the intertidal zone live in a constant
stressful environment. They undergo the disadvantages of terrestrial
habitats combined with disadvantages of aquatic environments (desicca-
tion, freezing, osmotic stress, UV radiation, and variable temperatures), to
which the mechanical stresses imposed by waves are added. In this inter-
tidal zone, macroalgae, particularly brown algae, have a remarkable abil-
ity to adapt and acclimate to many different adverse conditions. Hence,
these organisms should possess key processes that have evolved to cope
with these frequent changes in environmental conditions.
Brown algae are a large group of marine seaweeds including almost
1800 species with a characteristic olive-green to dark-brown color
derived from fucoxanthin (Wei et al. 2013). They are multicellular and
have evolved independently from animals and land plants. They belong
to the Stramenopiles (Heterokonta), a phylum that diverged from the
Archaeplastida and Opisthokonta 1 billion years ago (Yoon et al. 2004).
Within the Stramenopiles, brown algae are a relatively recent lineage
that emerged about 200 million years ago (Brown and Sorhannus 2010;
Silberfeld et al. 2010). Little is known about their biology, as they were
mostly studied for their taxonomic and ecological diversity. So, knowl-
edge about primary metabolic processes is essential for the understand-
ing of the physiology and ecology of seaweeds.
Global genome sequencing efforts have generated large numbers of new
genes, the biological or biochemical functions of which remain unknown.
The complete sequences of more than 24,000 genomes are currently avail-
able in the public databases (https://www.ncbi.nlm.nih.gov/genome/).
Chapter thirty: Cloning and expression strategies 455
However, up to now, only three draft genomes of brown algae have been
decoded: Ectocarpus siliculosus (E. siliculosus), order Ectocarpales (Cock
et al. 2010); Saccharina japonica, order Laminariales (Ye et al. 2015); and
Cladosiphon okamuranus S-strain, order Chordariales (Nishitsuji et al.
2016). In newly sequenced genomes, genes are annotated on the basis of
sequence similarity to other proteins that have already been characterized
(Altschul et al. 1997). One major challenge is to assign biological function
and to elucidate the mechanism of action of such genes. This challenge
involves techniques to elucidate the structure and function of the gene
products, interactions between proteins, and/or global protein changes.
Overexpression of recombinant proteins is one of the possible solutions to
answer these biological questions.
30.3 Materials
The protocol described below is based on techniques used for the purifi-
cation of recombinant M1Pase and M1PDH from E. siliculosus (Groisillier
et al. 2014; Bonin et al. 2015).
Chapter thirty: Cloning and expression strategies 457
30.3.1.2 Vector
• pFO4 vector: AmpR, T7lac promoter hexa-histidine tail at the
N-terminal of recombinant (a vector modified from pET15b, Novagen)
(Groisillier et al. 2010) (Note 1).
30.3.5 Equipment
• Water bath set at 42°C
• Incubator set at 37°C
• Shaking incubator at 20°C or 37°C
• Autoclave
• 96-well thermocycler with heated lid
• OD600 absorbance reader
• Nanodrop spectrophotometer
• Sonicator
• French press
• ÄKTA Avant system (GE Healthcare, USA) or equivalent
Chapter thirty: Cloning and expression strategies 459
30.3.6 Software
• Hyper 32 (Informer Technologies, model: hyper32)
• Microsoft Excel or similar spreadsheet
• UNICORN software associated with ÄKTA Avant
30.4.2 Cloning
Gene of interest was synthetized and codon-optimized for expression
in E. coli by GeneArtR Gene Synthesis, Life Technologies (USA) (Bonin
et al. 2015) or searched in the genome sequence or the GenBank Expressed
Sequence Tag (EST) library of E. siliculosus (Groisillier et al. 2014) (Note 2).
2000
Fraction number
4 5 6
1600
Absorbance at 280 nm
1200
800
600
0
0 1 2 3 4 5 6 7 8 9 10 11 12
(a) Fraction number
6
5
Ln molecular mass
250
Fraction number
Absorbance at 280 nm
4 200 EsM1PDH1cat
150
100
3
50
Fraction number
0
20 25 30 35 40 45 50 55 60 65 70
2
1 1.2 1.4 1.6 1.8 2 2.2
(b) Ve/V0
Figure 30.1 Purification (a) and estimation of molecular mass (b) of the recombi-
nant His-tagged EsM1PDH1cat domain. (a) Proteins were purified by Ni2+ affin-
ity chromatography, and eluted fractions were analyzed by SDS–PAGE (inset);
(b) proteins were resolved by gel filtration onto a Superdex 200 HiLoad™ column
(inset), and the following proteins were used as standards for column calibration:
carbonic anhydrase (29 kDa), albumin (66 kDa), alcohol dehydrogenase (150 kDa),
b-amylase (200 kDa); Ve, elution volume; V0, void volume. (From Bonin, P. et al.,
Phytochemistry, 117, 509–520, 2015.)
30.5 Notes
Note 1: The E. coli system is the first-choice host for the initial screen-
ing of the recombinant protein expression, because these cells can
be readily manipulated, are cultured inexpensively, and grow rap-
idly. In recent years, numerous new strains, vectors, and tags have
been developed to overcome the limitations of this system, which
Chapter thirty: Cloning and expression strategies 465
75
50
37
Figure 30.2 Western blot analysis of insoluble and soluble fractions of three
clones of E. coli cell-free extracts containing catalytic domain of M1PDH (M1Short
NC, without induction), M1PDH (M1Short N), and full M1PDH (M1full NC)
of E. siliculosus. M: molecular weight marker in kDa. (From Bonin, P., Étude du
métabolisme du mannitol chez l’algue brune modèle Ectocarpus siliculosus: carac-
térisation de l’enzyme clé mannitol-1-phosphate déshydrogénase. Thèse de doctorat
de Biologie. Université Pierre et Marie Curie, Paris, France, 2014.)
466 Protocols for Macroalgae Research
References
Altschul, S.F., Madden, T.L., Schaffer, A.A., Zhang, J., Zhang, Z., Miller, W.,
Lipman, D.J. 1997. Gapped BLAST and PSI-BLAST: A new generation of pro-
tein database search programs. Nucleic Acids Res 25: 3389–3402.
Baharum, H., Morita, H., Tomitsuka, A., Lee, F.-C., Ng, K.-Y., Rahim, R.A., Abe, I.,
Ho, C.-L. 2011. Molecular cloning, modeling, and site-directed mutagenesis
of Type III polyketide synthase from Sargassum binderi (Phaeophyta). Mar
Biotechnol 13: 845–856.
Bendtsen, J.D., Nielsen, H., von Heijne, G., Brunak, S. 2004. Improved prediction of
signal peptides: SignalP 3.0. J Mol Biol 340: 783–795.
Bonin, P. 2014. Étude du métabolisme du mannitol chez l’algue brune modèle
Ectocarpus siliculosus: Caractérisation de l’enzyme clé mannitol-1-phosphate
déshydrogénase. Thèse de doctorat de Biologie. Université Pierre et Marie
Curie, Paris, France.
Bonin, P., Groisillier, A., Raimbault, A., Guibert, A., Boyen, C., Tonon, T. 2015.
Molecular and biochemical characterization of mannitol-1-phosphate
dehydrogenase from the model brown alga Ectocarpus sp. Phytochem 117:
509–520.
Brown, J.W., Sorhannus, U. 2010. A molecular genetic timescale for the diversifica-
tion of autotrophic stramenopiles (Ochrophyta): Substantive underestima-
tion of putative fossil ages. PLoS One 5: e12759.
Callebaut, I., Labesse, G., Durand, P., Poupon, A., Canard, L., Chomilier, J.,
Henrissat, B., Mornon, J.P. 1997. Deciphering protein sequence information
through hydrophobic cluster analysis (HCA): Current status and perspec-
tives. Cell Mol Life Sci 53: 621–645.
Cock, J.M., Sterck, L., Rouzé, P. et al. 2010. The Ectocarpus genome and the indepen-
dent evolution of multicellularity in brown algae. Nature 465: 617–621.
de Franco, P.O., Rousvoal, S., Tonon, T., Boyen, C. 2009. Whole genome survey of
the glutathione transferase family in the brown algal model. Mar Genomics
1: 135–148.
Festa, F., Steel, J., Bian, X., Labaer, J. 2013. High-throughput cloning and expression
library creation for functional proteomics. Proteomics 13: 1381–1399.
Fischl, R., Bertelsen, K., Gaillard, F., Coelho, S., Michel, G., Klinger, M., Boyen, C.,
Czjzek, M., Hervé, C. 2016. The cell-wall active mannuronan C5-epimerases
in the model brown alga Ectocarpus: From gene context to recombinant pro-
tein. Glycobiology 26: 973–983.
Gopal, J., Kumar, A. 2013. Strategies for the production of recombinant protein in
Escherichia coli. Protein J 32: 419–425.
Chapter thirty: Cloning and expression strategies 467
Groisillier, A., Hervé, C., Jeudy, A., Rebuffet, E., Pluchon, P.F., Chevolot, Y.,
Flament, D. et al. 2010. MARINE-EXPRESS: Taking advantage of high
throughput cloning and expression strategies for the post-genomic analysis
of marine organisms. Microb Cell Fact 9: 45.
Groisillier, A., Shao, Z., Michel, G., Goulitquer, S., Bonin, P., Krahulec, S.,
Nidetzky, B., Duan, D., Boyen, C., Tonon, T. 2014. Mannitol metabolism in
brown algae involves a new phosphatase family. J Exp Bot 65: 559–570.
Inoue, A., Satoh, A., Morishita, M., Tokunaga, Y., Miyakawa, T., Tanokura, M.,
Ojima, T. 2016. Functional heterologous expression and characterization of
mannuronan C5-epimerase from the brown alga Saccharina japonica. Algal
Res 16: 282–291.
Iwamoto, K., Shiraiwa, Y. 2005. Salt-regulated mannitol metabolism in algae. Mar
Biotechnol 7: 407–415.
Khow, O., Suntrarachun, S. 2012. Strategies for production of active eukaryotic
proteins in bacterial expression system. Asian Pac J Trop Biomed 2: 159–162.
Krogh, A., Larsson, B., von Heijne, G., Sonnhammer, E.L. 2001. Predicting trans-
membrane protein topology with a hidden Markov model: application to
complete genomes. J Mol Biol 305: 567–580.
Kurland, C., Gallant, J. 1996. Errors of heterologous protein expression. Curr Opin
Biotechnol 7: 489–493.
Meslet-Cladière, L., Delage, L., Leroux, C.J.-J., Goulitquer, S., Leblanc, C., Creis, E.,
Ar Gall, E., Stiger-Pouvreau, V., Czjzek, M., Potin, P. 2013. Structure/function
analysis of a Type III polyketide synthase in the brown alga Ectocarpus silicu-
losus reveals a biochemical pathway in phlorotannin monomer biosynthesis.
The Plant Cell 25: 3089–3103.
Nishitsuji, K., Arimoto, A., Iwai, K., Sudo, Y., Hisata, K., Fujie, M., Arakaki, N. et al.
2016. A draft genome of the brown alga, Cladosiphon okamuranus, S-strain:
A platform for future studies of “mozuku” biology. DNA Res 23: 561–570.
Quax, T.E., Claassens, N.J., Söll, D., van der Oost, J. 2015. Codon bias as a means to
fine-tune gene expression. Mol Cell 59: 149–161.
Rosano, G.L., Ceccarelli, E.A. 2014. Recombinant protein expression in Escherichia
coli: Advances and challenges. Front Microbiol 5: 172.
Rousvoal, S., Groisillier, A., Dittami, S.M., Michel, G., Boyen, C., Tonon, T. 2011.
Mannitol-1-phosphate dehydrogenase activity in Ectocarpus siliculosus, a key
role for mannitol synthesis in brown algae. Planta 233: 261–273.
Shao, Z., Zhang, P., Li, Q., Wang, X., Duan, D. 2014. Characterization of mannitol-
2-dehydrogenase in Saccharina japonica: Evidence for a new polyol-specific
longchain dehydrogenases/reductase. PLoS One 9: e97935.
Silberfeld, T., Leigh, J.W., Verbruggen, H., Cruaud, C., de Reviers, B., Rousseau, F. 2010.
A multi-locus time-calibrated phylogeny of the brown algae (Heterokonta,
Ochrophyta, Phaeophyceae): Investigating the evolutionary nature of the
“brown algal crown radiation”. Mol Phylogenetics Evol 56: 659–674.
Studier, F.W. 2005. Protein production by auto-induction in highdensity shaking
cultures. Protein Expr Purif 41: 207–234.
Tenhaken, R., Voglas, E., Cock, J.M., Neu, V., Huber, C.G. 2011. Characterization
of GDP-mannose dehydrogenase from the brown alga Ectocarpus silicu-
losus providing the precursor for the alginate polymer. J Biol Chem 286:
16707–16715.
468 Protocols for Macroalgae Research
Polyethylene glycol-mediated
transformation in the green
macroalga Ulva mutabilis
(Chlorophyta)
A forward genetics approach
Jens Boesger, Michiel Kwantes, and Thomas Wichard
Contents
31.1 Introduction ......................................................................................... 470
31.2 State of the art ...................................................................................... 471
31.3 Materials ................................................................................................474
31.3.1 Equipment, consumables, and reagents .............................474
31.3.1.1 Equipment and consumables ..............................474
31.3.1.2 Reagents .................................................................474
31.3.2 Ulva culture medium ............................................................ 475
31.4 Experimental procedures................................................................... 475
31.4.1 Appropriate vector preparation for transformation
of U. mutabilis ......................................................................... 475
31.4.2 Preparation of gametes of U. mutabilis for
transformation ....................................................................... 475
31.4.3 Transformation of U. mutabilis gametes ............................. 476
31.4.4 Cultivation of transgenic Ulva lines and selection by
phleomycin treatment .......................................................... 477
31.4.5 Determination of the transformation efficiency and
further single-strain cultivation.......................................... 478
31.4.6 Confirmation of stable transformation .............................. 478
31.5 Notes ..................................................................................................... 481
Acknowledgments ......................................................................................... 482
References........................................................................................................ 482
469
470 Protocols for Macroalgae Research
31.1 Introduction
The number of research groups looking for opportunities to transform
algae has increased steadily with the increasing interest in algae as novel
plant model organisms and potential next-generation fuel producers
(Walker et al. 2005, Hallmann 2007, Mikami 2013). Genetic engineering
programs aim to manipulate algal metabolism, but a stable transforma-
tion is still a prerequisite and often the limiting factor to achieving the
next level of skills in macroalgal research.
Vector transformation is the process by which exogenous DNA is
transferred into a host organism. Methods based on polyethylene gly-
col (PEG)-mediated DNA transfer, particle bombardment, electropora-
tion, and gene transfer using Agrobacterium are common approaches to
create transgenic cell lines, which were also all applied to green algae
(Kathiresan et al. 2009, Oertel et al. 2015). Transformation can be divided
into genetic (stable) and transient transformations, which has recently
been more frequently reported for macroalgae (Mikami 2013). On trans-
formation, genes introduced by genetic recombination are maintained in
the genome through generations of cells, whereas in transient transforma-
tion, rapid loss of introduced foreign genes is usually observed. At this
moment, the critical step for genetic transformation is to generate a plas-
mid vector containing all the genetic elements for expression, including
promoter, enhancer, and transcription termination sequences. The vector
involves the manipulation of selection marker expression cassettes (usu-
ally a gene conferring resistance to an antibiotic) for either selection of
the plasmid in bacteria during the cloning steps or gene transfer in the
host organism. Recent genome data of several macroalgae now support
the application of recombinant techniques for these species (Cock et al.
2010, Collén et al. 2013), because they provide a rich resource of DNA ele-
ments, such as, for example, promoters that can be chosen for transgenic
expression.
Macroalgae very often reveal a strong resistance to antibiotics, so that
an appropriate selection of antibiotic markers is hard to find. At the same
time, morphogenesis of macroalgae (i.e., Ulva) depends on its associated
bacteria (Wichard 2015). Therefore, those essential growth-promoting
and/or morphogenesis-inducing bacteria must be equally resistant to the
chosen antibiotics such as the transgenic Ulva (Oertel et al. 2015).
Establishing the genetic-transformation system requires five main
parameters (Mikami 2013): (1) easily accessible target cells (e.g., Ulva
gametes); (2) an efficient gene transfer system; (3) an efficient expression
system for foreign genes, considering the codon usage of the host; (4) an
integration (and targeting) system to integrate the foreign gene into the
genome; and (5) a selection system for transformed lines that does not
interfere with the growth of the bacteria associated to Ulva. In fact, a
Chapter thirty one: Transformation of Ulva 471
TCCATCACACTCAACAC
Figure 31.1 Workflow of the forward genetic approach. The selection criteria and screenings for the phenotype of interest are the
bottleneck before the (multiple) insertion sites of the phleomycin-resistant cassette can be determined.
Protocols for Macroalgae Research
NcoI 4466 NcoI 4506 NcoI 5168
EcoRI 4460 NotI 73 NotI 73 EcoRI 5162 NotI 73
Chapter thirty one:
PvuI 4063
U. mutabilis U. mutabilis U. mutabilis XcmI 711
RbcS promoter XcmI 711 XcmI 711 RbcS promoter
bla RbcS promoter
Figure 31.2 Vector plasmids for the transformation of Ulva mutabilis (Oertel et al. 2015). (a–c) Codon adapted phleomycin-resistant
cassettes: RBCS. Ribulose-1.5-bisphosphate carboxylase/oxygenase small subunit; ble. phleomycin resistant gene; bla. ß-lactamase.
Suitable restriction site marked with the frame.
473
474 Protocols for Macroalgae Research
31.3 Materials
31.3.1 Equipment, consumables, and reagents
31.3.1.1 Equipment and consumables
• Bead mill, for example, Tissuelyser (Qiagen, Hilden, Germany)
• 2–3 mm diameter steel beads
• Erlenmeyer flasks (2 L)
• Nylon sieve (mesh size 30 µm)
• Micro centrifuge tubes (1.5 mL, 2 mL, and for PCR)
• Centrifuge tubes (15 and 50 mL)
• Membrane filters (0.2 µm)
• Disposable syringes (sterile; 10/20/50 mL range)
• Pipettes and tips (2/10/100/1000 µL range)
• Petri dishes (diameter 145 mm) or culture flasks (surface area
182 cm2, capacity 550 mL)
• Pasteur pipettes with long, thin opening
• Light source (e.g., a mobile fluorescent lamp)
• Common PCR thermal cycler
• Aluminum foil and paper tissues
31.3.1.2 Reagents
• Ulva culture medium (UCM, Stratmann et al. 1996)
• Ulva transformation buffer 1 (UTB1, Table 31.1)
• Ulva transformation buffer 2 (UTB2, Table 31.2)
• Phleomycin (InvivoGen, San Diego CA, USA; stock solution:
10 mg mL−1)
• Bacterial growth factors of Ulva or stationary cultures of Maribacter
sp. MS6 and Roseovarius sp. MS2 (Spoerner et al. 2012, Grueneberg
et al. 2016)
• Vector plasmids for the transformation of U. mutabilis, for example,
pPIBT7 (EU176859.1), pPIBT3 (EU176858.1), pPIBGT8 (EU196041.1)
(Figure 31.2; Oertel et al. 2015)
• Oligonucleotides: Ph-2–5′-TTAGTCCTGCTCCTCGTCCACGAAGTG-3′;
RbcS_F1—GGCTTTCCAGGAGGGCAGTC; Ubi1-F1–5′-TACGGAGGC
TGTTGCACACG-3′; Ubi1_R2—AGCAGCATGACAGACCATCTTGTG
• PrimeSTAR GXL DNA Polymerase (Takara Bio Inc, USA); alternative
Pfu-Turbo DNA Polymerase (Agilent, USA)
• Dimethyl sulfoxide
• Silica-based DNA purification kit, for example, QIAquick (Qiagen,
Hilden, Germany)
• Genomic DNA purification kit, for example, GenElute Plant Genomic
DNA Miniprep Kit (Sigma, St. Louis, MO, USA)
• Suitable restriction endonucleases
Chapter thirty one: Transformation of Ulva 475
Gametes must be used directly as they are depleted of a cell wall for
a few hours only. Therefore, the transformation procedure must be per-
formed not later than 4 h after the hatching of gametes. Moreover, zoids
from parthenosporophytes lack also a rigid cell wall, making them suit-
able for this method as well (see Note 2).
12345678
−5000 bp
−1000 bp
−5000 bp
−1000 bp
Figure 31.3 Transformants of the Ulva mutabilis mutant (slender) derived from
gametes. (a) The negative control (=transformation experiment without DNA) did
not show any surviving germling or contaminant. (b) U. mutabilis gametes were
transformed with the linearized vector plasmid pPIBT7. The picture was taken
by inverted microscopy in the third week after transformation. The transforma-
tion was conducted under the conditions described. (c) Agarose gel photograph
showing PCR genotyping results. The upper part of the gel was loaded with 5 µL
PCR product from the reaction, using primers Ph-2 and RbcS_F1 (amplicon length
1376 bp), and the lower part product from the control reaction with primers UBQ1_F1
and Ubi_R2 (amplicon length 1601 bp). PCR products, lanes 1–4: gDNA from inde-
pendently selected transformed Ulva lines; lane 5: gDNA from untransformed
Ulva; lane 6: 0.5 pg SspI-linearized pPIBT7; lane 7: water. Lane 8 contains 5 µL of
molecular marker.
478 Protocols for Macroalgae Research
Sl fl ab
3 cm 0.5 cm 500 μm
ro wr br
1 cm 1.6 cm 150 μm
tw bu sk
1 cm 1 cm 500 μm
Genomic DNA Miniprep Kit (Sigma, St. Louis, USA). Regarding PCR,
the best results were obtained using PrimeSTAR GXL DNA polymerase
(Takara Bio Inc, USA) (alternative: Pfu-Turbo DNA polymerase).
31.5 Notes
Note 1: Before the use of any transformation buffer, sterilize the buffer
by passing through a membrane filter (0.2 µm) using a sterile dispos-
able syringe.
Note 2: Ulva zoids from parthenosporophytes were transformed by the
same procedure as gametes. However, a partial purification of these
cells by their movement to the light could not be applied, because
zoids are not phototactic. Therefore, zoids were used directly after
hatching from mature sporangia. For further applications, cells must
be processed within 30 min after the sporangia have discharged the
zoids and before they start settling or aggregating.
Note 3: Development and morphogenesis in a variety of green macroal-
gae are strongly dependent on the appropriate microbiome in the
environment. Therefore, it is necessary for certain scientific issues
to work further with axenic algal cultures or in the presence of a
defined microbiome. A smart way to separate gametes from their
accompanying bacteria can be performed by the horizontal move-
ment of phototactic gametes through glass capillary pipettes (for a
detailed protocol, see Chapter 9 by Califano and Wichard 2018). The
generation of axenic insertional mutants also requires highly puri-
fied DNA, and, therefore, additional purification steps by isopropa-
nol or ethanol precipitation might be necessary.
Note 4: Increasing concentrations of the transformation vector not
only result in better transformation efficiencies but also may cause
multiple insertion events accompanied by problems in further
characterization, including the identification of the insertional
mutants.
Note 5: Because of the high viscosity of PEG 4000 and the UTB2, careful
mixing is recommended through repeated aspiration with a micro-
liter pipette with a wide-open tip.
Note 6: Based on experience, gametes do not precipitate in the highly
concentrated UTB2 solution at the tip of the 1.5 mL micro centrifuge
tubes after centrifugation. Gametes usually stick around the wall of
the microcentrifuge tubes and are only visible as a light-green halo.
Therefore, resuspend the transformed gametes carefully by repeated
aspiration with a microliter pipette with a wide-open tip or gently
shaking the tube in 500 µL UCM after discarding the supernatant.
Note 7: The concentration of phleomycin can be increased up to
50 µg mL−1.
Note 8: A cross-kingdom cross-talk between macroalgae and bacte-
ria controls a variety of algal morphogenic processes, such devel-
opment, settlement, and growth. For that reason, it is essential
to inoculate each medium with the appropriate marine bacteria.
482 Protocols for Macroalgae Research
Acknowledgments
This work is supported by the Deutsche Forschungsgemeinschaft (DFG)
through the Collaborative Research Centre 1127 “Chemical Mediators in
complex Biosystems” (T.B., M.K., and T.W.).
References
Califano, G., and T. Wichard. 2018. Preparation of axenic cultures in Ulva
(Chlorophyta). In Protocols for Macroalgae Research, B. Charrier, T. Wichard,
C.R.K. Reddy (Eds.), Chapter 9. Boca Raton, FL: CRC Press, Taylor & Francis
Group.
Cock, J.M., L. Sterck, P. Rouze et al. 2010. The Ectocarpus genome and the indepen-
dent evolution of multicellularity in brown algae. Nature 465: 617–621.
Collén, J., B. Porcel, W. Carré et al. 2013. Genome structure and metabolic features
in the red seaweed Chondrus crispus shed light on evolution of the archae-
plastida. Proc. Natl. Acad. Sci. 110: 5247–5252.
Freshwater, D.W., J.A. Dutcher, D.F. Kapraun, and R.K. Sizemore. 1990. Variation
in nuclear DNA base composition (mol % G + C) in three orders of marine
green algae. Hydrobiologia 204: 167–172.
Grueneberg, J., A. Engelen, R. Costa, and T. Wichard. 2016. Macroalgal morpho-
genesis induced by waterborne compounds and bacteria in coastal seawater.
PLoS One 11: e0146307–e07.
Hallmann, A. 2007. Algal transgenics and biotechnology. Transgenetic Plant J.
1: 81–98.
Haug, A. 1976. Influence of borate and calcium on gel formation of a sulfated poly-
saccharide from Ulva lactuca. Acta Chem. Scandinaviaca B 30: 562–566.
Hinnen, A., J.B. Hicks, and G.R. Fink. 1978. Transformation of yeast. Proc. Natl.
Acad. Sci. 75: 1929–1933.
Chapter thirty one: Transformation of Ulva 483
Kapraun, D.F. 2007. Nuclear DNA content estimates in green algal lineages:
Chlorophyta and Streptophyta. Ann. Bot. 99: 677–701.
Kathiresan, S., A. Chandrashekar, G.A. Ravishankar, and R. Sarada. 2009.
Agrobacterium-mediated transformation in the green alga Haematococcus plu-
vialis (chlorophyceae, volvocales). J. Phycol. 45: 642–649.
Kessler, R.W., T. Alsufyani, and T. Wichard. 2018. Purification of sporulation and
swarming inhibitors from Ulva: Application in algal life cycle controlling.
In Protocols for Macroalgae Research, B. Charrier, T. Wichard, C.R.K. Reddy
(Eds.), Chapter 8. Boca Raton, FL: CRC Press, Taylor & Francis Group.
Mikami, K. 2014. A technical breakthrough close at hand: Feasible approaches
toward establishing a gene-targeting genetic transformation system in sea-
weeds. Front. Plant Sci. 5: 498.
Mikami, K. 2013. Current advances in seaweed transformation. In Biochemistry,
Genetics and Molecular Biology an Integrated View of the Molecular Recognition
and Toxinology—From Analytical Procedures to Biomedical Applications, G.R.
Baptista (Ed.), Chapter 13. Rijeka, Croatia, InTech.
Oertel, W., T. Wichard, and A. Weissgerber. 2015. Transformation of Ulva mutabilis
(chlorophyta) by vector plasmids integrating into the genome. J. Phycol.
51: 963–979.
Pakhomov, M.V., G.N. Zaitseva, and A.N. Belozerskii. 1968. Presence of
5-methylcytosine and 6-methylaminopyrine in composition of DNA of cer-
tain algae. Dok. Akad. Nauk SSSR 182: 712–714.
Spoerner, M., T. Wichard, T. Bachhuber, J. Stratmann, and W. Oertel. 2012. Growth
and thallus morphogenesis of Ulva mutabilis (Chlorophyta) depends on a
combination of two bacterial species excreting regulatory factors. J. Phycol.
48: 1433–1447.
Stratmann, J., G. Paputsoglu, and W. Oertel. 1996. Differentiation of Ulva mutabilis
(chlorophyta) gametangia and gamete release are controlled by extracellular
inhibitors. J. Phycol. 32: 1009–1021.
Suzuki, R., S. Ota, T. Yamazaki et al. 2016. Morphological changes of giant mito-
chondria in the unicellular to multicellular phase during parthenogenesis of
Ulva partita (Ulvophyceae) revealed by expression of mitochondrial target-
ing GFP and PEG transformation. Phycol. Res. 64: 176–184.
Walker, T.L., C. Collet, and S. Purton. 2005. Algal transgenics in the genomic era.
J. Phycol. 41: 1077–1093.
Wichard, T. 2015. Exploring bacteria-induced growth and morphogenesis in the
green macroalga order ulvales (chlorophyta). Front. Plant Sci. 6: 86.
Wichard, T., B. Charrier, F. Mineur et al. 2015. The green seaweed Ulva: A model
system to study morphogenesis. Front. Plant Sci. 6: 72.
Index
Note: Page numbers followed by f and t refer to figures and tables respectively.
485
486 Index