(Methods in Molecular Biology 1290) Anoop Kumar, András Simon (Eds.) - Salamanders in Regeneration Research - Methods and Protocols
(Methods in Molecular Biology 1290) Anoop Kumar, András Simon (Eds.) - Salamanders in Regeneration Research - Methods and Protocols
(Methods in Molecular Biology 1290) Anoop Kumar, András Simon (Eds.) - Salamanders in Regeneration Research - Methods and Protocols
Anoop Kumar
Andrs Simon Editors
Salamanders
in Regeneration
Research
Methods and Protocols
METHODS
IN
M O L E C U L A R B I O LO G Y
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Salamanders in Regeneration
Research
Methods and Protocols
Edited by
Anoop Kumar
Institute of Structural and Molecular Biology, Division of Biosciences,
University College London, London, UK
Andrs Simon
Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
Editors
Anoop Kumar
Institute of Structural and Molecular
Biology, Division of Biosciences
University College London
London, UK
Andrs Simon
Department of Cell and Molecular Biology
Karolinska Institute
Stockholm, Sweden
ISSN 1064-3745
ISSN 1940-6029 (electronic)
Methods in Molecular Biology
ISBN 978-1-4939-2494-3
ISBN 978-1-4939-2495-0 (eBook)
DOI 10.1007/978-1-4939-2495-0
Library of Congress Control Number: 2015931955
Springer New York Heidelberg Dordrecht London
Springer Science+Business Media New York 2015
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction
on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and
regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this book are believed to
be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty,
express or implied, with respect to the material contained herein or for any errors or omissions that may have been made.
Printed on acid-free paper
Humana Press is a brand of Springer
Springer Science+Business Media LLC New York is part of Springer Science+Business Media (www.springer.com)
Preface
Many of the most fundamental discoveries in experimental biology, such as the embryonic
organizers, neuronal specificity, nerve guidance, and units of DNA transcription, originate
from salamander research. Salamanders are the only tetrapods capable of repeatedly regenerating entire limbs as adults, and they also display the widest range of regeneration capacities of other complex tissues and organs. These animals constitute unique models for
understanding critical processes underlying morphological and functional restoration of
lost or damaged structures in vertebrates. The present volume focuses on this particular
aspect of salamander biology, which has gained new momentum during the past 1015
years, partly due to the general interest in stem cells and regenerative medicine. A combined
search on Google scholar using the terms salamander and regeneration shows a steady
growth in the number of yearly publications with a 140 % increase between 2001 and 2013,
resulting in more than 10,000 published articles during this period.
There are considerable variations among the most commonly studied salamanders in
the laboratory in terms of their general physiology, life cycle, regeneration spectrum, and
also mechanisms by which replacement structures form. The first part of the book outlines
the best practices and conditions for maintaining the most commonly used salamander species in the laboratory. The chapters of the two following parts describe experimental manipulations in vivo and in vitro, respectively. These include methods targeting a wide variety of
structures, ranging from the limb to the heart and to the brain. The two final sections deal
with genetically modified organisms and tools for mining in the genomic databases. These
chapters illustrate the boom of recent technical developments, which provide new platforms for understanding salamander regeneration using the most modern molecular tools.
The methods chapters of this book are preceded by an inspiring essay on salamander regeneration from phylogenetic and evolutionary perspectives by Jeremy Brockes, who has
greatly contributed to revitalize this research field.
Finally, we thank all the colleagues for their invaluable time and efforts to provide with
all the finer details to produce this comprehensive collection of methods chapters. We
hope that this collection will be useful to all, who already are devoting our activities to
salamander regeneration, as well as for those who are just considering to dwell on to this
intriguing problem.
London, UK
Stockholm, Sweden
Anoop Kumar
Andrs Simon
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
PART I
SALAMANDERS
PART II
v
ix
17
27
47
71
vii
81
91
101
115
127
141
147
viii
Contents
PART III
PART IV
171
187
197
205
227
241
253
TRANSGENESIS IN SALAMANDERS
PART V
159
269
279
297
GENE EXPRESSION
309
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
353
321
337
Contributors
ANTONY ATHIPPOZHY, PH.D. Department of Biology, University of Kentucky, Lexington,
KY, USA
NOUR W. AL HAJ BADDAR Department of Biology, University of Kentucky, Lexington,
KY, USA
LIYAKATH ALI SHAHUL HAMEED Department of Cell and Molecular Biology,
Karolinska Institute, Stockholm, Sweden
THOMAS BRAUN, PH.D. Max-Planck-Institute for Heart and Lung Research, Bad Nauheim,
Germany
JEREMY P. BROCKES, PH.D. Institute of Structural and Molecular Biology, Division of
Biosciences, University College London, London, UK
CLAUDIA MARCELA ARENAS Grupo Gentica, Regeneracin y Cncer, Instituto
de Biologa. Universidad de Antioquia, Medelln, Colombia
PEGGY S. COOTS Department of Biology, University of Kentucky, Lexington, KY, USA
CRAIG M. CREWS, PH.D. Department of Molecular, Cellular and Developmental Biology,
Yale University, New Haven, CT, USA
RYAN J. DEBUQUE Australian Regenerative Medicine Institute (ARMI),
Monash University, Clayton, VIC, Australia
JEAN PAUL DELGADO, PH.D Grupo Gentica, Regeneracin y Cncer, Instituto de Biologa.
Universidad de Antioquia, Medelln, Colombia
JEAN-FRANOIS DENIS Department of Biochemistry, Faculty of Medicine,
Universit de Montral, Montral, QC, Canada
KAREN ECHEVERRI, PH. D. Department of Genetics, Cell Biology and Development,
Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
TETSUYA ENDO, PH.D. Division of Liberal Arts and Sciences, Aichi Gakuin University,
Nissin, Aichi, Japan
JAMI R. ERICKSON Department of Genetics, Cell Biology and Development, Stem Cell
Institute, University of Minnesota, Minnesota, USA
JOHANNA E. FARKAS Department of Biology, Northeastern University, Boston, MA, USA
PATRIZIA FERRETTI, PH.D. UCL Institute of Child Health, University College London,
London, UK
G. PARKER FLOWERS, PH.D. Department of Molecular, Cellular and Developmental
Biology, Yale University, New Haven, CT, USA
DAVID M. GARDINER, PH.D. Department of Developmental and Cell Biology, University
of California, Irvine, CA, USA
JAMES W. GODWIN, PH.D. Australian Regenerative Medicine Institute (ARMI),
Monash University, Clayton, VIC, Australia
ANDREA GMEZ-MOLINA Grupo Gentica, Regeneracin y Cncer, Instituto de Biologa.
Universidad de Antioquia, Medelln, Colombia
TOSHINORI HAYASHI, PH.D. Department of Biomedical Sciences, School of Life Science,
Faculty of Medicine, Tottori University, Tottori, Japan
ALBERTO JOVEN, PH.D. Department of Cell and Molecular Biology, Karolinska Institute,
Stockholm, Sweden
ix
Contributors
SHIVAM KHATRI Paul Laurence Dunbar High School, Lexington, KY, USA
SHAHRYAR KHATTAK, PH.D. Technische Universitt Dresden, DFG Center for Regenerative
Therapies Dresden, Dresden, Germany
MATTHEW KIRKHAM, PH.D. Department of Cell and Molecular Biology, Karolinska
Institute, Stockholm, Sweden
ANOOP KUMAR, PH.D. Institute of Structural and Molecular Biology, Division of
Biosciences, University College London, London, UK
D. KEVIN KUMP Department of Biology, University of Kentucky, Lexington, KY, USA
TZU-HSING KUO Brigham Regenerative Medicine Center and the Department
of Orthopedic Surgery, Brigham & Womens Hospital, Cambridge, MA, USA; Harvard
Medical School, Harvard Stem Cell Institute, Cambridge, MA, USA
MARIO LOOSO, PH.D. Max-Planck-Institute for Heart and Lung Research, Bad Nauheim,
Germany
AKI MAKANAE, PH.D. Research Core for Interdisciplinary Sciences (RCIS), Okayama
University, kitaku, Okayama, Japan
NOBUYASU MAKI, PH.D Institute of Protein Research, Osaka University, Osaka, Japan
JAMES R. MONAGHAN, PH.D. Department of Biology, Northeastern University, Boston,
MA, USA
SHANNON ODELBERG, PH.D. Molecular Medicine Program, Eccles Institute of Human
Genetics, Department of Internal Medicine, Cardiology Division, University of Utah,
Salt Lake City, UT, USA
TANJA PIATKOWSKI Max-Planck-Institute for Heart and Lung Research, Bad Nauheim,
Germany
AIDA RODRIGO ALBORS, PH.D. DFG Center for Regenerative Therapies TU Dresden
(CRTD), Technische Universitt Dresden, Dresden, Germany
STPHANE ROY, PH.D. Department of Stomatology, Universit de Montral, Montral,
QC, Canada
FADI SADER Department of Biochemistry, Faculty of Medicine, Universit de Montral,
Montral, QC, Canada
AKIRA SATOH, PH.D. Research Core for interdisciplinary sciences (RCIS),
Okayama University, Okayama, Japan
ASHLEY W. SEIFERT, PH.D. Department of Biology, University of Kentucky, Lexington,
KY, USA
ANDRS SIMON, PH.D. Department of Cell and Molecular Biology, Karolinska Institute,
Stockholm, Sweden
HANS-GEORG SIMON, PH.D. Department of Pediatrics, Lurie Childrens Hospital
of Chicago Research Center, Northwestern University Feinberg School of Medicine,
Chicago, IL, USA
TAKASHI TAKEUCHI, PH.D. Department of Biomedical Sciences, School of Life Science,
Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
ELLY M. TANAKA, PH.D. DFG Center for Regenerative Therapies TU Dresden (CRTD),
Technische Universitt Dresden, Dresden, Germany
RUTH R. TAYLOR, PH.D. UCL Ear Institute, University College London, London, UK
S. RANDAL VOSS, PH.D. Department of Biology, University of Kentucky, Lexington, KY, USA
JESSICA L. WHITED, PH.D. Harvard Medical School, Harvard Stem Cell Institute,
Cambridge, MA, USA
M. RYAN WOODCOCK, PH.D. Department of Biology, University of Kentucky,
Lexington, KY, USA
Part I
Salamanders
Chapter 1
Variation in Salamanders: An Essay on Genomes,
Development, and Evolution
Jeremy P. Brockes
Abstract
Regeneration is studied in a few model species of salamanders, but the ten families of salamanders show
considerable variation, and this has implications for our understanding of salamander biology. The most
recent classification of the families identifies the cryptobranchoidea as the basal group which diverged in
the early Jurassic. Variation in the sizes of genomes is particularly obvious, and reflects a major contribution from transposable elements which is already present in the basal group. Limb development has been
a focus for evodevo studies, in part because of the variable property of pre-axial dominance which distinguishes salamanders from other tetrapods. This is thought to reflect the selective pressures that operate on
a free-living aquatic larva, and might also be relevant for the evolution of limb regeneration. Recent fossil
evidence suggests that both pre-axial dominance and limb regeneration were present 300 million years ago
in larval temnospondyl amphibians that lived in mountain lakes. A satisfying account of regeneration in
salamanders may need to address all these different aspects in the future.
Key words Newt, Axolotl, Limb regeneration
Introduction
This collection of articles is about regeneration in salamanders and
various experimental approaches for working with these animals. It
is largely focused on the axolotl and some species of newt, which are
the two most widely used laboratory animals. The newt and axolotl
fall into two of the ten families of salamanders, and regeneration
research is mainly concerned to identify common, ancestral, or unifying aspects of the underlying mechanisms. In a recent study that
provided an interesting perspective, the origin of muscle-derived
cells in the limb blastema was traced to satellite cells in the axolotl,
but to multinucleate myofibers in the newt [1]. Salamanders present considerable variation between and within families in certain
aspects of their biology, and this chapter explores some examples. I
will also consider the related problems of diversity in salamander
genomes and in embryonic and larval development. The origin of
salamanders is discussed in relation to the fossil evidence from dif-
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_1, Springer Science+Business Media New York 2015
Jeremy P. Brockes
Batrachoseps major
30 nuclear genes
Eurycea bislineata
Aneides hardii
Plethodontidae
Amphiuma means
0.1 subsititutions/site
Rhyacotriton variegatus
Amphiumidae
Rhyacotritonidae
Necturus beyeri
Proteus anguinus
Proteidae
Cynops orientalis
Tylototriton asperrimus
Salamandridae
Salamandroidea
Plethodon jordani
Salamandra salamandra
99/1.0/1.0/83
1
Dicamptodon aterrimus
Ambystoma mexicanum
Dicamptodontidae
Ambystomatidae
Pseudobranchus axanthus
Sirenidae
Batrachuperus yenyuanensis
Ranodon sibiricus
99/1.0/1.0/74
Hynobiidae
Onychodactylus fischeri
Andrias davidianus
Cryptobranchidae
Bombina fortinuptialis
Cryptobranchoidea
Siren intermedia
ANURA
Silurana tropicalis
Ichthyophis bannanicus
Typhlonectes natans
GYMNOPHIONA
Gallus gallus
Chrysemys picta bellii
Mus musculus
Homo sapiens
Non-amphibian
Outgroup
Latimeria chalumnae
Fig. 1 This tree is taken from Shen et al. [4] and shows the tree for the 10 salamander families based on the
analysis of 30 genes in the 19 species shown here. Note that the basal group comprises the hynobiid and
cryptobranchid salamanders
ferent Paleozoic amphibians. I suggest that ultimately it will be necessary to understand the evolution of regenerative ability in
conjunction with these different aspects of salamander biology.
The ten families have historically been grouped into different
trees using a variety of approaches [2, 3], but perhaps the most
compelling of these has been the recent molecular analysis by
Zhang and colleagues [4]. It employed 30 nuclear protein-coding
loci that were identified by PCR from 19 salamander species
representing the 10 families. The resulting tree is shown in Fig. 1.
Jeremy P. Brockes
for complete regeneration varying from 2 to 9 months. These relationships were observed in certain plethodontid lineages and not
others. In later studies, large genomes in salamanders have been
correlated with changes in the neural circuitry [16] and in the circulatory system [17].
The most informative studies to date on the causes of genomic
gigantism in salamanders have emphasized the importance of
transposable elements (TE). Although at the time of writing no
complete salamander genome sequences are available, low-coverage
shotgun data has been generated for six plethodontid species covering a threefold range of genome size [8]. Up to 47 % of the
genome (for the species Aneides flavipunctatus) can be attributed
to TEs, but the predominant class is the long terminal repeat
(LTR) retrotransposon, which is estimated to be 30 % of the
A. flavipunctatus genome. These data raise the question of when
these sequences entered the genome during salamander evolution.
Recent analysis of the basal group genome of the hellbender, a
cryptobranchid species, has indicated that the high level of LTR
retrotransposons is likely to be characteristic of modern salamanders as a whole and to reflect persistence and diversification of
ancestral TE families [18].
It is likely that much remains to be discovered about the role
of TEs in salamander biology, but the study by Zhu et al. provides
a valuable entry point for some of the issues in relation to limb
regeneration [19]. The non-LTR long interspersed nucleotide
element-1 (LINE-1) retrotransposon was found to be markedly
upregulated in axolotl limb regeneration in both the mesenchymal
blastema and the wound epidermis. The genomic content of
LINE-1 elements was found to progressively increase by 1.52fold comparing the normal limb with two successive rounds of
regeneration, suggesting that productive retrotransposition is
induced by limb transection. Presumably this would apply to tissue
removal or injury in other regenerative contexts in the salamander.
If this occurs, and the data remain fragmentary at present, it needs
to be reconciled with the ability of salamanders to undergo multiple rounds of regeneration, for example, in the case of the newt
lens [20], without a detectable change in outcome. The recent
transcriptome analyses of regenerating newt heart [21] and axolotl
limb [22] have found that many different TE and endogenous retrovirus sequences are markedly upregulated in regeneration. What
might be the significance of these findings?
A recent analysis of mammalian development suggests that
regulation by endogenous retroelements plays a critical role at
early stages [23]. In the totipotent cells of the 2-cell (2C) stage
mouse embryo and in rare cells that arise in ES cell populations in
culture, retroviral GAG protein is expressed as well as many transcripts with junctions to endogenous retroelements. This promoter
activity of retroelement LTRs is turned off by the transition to
pluripotency after the 2C stage in mouse embryogenesis or by
Jeremy P. Brockes
Fig. 2 Diagram illustrating the difference between postaxial and preaxial dominance in outgrowth of the digits.
The large 2 and 1 numerals illustrate the first digits to extend in the case of salamanders or the other tetrapods. Modified from a similar diagram in Frobisch and Shubin [27]
regarded as a salamander innovation or apomorphy [27]. The analysis of Hox A and D complex gene expression in axolotl limb buds has
not revealed any differences from other tetrapods that appear to
explain the mechanism of preaxial dominance [29].
The significance of preaxial dominance can be appreciated in
terms of the selective pressures on aquatic larvae (or embryos for
direct developing species) in different locales. Most salamanders
have extended larval periods, which can last for more than a year in
some species. In species with pond-type larvae, limb development
proceeds slowly after hatching while the larvae move aroundthe
limbs are moved as they are developing. These larvae use the preaxial part of the autopod for locomotion and support. Such larvae
often have transient balancer organs at hatching to hold the animal
in place during the initial phase of limb elongation. The larvae of
Salamandrella keyserlingii in the Urals have a long fin-like mesenchymal membrane between digits 2 and 1 to assist movement and
floating during active feeding [30]. In stream-adapted larvae, for
example, the Pacific giant salamander, limb development is
advanced at hatching and the forelimb elements are completely
differentiated [31]. In plethodontids with direct development,
there is no larva, and the limbs develop within the capsule [32].
Nonetheless all of these cases show the extension of the limb axis
through digit 2, not digit 4. This is particularly marked for the
pond and stream-dwelling larvae, but it has also been observed for
the plethodontid case [32]. This is noteworthy because the latter
appears in other respects to be more reminiscent of amniote limb
development. The limb bud extends in a paddle configuration and
the digits form in a concerted fashion, in contrast to the iterative
episodes seen in digit development in the larval forms [32]. The
variations in limb development illustrate the operation of strong
selective pressures, particularly on the free-living aquatic larva,
which is considered to be the ancestral life history for salamanders
[26, 33]. It is possible to suggest that these selective pressures
might operate for regeneration of the limb in a context where predation and density-dependent biting behavior have been observed
[34, 35].
Although the basale commune is specific to salamanders, it is an
example of the reduction of bones in the limb, a widespread feature of tetrapod evolution [36]. In some salamander familiesthe
amphiumids, proteids, and sirenidsthe limbs may be reduced so
as to have only two digits. For example, the cave-dwelling salamander Proteus has two digits in the foot and three in the hand. In
these highly reduced salamander limbs, the remaining digits are
always the preaxial 1 and 2. It is interesting that in this context the
missing digits do not form in development, whereas a recent
analysis of digit loss in crocodiles and birds has provided examples
where digit remnants are clearly observed [37].
The nature of digit development in salamanders is of particular
interest at present, when the currently favored model for digit formation in amniotes posits a Turing-type reaction-diffusion network in the limb bud, leading to the concerted formation of the
digits [38]. Yet it is well documented that larval salamanders may
show extension of digits 2/1, followed by a gap of several days,
then formation of digit 3, another gap, and formation of digit 4
[32]. It seems somewhat unlikely that this could reflect the operation of a reaction-diffusion network as proposed for mice. It will be
interesting to see if the salamander is just a special case, even a
distinct mechanism for generating the digits, or whether the
reaction-diffusion network remains the most supported possibility
for amniotes.
10
Jeremy P. Brockes
11
Fig. 4 Fossil of the early Permian from Rhineland-Pfalz showing a single large
Micromelerpeton at the right and three smaller examples of Apateon at the left
(authors collection). Scale bar, 5 cm
12
Jeremy P. Brockes
contents. Several examples of Micromelerpeton display limb abnormalities considered characteristic of regeneration in modern salamanders [44]. For example, the fusion of digits along the
proximodistal axis is observed, as well as the formation of narrow
additional digits. These are distinct from the abnormalities
observed in limb development in salamanders. This evidence for
limb regeneration in the fossil record is not as direct as that for
preaxial dominance [27], but nonetheless it constitutes prima facie
evidence that Micromelerpeton was capable of limb regeneration.
Gerobatrachus is a single fossil from Texas dating to the early
Permian and described originally in 2008 [45]. It is classified as a
dissorophid but its precise relationship to urodeles and anurans is a
matter of active debate [41]. The leg skeleton possesses a definitive
salamander character that is the presence of a basale commune. In
summary there is strong evidence for preaxial dominance and significant evidence for limb regeneration and the basale commune in the
possible Paleozoic ancestors of salamanders. It is important to note
that this is about 100 MY before the divergence of cryptobranchoids
and emergence of crown-group salamanders. The lake system that
may have provided the habitat for evolution of salamander-like features in dissorophids, as well as being important for fossil preservation, did not persist beyond the early Permian [41]. The changes
that occurred before the appearance of lower or middle Jurassic
cryptobranchoids were in part concerned with the challenges of terrestrial life, such as feeding or hearing [39]. They include the development of a tongue, which is apparently not a regenerative structure
in modern salamanders, in contrast to the jaws [46].
Conclusions
Our ability to deliver meaningful insights into the possible extension of mammalian regeneration will be enhanced by a more
detailed understanding of the evolution of regeneration and the
factors underlying the extensive repertoire in salamanders. Limb
regeneration is a property that distinguishes salamanders from
other adult tetrapods, that is, the anuran amphibians as well as
amniotes. On the other hand, preaxial dominance is a property of
limb development that also distinguishes salamanders from other
adult tetrapods. There is a significant consensus among evolutionary biologists that preaxial dominance evolved in the salamander
lineage, possibly in response to the selective pressures operating on
free-living aquatic larvae [27]. It is plausible, as suggested above,
that the same or related pressures may extend to regeneration of
the limb. Limb regeneration could be a purely ancestral property
for tetrapods that was lost in adult anurans and amniotes, and this
remains the most popular hypothesis. Alternatively, it may be that
certain salamander-specific novelties were required in addition to
13
Acknowledgments
I thank Peng Zhang for his help in relation to salamander phylogeny and Anoop Kumar for help with the figures.
References
1. Sandoval-Guzman T, Wang H, Khattak S,
Schuez M, Roensch K, Nacu E, Tazaki A,
Joven A, Tanaka EM, Simon A (2014)
Fundamental differences in dedifferentiation
and stem cell recruitment during skeletal muscle regeneration in two salamander species.
Cell Stem Cell 14:174187
2. Larson A, Dimmick WW (1993) Phylogenetic
relationships of the salamander families: an
analysis of congruence among morphological
and molecular characters. Herpetol Monogr
7:7793
3. Duellman WE, Trueb L (1994) Biology of
amphibians. Johns Hopkins University Press,
Baltimore, MD
4. Shen XX, Liang D, Feng YJ, Chen MY, Zhang
P (2013) A versatile and highly efficient toolkit
including 102 nuclear markers for vertebrate
phylogenomics, tested by resolving the higher
level relationships of the caudata. Mol Biol
Evol 30:22352248
5. Carroll R (2009) The rise of amphibians.
The Johns Hopkins University Press,
Baltimore, MD
14
Jeremy P. Brockes
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
15
Chapter 2
Maintaining Eastern Newts (Notophthalmus viridescens)
for Regeneration Research
Hans-Georg Simon and Shannon Odelberg
Abstract
The adult Eastern newt, Notophthalmus viridescens, has long served as a model for appendage as well as
heart muscle regeneration studies. Newt tissues include all major cell types known in other vertebrates and
mammals, including bone, cartilage, tendon, muscle, nerves, dermis, and epidermis. Therefore, these
aquatic salamanders make an excellent model for studying the regeneration of complex tissues. Regeneration
of adult tissues requires the integration of new tissues with preexisting tissues to form a functioning unit
through a process that is not yet well understood. Scale is also an issue, because the regenerating tissues or
structures are magnitudes larger than their embryonic counterparts during development, and therefore, it
is likely that different physics and mechanics apply. Regardless, regeneration recapitulates to some degree
developmental processes. In this chapter, we will describe basic methods for maintaining adult Eastern
newts in the laboratory for the study of regeneration. To determine similarities and differences between
development and regeneration at the cellular and molecular level, there is also a need for embryonic newt
tissue. We therefore also outline a relatively simple way to produce and raise newt embryos in the
laboratory.
Key words Eastern newt, Red-spotted newt, Notophthalmus viridescens, Embryo, Larva, Red eft,
Breeding, Spawning, Regeneration
1
1.1
Introduction
Eastern Newts
Eastern newts are urodele amphibians, commonly called salamanders [1]. A native of eastern North America, Eastern newts belong
to the genus Notophthalmus of which there are three known species: N. viridescens (Eastern newt), N. meridionalis (black-spotted
newt), and N. perstriatus (striped newt). Notophthalmus is one of
only two genera of newts native to the United States, the other
genus being Taricha, which inhabits primarily the coastal regions
of western North America. Taxonomists currently classify Eastern
newts into four subspeciesN. v. viridescens (red-spotted newt),
N. v. dorsalis (broken-striped newt), N. v. louisianensis (central
newt), and N. v. piaropicola (peninsula newt). Red-spotted newts
are endemic to the northeastern region of the United States but
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_2, Springer Science+Business Media New York 2015
17
18
Life Cycle
Eastern newts have two distinct features that set them apart from
most other vertebrate speciestheir remarkable regenerative abilities and their unusual and complex life cycle, which can be divided
into four major stages, including the embryo, larval, red eft, and
adult stages (Fig. 1). Newts have two fascinating courtship and
breeding behaviors. Courtship may involve a stereotypical behavior known as hula during which the male undulates his body and
tail in an effort to entice the female to nudge his tail. After receiving this stimulus, the male deposits a spermatophore. The female
then uses her cloaca to pick up the sperm and stores them in a
special cavity known as the spermatheca to be used later for fertilizing her eggs. Alternatively, a more common courtship behavior
involves amplexus, in which the male grasps the females trunk
with his large hind limbs and nuzzles her with his snout. Amplexus
can go on for several hours before the male dismounts and deposits
a spermatophore in front of the female. A female newt fertilizes
Newt Husbandry
19
each egg with stored sperm just before depositing the egg on an
underwater leaf. She then gently wraps the egg with the leaf to
form a protective shield. The fertilized egg develops into an
embryo that hatches in about 2035 days. Over a period of
25 months, the gilled larvae develop first forelimbs followed by
the hind limbs. When the larvae reach a length of about 3538 mm,
they metamorphose into land-dwelling red efts. These red efts typically grow over a 3-year period before developing into mature
adults that are mostly aquatic (Fig. 1). In the wild, newts can live
for 1215 years, which is remarkable longevity for a vertebrate that
only weighs 23 g. More details concerning the life cycle of the
newt have been described elsewhere [2].
1.3 Regenerative
Abilities
Although the ancient Greeks knew about regeneration in vertebrate species as evidenced both by Aristotles notation in Historia
Animalium that lizards can regenerate their tails and the Greek
myth of liver regeneration in Prometheus, the first known scientific
study of regeneration in a vertebrate was published by the Italian
scientist Lazzaro Spallanzani in 1768 [3]. Spallanzani was able to
show that the aquatic salamander (most likely a newt) is able to
regenerate its forelimbs and hind limbs, tail, upper and lower jaws,
and caudal spinal cord. He also showed that aquatic salamanders
could repeatedly regrow a limb even after multiple amputations.
Over the past decades, major discoveries have been made using the
newt as a model organism for regeneration. In fact, the adult
Eastern newt (most often the red-spotted newt) has long served as
a primary model for the study of epimorphic regeneration of amputated limbs and tails [4, 5]. Similar to the limb, in a study spanning
16 years, Goro Eguchi and coworkers demonstrated that the
regenerative capacity of the newt lens is not altered by repeated
regeneration and aging [6]. It has also been shown that during lens
regeneration, pigment epithelial cells of the iris can transdifferentiate to lens cells [7]. The plasticity of differentiated cells has been a
great focus in regeneration studies. Adult newt cardiomyocytes
were shown to reenter the cell cycle [8, 9], and high resolution 3D
imaging as well as modern lineage tracing methods have revealed
that during limb regeneration, newt multinucleate myofibers
dedifferentiate and fragment to form proliferating mononuclear
cells that give rise to new skeletal muscle in the regenerated limb
[10, 11, 12, 13]. Moreover, the newt not only regenerates the
caudal spinal cord following tail amputation but can also regenerate the trunk spinal cord following a complete transection and
regain function of initially paralyzed appendages caudal to the
injury site [14, 15]. Modern cellular and molecular studies have
also shown that parts of the brain can regenerate by activation of
quiescent regions of the adult newt brain [16, 17].
As illustrated above, the advent of new methodologies has opened
an era in which the newt has become an attractive model to study the
cellular and molecular basis of regeneration in a vertebrate species.
20
Materials
Prepare all solutions using ultrapure water and analytical grade
reagents. Prepare and store all reagents at room temperature
(unless indicated otherwise). Diligently follow all waste disposal
regulations when disposing waste materials. The housing and
maintenance of animals must follow the guidelines of and be
approved by the Institutional Animal Care and Use Committee.
2.1
Adult Newts
2.2
Newt Water
Newt Husbandry
21
Fig. 2 Aquarium setup and swimming adult newts. (a) 80 L aquariums are
supported by metal shelves in temperature- and light cycle-controlled room. (b) A
layer of pea gravel covers the aquarium bottom with (c) aquatic plants rooted into
the gravel
22
2.4
Food
2.5 Breeding,
Spawning,
and Rearing of Newt
Larvae
Methods
Carry out all procedures at room temperature unless otherwise
specified. The use of animals must follow the guidelines of and be
approved by the Institutional Animal Care and Use Committee.
3.2 Breeding,
Spawning,
and Rearing
of Newt Larvae
Newt Husbandry
23
Notes
1. Newts freshly shipped from the vendor should be kept separate
for 2 weeks before using for experimentation. Occasionally, a
sick newt is among the shipments, and keeping animals in
quarantine helps to prevent spreading of a potential disease.
2. Newts can be handled by gently picking them up by the tail
with gloved hands. Unlike some lizards, which readily discard
their tails as a defense mechanism, the tail of the newt cannot
be discarded and therefore picking up the newt by the tail is an
appropriate method for quickly transferring animals from one
location to another. If the newt is to be handled for any length
of time beyond a simple transfer, its body should be supported
by the palm of the hand.
3. The skin of both Eastern and Western newts often contains a
potent neurotoxin known as tetrodotoxin, which binds to and
blocks voltage-gated sodium ion channels. The concentration
of tetrodotoxin has been shown to vary widely between
individuals depending on location, diet, and possibly other
unknown factors. Although controversy still remains as to the
origin of the tetrodotoxin, several studies suggest that in
Eastern newts, the toxin might be of dietary origin and if newts
24
are fed a toxin-free diet, they can lose their toxicity over a
period of several years [20]. Regardless, when handling newts
at any stage of their lives, it is best to wear gloves. If a newt is
handled without gloves, the hands should be washed immediately afterwards. Needless to say, unlike axolotls, which were
regularly eaten in Mexico before they became critically endangered (or possibly extinct in the wild), newts should never be
consumed.
4. In the wild, newts live in and by water streams, and therefore, in
principle, they can be kept in tap water in the laboratory.
However, in most cities, the drinking water is chlorinated to an
extent that it could be harmful to the animals. Therefore, if chlorinated tap water is used, it should be sufficiently dechlorinated
by allowing it to stand exposed to the air for a sufficient time
period for the chlorine to evaporate (usually several days).
However, many municipalities are treating their water supply
with monochloramine, rather than chlorine. Monochloramine
cannot be removed by evaporation. Therefore, we prefer to carefully prepare our own newt water by supplementing the deionized water supply in the laboratory with low amounts of Instant
Ocean salt to achieve a conductivity of approximately 600 SI
(equivalent to Evian Water). We have found that the method
described in this chapter produces reliable results that allow for
the maintenance and care of a healthy population of newts.
5. California blackworms can also be purchased from Eastern
Aquatics. Website: currently easternaquatics.com.
6. There is considerable competition among males for the right
to mate with females. Often males will try to dislodge a male
that is in amplexus with a female. Occasionally, a different male
not in courtship deposits his spermatophore between the spermatophore of a male in courtship and the female in an attempt
to get the female to collect sperm from his spermatophore
rather than the spermatophore from the male in courtship.
Acknowledgments
We would like to acknowledge both current and former researchers in our respective laboratories who helped develop the protocols
described in this chapter. These individuals include Paul Khan,
Barbara Linkhart, Claudia Guzman, Sarah Calve, Sarah Mercer,
Donald Atkinson, Vladimir Vinarsky, Tamara Stevenson, David
Kent, and Katherine Zukor. We would also like to acknowledge
several of our colleagues who provided us with invaluable advice
when we were just beginning our studies on regeneration using
this remarkable animal, including Cliff Tabin, Mark Keating,
Jeremy Brockes, David Stocum, Roy Tassava, Panagiotis Tsonis,
and Anthony Mescher.
Newt Husbandry
25
References
1. Duellman WE, Trueb L (1986) Biology of
amphibians. McGraw-Hill Book Company,
New York, NY
2. Petranka JW (1996) Salamanders of the United
States and Canada. Smithsonian Institution
Press, Washington, DC
3. Spallanzani L (1769) An essay on animal reproductions. Translated from Italian by M. Maty.
Printed for T. Becket, and P. A. de Hondt, in
the Strand, London. Available through UMI
Books on Demand
4. Wallace
H
(1981)
Vertebrate
Limb
Regeneration. J Wiley and Sons Ltd., Toronto,
ON
5. Nye HL, Cameron JA, Chernoff EA, Stocum
DL (2003) Regeneration of the urodele limb: a
review. Dev Dyn 226:280294
6. Eguchi G, Eguchi Y, Nakamura K, Yadav MC,
Lillan JL, Tsonis PA (2011) Regenerative
capacity in newts is not altered by repeated
regeneration and ageing. Nat Commun 2:384.
doi:10.1038/ncomms1389
7. Eguchi G, Itoh Y (1982) Regeneration of the
lens as a phenomenon of cellular transdifferentiation: regulability of the differentiated state
of the vertebrate pigment epithelial cell. Trans
Ophthalmol Soc U K 3:380384
8. Oberpriller J, Oberpriller JC (1971) Mitosis in
adult newt ventricle. J Cell Biol 49:560563
9. Bettencourt-Dias M, Mittnacht S, Brockes JP
(2003) Heterogeneous proliferative potential
in regenerative adult newt cardiomyocytes.
J Cell Sci 116:40014009
10. Calve S, Odelberg SJ, Simon H-G (2010) A
transitional extracellular matrix instructs cell
behavior during muscle regeneration. Dev Biol
344: 259-271
11. Calve S and Simon H-G (2011) High resolution 3D imaging: Evidence for cell cycle reentry
in
regenerating
skeletal
muscle.
Developmental Dynamics 240:1233-1239
Chapter 3
Housing and Maintenance of Ambystoma mexicanum,
the Mexican Axolotl
Johanna E. Farkas and James R. Monaghan
Abstract
The aim of this paper is to assemble a significant amount of information on Ambystoma mexicanum, the
axolotl salamander, to assist in the basic knowledge needed to raise, breed, and study most aspects of axolotl biology. It is important to understand the basic biology of the axolotl in order to make informed decisions on their proper care and use in experiments. Therefore, we will provide necessary information to
the non-herpetologist that will assist in their study of this unique and fascinating animal. We also aim to
provide a resource on the general anatomy, behavior, and experimental tips specific to the Mexican axolotl
that will be of use to most axolotl laboratories. Axolotls have been actively researched since the 1860s,
giving testament to their relatively straightforward maintenance and their versatility as an animal model for
development and regeneration. Interest in using the axolotl in laboratory research has grown tremendously over the past decade, so dedicated resources to support the study of this species are needed and
encouraged.
Key words Salamander, Limb regeneration, Animal model, Axolotl anatomy
Introduction
1.1 Taxonomy,
Habitat,
and the Laboratory
Strain
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_3, Springer Science+Business Media New York 2015
27
28
Fig. 1 Image of female leucistic (left ), female albino (middle), and male wild-type
(right ) axolotls. Notice the large bellies of the two females indicating they are
filled with eggs and the large cloaca of the mature male wild-type
~40 km2 area of artificial canals just outside the city limits of
Mexico City [2]. This high-altitude lake system (~2,200 m above
sea level) has been inhabited for centuries, most notably by the
Aztecs. This is where the name axolotl originates, as the animal
was named after the Aztec god Xolotl [3]. The water conditions
of Lake Xochimilco between 1978 and 1988 were estimated to be
between 16 and 20 C, pH 7.48.0, with a conductivity between
975 and 1,650 microsiemens (S)/cm [1]. Today, axolotls are
critically endangered and are on the brink of extinction due to
habitat loss, the introduction of invasive species, and shifts in
water quality [4]. It has been estimated that densities were at
6,000 ind./km2 in 1998, 1,000 ind./km2 in 2000, and 100 ind./
km2 in 2008 [2], and only a few axolotls were cited in the lake
system after months of surveying in 2013 [5]. Unfortunately, the
majority of axolotls today are found in aquaria and laboratories
around the world.
The modern axolotl strain used in most laboratories is a highly
inbred population that most likely arose from a donation of seven
wild axolotls (six wild-type and one white mutant) between 1863
and 1866 to the Paris Natural History Museum [6]. In fact, most
modern-day laboratory axolotls likely have a direct lineage to these
founders, and all white mutants are descendants from this single
white animal [7]. A few wild-caught axolotls were introduced into
the colony strain in the 1960s including an albino tiger salamander
(Ambystoma tigrinum) [8], but overall the present-day laboratory
strain is likely one of the most long-running inbred strains of any
laboratory species. The 150-year history of laboratory breeding
seems to have selected against spontaneous metamorphosis (currently <1 % frequency), as it is more prevalent in wild strains than
the lab strain [7, 9]. The most extensive colony of laboratory axolotls is maintained at the Ambystoma Genetic Stock Center (AGSC)
at the University of Kentucky (www.ambystoma.org), which is a
continuation of the Indiana University Axolotl Colony initiated by
29
Humphrey R.R. in 1957 [10]. Over the past 50 years, the Axolotl
Colony has provided the majority of axolotls and housing information to labs worldwide. Valuable information on housing, breeding, and diseases of axolotls can be found at www.ambystoma.org
as well as in the archives of the Axolotl Newsletter.
1.2 Laboratory
Research Using
the Axolotl
1.3
Gross Anatomy
30
Fig. 2 Cartoons of the exterior (top) and interior (bottom) of an adult leucistic axolotl. Each organ is drawn to
approximate scale and represents the approximate position in the animal. Colors also represent the approximate color of each organ. The regenerating limb and tail represent mid-bud blastema stages. Internal histology
of a mid-bud blastema is represented in Fig. 3c
31
Aquatic and lacking a middle-ear structure, axolotls rely on olfaction far more than audition when they are searching for food. The
urodele olfactory system is quite complex and allows the animal to
discriminate between very similar odorants and chemicals. Because
their olfactory epithelium and olfactory bulb are large and easily
accessible, tiger salamanders were a favored model for the study of
olfaction and neuronal signaling during the 1970s and 1980s
[2224]. Axolotls possess true teeth and a calcified skeleton with
cartilaginous joints that are anatomically similar to mammalian
joints [25, 26].
Although their eyesight is poor and their vision is largely limited to the detection of movement, the axolotl retina displays the
layered structure typically seen in vertebrates. In fact, many early
studies of retinal intracellular signaling were conducted in salamanders because of their large and easy-to-access retinal cells [2729].
Tiger salamanders can detect ultraviolet light, and their UV-sensitive
photoreceptors express three different opsins [30, 31].
Furthermore, like fish and other aquatic amphibians, axolotls possess a lateral line system that is used to detect both electrical currents (via ampullary organs located on the head) and water
movement (via mechanoreceptive neuromasts that run along the
side of the animal). The lateral line develops from neurogenic placodes, and this developmental process may provide insights into the
evolution of vertebrate sensory systems [32]. However, the study
of the axolotls development and evolutionary history has been
complicated by its massive genome. Consisting of around 32 109
base pairs located across 14 haploid chromosomes [33], the axolotl
diploid genome is among the largest of all tetrapods. Our understanding of this animal is sure to increase rapidly as genetic technology advances and more techniques are adapted for use with
salamanders.
1.4 Structure
of the Circulatory
and Respiratory
System
32
blood loss, and clotting occurs very quickly even after injury to
major arteries. As a result, axolotls are at only minimal risk of
bleeding to death during and after surgery. They are a fairly popular model for the study of cardiac development because they can
exhibit a recessive mutation, dubbed c, that is cardiac lethal within
2 weeks after hatching. The hearts of c/c mutants do not beat, and
studies have found that these mutant hearts are largely differentiated
but lack tropomyosin [36] and organized sarcomeres [37].
Aquatic throughout their lives, axolotls utilize multiple strategies for obtaining oxygen from their environment. They exchange
oxygen with their three-paired external gills and can respire
through their skin via cutaneous gas exchange. However, axolotls
also possess rudimentary lungs and may obtain at least 4060 % of
their oxygen through surface breathing [38]. These lungs are elongated and translucent in appearance and run parallel to the spinal
column for virtually the entire length of the body cavity (Fig. 2).
Very little research on axolotl respiration has been conducted, but
previous studies of tiger salamander larvae have provided likely
insights into the mechanism of this process. These larvae inhale
using a two-stroke buccal pump system that mixes expired and
fresh air in the buccal cavity before pumping it into the lungs.
Exhalation is active and very rapid, minimizing the amount of fresh
air that is expelled through the mouth and gills. The ventilation
efficiency of this system is comparable to that of mammalian ventilation [39]. Axolotls housed in hypoxic tanks make frequent trips
to the surface to breathe [40], which can increase the probability
that they will swallow air and cause the formation of an air bubble
within the body that can disrupt the animals locomotion and feeding. Because of their numerous options for respiration, axolotls can
survive for hours outside of their tanks so long as they are not
allowed to desiccate. This ability proves useful for the purpose of
surgical procedures, as axolotls will heal very quickly if kept moist
and left sedated after surgery.
1.5 The
Immune System
33
34
Regeneration
Limb regeneration is the classic paradigm of complex tissue regeneration and is currently the primary focus of laboratory research on
the axolotl (Fig. 2). Axolotls regenerate limbs by generating a mass
of highly proliferative cells called a blastema at the distal tip of a
limb stump (Fig. 3c). After amputation, locally derived lineagerestricted progenitor cells or dedifferentiated cells proliferate and
recapitulate developmental processes in order to regrow the missing portion of the limb [59]. As long as animals are well cared for,
limb regeneration is a robust, dependable assay. A juvenile animal
(8.510 cm SVL) will reach the differentiation stage of regeneration around approximately 32 days post amputation but will not
replace 100 % of the missing limb for at least 100 days post amputation. Three-month-old axolotls (~5 cm SVL) will reach the differentiation stage of regeneration at approximately 22 days post
amputation and will replace 100 % of their limbs by approximately
66 days post amputation [12].
Perhaps because axolotls are a neotenic member of an evolutionarily primitive order, their brains are very simple and share
several similarities with the mammalian embryonic brain (Fig. 3d).
The axolotl brain is relatively flat and elongated with clear delineations between the telencephalon, mesencephalon, and rhombencephalon (Fig. 2). The olfactory bulbs are large, but the optic
lobes are small and poorly separated, and the cerebellum is relatively undersized and weakly developed as well [21]. The cerebellum contains the only neurons in the axolotl central nervous
system that are not paraventricularall other neurons remain stationary and do not migrate from the germinal site during development [60] (Fig. 3d). Though less is documented about the
terrestrial axolotl brain, it is known that the brain undergoes
35
Fig. 3 Histological images of axolotl tissues. (a) Massons trichrome histological staining of a juvenile axolotl
epidermis. Notice the large Leydig cells (L) throughout the epidermis, dermal tissue underneath the epidermis
(D), and muscle lying underneath the dermis (M). (b) A transmission electron micrograph of a Leydig cell in a
juvenile axolotl epidermis. Notice the nucleus in the middle (N), which is surrounded by rough endoplasmic
reticulum and a large number of dense vesicles (V). PM represents the Leydig cell plasma membrane. K represents a keratinocyte with N representing the nucleus of the keratinocyte. (c) Massons trichrome histological
staining of a juvenile mid-bud limb blastema (BL). The bone is indicated with a B. Notice the thickened epidermis on the distal tip of the blastema and mesenchymal cell that makes up the blastema. (d) Hematoxylin and
eosin histological staining of a juvenile axolotl brain. The cross section is taken through the posterior telencephalon (forebrain; see Fig. 2). Notice the ventricular zone that contains highly proliferative neural progenitor
cells [66]
36
Axolotls reproduce sexually and fertilize internally. Males lay spermatophores which are then taken up and dissolved by the female in
the spermatheca, allowing spermatozoa to be stored in the cloaca
until it is time for spawning to occur. As eggs leave the oviducts and
enter the cloacal chamber, spermatozoa come into contact with the
egg and more than one sperm enter the egg cytoplasm [69]. Eggs
are usually laid in strings and are protected by a thick coating of
sticky jelly, which must be removed prior to embryonic injections or
grafting experiments. Though spawnings vary considerably in size,
most females will lay between 200 and 1,000 eggs per spawn.
Breeding ease and efficiency in the laboratory peaks in the spring
and decreases throughout the summer, suggesting that axolotl
breeding is seasonal even in animals which have been removed from
37
2
2.1
Materials
Housing
1. A windowless (see Note 1) or blacked-out room held at a constant temperature of 1620 C (see Note 2).
2. An automatic light timer which controls lighting conditions
for a 12L:12D light cycle (see Note 3).
38
2.3
Breeding
2.4 Disease
Treatment
2.5
Shipping
2. Packing materials.
3. Thick, sealable plastic bags.
4. Disposable ice packs.
3
3.1
39
Methods
Housing
40
Breeding
3.4 Disease
Treatment
3.5
Shipping
41
Notes
1. Restricting sources of natural light will increase the chances of
successful mating taking place during the typically nonbreeding months.
2. Water temperatures higher than 22 C are stressful on the animals and can have long-term negative effects on axolotl health.
3. A dimly lit room is preferable to a bright one, as axolotls can
be stressed by bright lights.
4. Success is also possible at a lower salinity of 750 S/cm [76].
Thus, axolotls thrive in a range of salinity conditions.
5. Free-standing water, especially tap water, should be treated
with the water conditioners Amquel Plus and Novaqua Plus
(Kordon LLC).
6. Aquarium filters should be used, but make sure that the water
flow is kept to a minimum, as high flow is irritating to axolotls.
Long-term housing in these tanks is reliable without any substrate, but small gravel can be used to act as a host for nitrifying
bacteria and supplemented with 20 % crushed coral to facilitate
pH balance (Jessica Whited, pers. comm.).
7. The advantage of these systems is that the footprint is relatively
small and many of the daily tasks are automated and alarmed
over network including water changes, water conductivity, pH,
water flow, and temperature. Water-cooling is highly recommended with axolotls considering the heat that is generated by
the automated systems. Most modern buildings can leverage
process-cooling water, but if this is not available, air-cooled units
can be used for chilling as long as an exhaust vent for hot air is
built into the animal room. The downside to an automated system is that the water is shared between tanks so disease could
spread between animals. Therefore, diligent observation of animal health is critical when using an automated system.
8. Commercial brine shrimp hatcheries work very well for high
yields. If the axolotl colony is small, a simple brine shrimp
hatchery can be generated using a 500 mL glass bottle with a
bubbler set at the bottom placed next to a window. Hatching
solution should include 35 g sea salt/L and 1.5 g brine shrimp
cysts per 500 mL. Cysts will hatch after approximately 24 h
depending upon salinity, heat, light, and batch of artemia so
optimization will be needed for each lab.
42
43
Acknowledgments
We would like to thank the Ambystoma Genetic Stock Center for
the continued support and advice on axolotl maintenance (R24
OD010435). We would also like to thank Panagiotis Katsaros and
William Fowle for the electron microscopy images, Matthew
Nguyen and Pankhuri Singhal for the histological staining, and
Alex Sweeney and Jason Langshaw for the careful reading of the
manuscript.
References
1. Shaffer HB (1989) Natural history, ecology,
and evolution of the Mexican Axolotls.
Axolotl Newslett 18:511
2. Contreras V, Martnez-Meyer E, Valiente E,
Zambrano L (2009) Recent decline and potential distribution in the last remnant area of the
microendemic Mexican axolotl (Ambystoma
mexicanum). Biol Conserv 142:28812885
3. Smith HM (1969) The Mexican axolotl: some
misconceptions and problems. Bioscience 19:
593615
4. Zambrano L, Reidl PM, McKay J, Griffiths R,
Shaffer HB, Flores-Villela O, Parra-Olea G,
Wake DB (2010) Ambystoma mexicanum.
IUCN 2013. IUCN Red List of Threatened
Species. Version 2013.2
5. Press A (2014) Axolotl, Endangered Water
Monster, Sighted In Mexico after fears of
disappearance. 02/24/2014
6. Smith H (1989) Discovery of the axolotl and
its history in research. In: Armstrong JB,
Malacinski GM (eds) Developmental biology
of the axolotl. Oxford University Press,
New York, NY, pp 312
7. Malacinski GM (1978) The Mexican axolotl,
Ambystoma mexicanum: its biology and developmental genetics, and its autonomous celllethal genes. Am Zool 18:195206
8. Humphrey RR (1967) Albino axolotls from an
albino tiger salamander through hybridization.
J Hered 58:95101
9. Voss SR, Smith JJ (2005) Evolution of salamander life cycles: a major-effect quantitative
trait locus contributes to discrete and continuous variation for metamorphic timing. Genetics
170:275281
10. Humphrey RR (1979) The axolotl colony at
Indiana University. Axolotl Newslett 1:38
11. Voss SR, Epperlein HH, Tanaka EM (2009)
Ambystoma mexicanum, the axolotl: a versatile
amphibian model for regeneration, development,
and evolution studies. Cold Spring Harb Protoc
2009:pdb.emo128. doi:10.1101/pdb.emo128
12. Monaghan JR et al (2014) Experimentally
induced metamorphosis in axolotls reduces
regenerative rate and fidelity. Regeneration
1:214
13. R: A language and environment for statistical
computing
14. Zhang P, Wake DB (2009) Higher-level salamander relationships and divergence dates
inferred
from
complete
mitochondrial
genomes. Mol Phylogenet Evol 53:492508
15. Sandoval-Guzman T, Wang H, Khattak S,
Schuez M, Roensch K, Nacu E, Tazaki A,
Joven A, Tanaka EM, Simon A (2014)
Fundamental differences in dedifferentiation
and stem cell recruitment during skeletal muscle regeneration in two salamander species.
Cell Stem Cell 14:174187
16. Blassberg RA, Garza-Garcia A, Janmohamed
A, Gates PB, Brockes JP (2011) Functional
44
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
45
46
Chapter 4
Husbandry of Spanish Ribbed Newts (Pleurodeles waltl )
Alberto Joven, Matthew Kirkham, and Andrs Simon
Abstract
Research on urodele amphibians, such as newts, is constantly contributing to our understanding of fundamental
biological processes. In the present chapter, we present detailed husbandry protocols for the Spanish
ribbed newt (Pleurodeles waltl ). We describe the main phases of their life cycle, with emphasis on the
progressive development of sensory, motor, and integration systems, which lead to the acquisition of specific
stereotyped (and conditioned) behaviors. The methods are outlined to manage housing, feeding, handling, captive breeding, health monitoring, and euthanasia in this species under laboratory conditions.
With minor changes, these protocols can also be applied to other species of urodele amphibians commonly
used in laboratory research.
Key words Salamander, Housing, Environmental enrichment, Reproduction, Breeding, Larvae,
Development
Introduction
Urodele amphibians became well known by scientists in the past,
mostly due to their large embryos, the relatively easy manipulation,
and their external and slow development that can be modulated by
temperature [14]. All these facts together permitted to easily analyze the key stages during ontogenetic development, and as a
result, research in urodeles provided significant knowledge regarding the ontogeny of vertebrates [2, 59]. In addition, the absence
of rejection after transplant [2, 10], their extraordinary regenerative abilities [1115], the large size of their neurons and neuroendocrine cells [2], and the secondary simplification that characterizes
their nervous system [16, 17] have attracted the attention of generations of researchers. Since urodeles have become amenable to
modern molecular technologies, such as transgenesis and genome
editing, they provide reemerging animal models in particular for
studying development and regeneration [2, 15, 1831]. The
Spanish ribbed newt is a relatively large urodele species natural of
the Iberian Peninsula and Morocco that can reach 31 cm long,
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_4, Springer Science+Business Media New York 2015
47
48
Am
141
4244
Life history
Embryonic
stages
Initial larval
stages
3538
034
Pw
Peg stage,
e2D stage
Tailbud stage,
limb field stage
bud stage
Nv
(continued)
Table 1
Comparative depiction of developmental periods of commonly used salamander species based on external characters and physiological and behavioral key events
56, 56+
Juvenile,
adult
Eft, adult
Forelimb: 2D,
e3D, 3DI,
3DII, 4D
Hind limb: Limb
field, bud I,
bud II, bud III
Nv
57, 57+
4655c
Late active
5357
larval stages
Pw
3945
Am
Early active
4552
larval stages
Life history
Table 1
(continued)
51
and active larval life (in which limbs and complex behaviors
develop) finishing at metamorphosis. In the following paragraph,
more details of the different stages are given.
Throughout embryonic development (stages 034), in addition to the body plan formation, visual and olfactory primary
organ primordia develop, together with the mechanoreceptory
and chemoreceptory lateral line [41, 48]. The motor system also
appears, and basic stereotyped movement acquisition ends up in
swimming [4951]. Before hatching, at each side of the head a
balancer appears (a filament-like appendix used for remaining
adhered to different surfaces), followed by the development of
three branchial arches. During initial larval life (stages 3538),
behind the branching gills, the forelimb buds arise, and the visual
and lateral line systems mature along with the escape response
associated to visual and mechanical stimuli, although most of the
time the larvae remain steady, absorbing the remaining yolk
[26, 27, 5254]. By the end of this period, larvae start to eat by
stereotyped suction mechanism, driven also by visual and vibrational stimuli, but without active food searching. Consequently,
in the laboratory, live foods such as Artemia nauplii should be
provided. At this point, the active larval life begins and lasts about
3 months in Pleurodeles waltl, during which many morphological
variations occur related to key physiological and behavioral
changes [26, 27, 41, 45]. The active larval period can be further
divided into two phases: early active larval period (stages 3945,
when forelimbs develop) and late active larval period (stages
4655c, when hind limbs develop) [26, 27]. In the early active
larval period, the most remarkable external changes include the
loss of the balancers and forelimb development. In terms of hunting behavior, this period is driven by a refinement of the suction
technique, related to scouting the surroundings by first forelimbdriven walking steps together with head movements from side to
side, and the acquisition of lunging. During the late active larval
period, the hind limbs are formed, a major growth in the body
occurs, and the main structures present in the adult brain can be
recognized [26, 27, 41, 45]. From this period onwards, the
olfactory system start to be important for feeding, as active seeking followed by sniffing results in the capacity of predating on
immobile food such as commercial fish pellets or frozen food. In
addition, learning processes by classical conditioning can be
assessed during this period, as larvae show different behaviors
when raised in different conditions.
The present chapter describes husbandry protocols for successfully caring, breeding, and raising Pleurodeles waltl in the laboratory in a simplified manner with the aim of making the caretaker
job as easy and efficient as possible.
52
Materials
2.1 Tanks,
Containers, and
Environmental
Enrichment
Complements
2.3 Laboratory
Supplies
and Equipment
2.4 Reagents
and Solutions
53
Methods
3.1 Housing
Metamorphic/Adult
Newts
54
Fig. 1 Setups for Spanish ribbed newts and key features of the different developmental stages. (a) Aquarium
for juveniles and adults. (b) Adult newts. Note the difference in the shape of the body and the specialization of
the male forelimbs (arrowhead). (c) Detail of a Petri dish containing Pleurodeles embryos. Note the contaminated eggs that should be removed. (d) Representative stages of embryonic development, according to Gallien
and Durocher [41]. The arrowheads point to the blastopore at stage 11, the closure of the neural tube at stage
18, the eye protrusion at stages 20 and 23, and the balancer at stage 30; the gill primordia are encircled at
stage 30. (e) Box for housing initial and early active larvae. (f) Dorsal, lateral, and ventral views of late embryo,
hatchling, and initial larva; note the progressive development of the forelimb buds (small circles in dorsal
view), the development of the digestive system (big circles in ventral view), and the maintenance of the balancer (arrowheads). (g) Box for late active larvae. Observe the even distribution of environmental enrichment
objects. (h) Early and late active larvae, showing the appearance and development of the hind limbs (circles)
55
Fig. 2 Examples of housing systems for Spanish ribbed newts and advisable maximum number of animals that
should be housed together. (a) Hypothetical rack system. (b) Housing in Petri dishes, plastic boxes and aquaria
of different sizes
3.3 Handling
and Sexing Postmetamorphic/Adult
Pleurodeles waltl
1. Use a net to take a newt out of the tank (see Note 12).
2. Grip the newt by the base of the tail with one hand.
3. Observe the cloaca (see Note 13).
4. Place it calmly in the palm of your other hand.
5. Observe the forelimbs (see Note 14 and Fig. 1b).
Frozen
food
Free-living
food
Type
Mysis
Bloodworms
Cyclops
Grindal worms
(Enchytraeus sp.)
310
28
310
0.52
0.5 10 to
1 40
0.5 2 to
14
Pet shop/aquarist
Pet shop/aquarist
0.4
Size (mm)
Pet shop/online
Table 2
Adequate for
812 g
812 g
812 g
812 g
812 g
Can be used as a
supplement:
~500 worms
Can be used as a
supplement:
~500 worms
Inadequate
Quantity for
feeding 200 late
active larvae of
~0.2 g/larvae
1215 g
1215 g
1215 g
1215 g
Inadequate
Inadequate
Inadequate
Inadequate
56
Alberto Joven et al.
Pellets
Pet shop/online
(www.hikari.info)
Pet shop/online
(www.jbl.de)
Pet shop/online
(www.tetra.net)
Pet shop/online
(www.nlsfishfood.com)
Pet shop/online
(www.tropical.pl)
Pet shop/online (www.
aquatic-nature.com)
Pet shop/online
(www.tetra.net)
Pet shop/online
(www.tetra.net)
Pet shop/online
(www.nlsfishfood.com)
Powder Microvit
BASIC (70342)
Tropical energy food
S (TENF190)
Cichlid mini granules
(T506387-CE)
TetraMin granules
(T505322CE)
New Life
SPECTRUM
Community fish
formula
Tropical Spirulina
granulat (60434)
New Life
SPECTRUM
Amphibian formula
Hikari tropical sinking
wafers (21521)
Early and small late
active larvae
Late active larvae
Late active larvae and
small juveniles
Late active larvae and
small juveniles
0.40.8
13
Metamorphic
juveniles (previously
broken) and adults
Big adults
6 12
78
12
13
12
0.1
Inadequate
Inadequate
Inadequate
0.51 g
0.51 g
0.51 g
0.51 g
0.51 g
Inadequate
Inadequate
12 g
12 g
12 g
12 g
12 g
Inadequate
Inadequate
Inadequate
58
3.4 Cleaning
Post-metamorphic/
Adult Pleurodeles
waltl
3.5 Promoting
Natural Breeding
59
3. Observe the behavior of the newts (see Note 20) and look for
eggs in the tank.
4. Collect all the eggs from the breeding tank using a widemouthed plastic pipette (see Note 21), and place them in a
clean container (see Note 22).
3.6 Artificial
Fertilization
A daily careful inspection of the eggs is essential to prevent microbial growth. Remove any unfertilized or contaminated eggs (see
Fig. 1c), and change the medium (chlorine-free tap water or MHS)
every 24 days.
1. Fill up the new housing place with chlorine-free tap water or
20 % MHS.
2. If necessary, release the egg from the jelly (see Note 33) either
mechanically using no. 5 forceps or chemically by rinsing them
in a solution of 2 % cysteine in 20 % MHS at pH 8.0 for
25 min. In the latter case, several washes in freshwater/20 %
MHS must be done afterwards.
3. Follow the correct development of the embryos (Fig. 1d), and
separate those that show abnormalities.
4. After hatching, when larvae start swimming, monitor daily the
development of the embryos for free-living food to be supplemented at appropriate timing (see Note 34 and Fig. 1f).
60
The big larvae can be easily collected using a net, but embryos and
small larvae are more fragile and can be easily killed when trying to
pick them up with a net. Use Pasteur pipette as described below.
1. Cut the tip of a plastic Pasteur pipette to have an opening
larger than the size of the larvae to be transferred.
2. Calmly approach the larva with the widemouthed Pasteur
pipette.
3. Capture the larva by a quick vacuum movement.
4. Use your finger tip to close the opening of the pipette
(see Note 47).
5. Release the larva into its new location.
61
3.12 Health
Inspection and
Treatment of Diseases
62
the plants with new ones, and treat all the tank mates as
described in steps 2 and 3. Refill the tank with chlorine-free
water and add antibiotic/antimycotic solution.
7. Every 2 or 3 days, change one half of the volume of the tank.
8. Stop the treatment when the animals look healthy, 2 days after
the signs disappear.
3.13
Euthanasia
Notes
1. During the periods of extreme temperatures, Pleurodeles waltl
may undergo hibernation and aestivation. During these processes, the skin becomes thicker and less permeable; Animals
decrease food intake and slow down the metabolic rate.
Nevertheless, Spanish ribbed newts have been found all along
the year having an active lifestyle in the water [3236].
2. Natural populations experience slight variations along the year
in the light-dark (LD) cycle which approximately ranges from
(LD 15:9) in summer to the opposite in winter (LD 9:15).
They can be kept in a (LD 12:12) in laboratory conditions,
although placing the newts under the influence of natural light
(i.e., close to a window) or alternatively adapting the timer of
the animal facility trying to mimic the natural LD cycle (15 min
of light/week) may benefit the adult newts and induce the
natural breeding at the corresponding time of the year.
3. The Spanish ribbed newt has a high temperature tolerance. In
nature, this species survives in regions in which absolute temperatures range from bellow 10 C to over 40 C. In the
laboratory, the stable temperature prevents most of the newts
undergoing hibernation and aestivation. Nevertheless, some
animals may decide to spend several days/weeks in the floating surfaces, eating very little, moving slowly, and showing
63
64
65
66
67
43. During the initial larval period, hatchlings live on their remaining
stalk; thus, no feeding is necessary. Once the bellies start to
lose their whitish-yellow appearance of the stalk, the initial
larval period is about to start, and the first meal (free-living
food) should be added. In the early active larval period, larvae
start to prey over small live food, which is recognized usually
by a combination of visual and vibrational cues. They do not
seek for food at these stages, but prey only over the small creatures that pass by their heads. Cannibalistic behaviors become
frequent; thus, it is recommendable to keep them at low densities and ad libitum conditions. At the beginning of the late
active larval period, larvae start to use odor cues to discriminate also steady food, and learning processes modulate food
intake. As a consequence, cannibalistic individuals should be
isolated if detected. Once cannibalism is solved by housing
larvae at appropriate densities, newts can learn that dry pellets
are a source of food. The pellet size after hydration should be
slightly smaller than the mouth of the animal.
44. For small and medium active larvae (stages 4554), cut the
frozen pellets as small as you can before the tablet gets defrost
(also the Tubifex). Mix it in freshwater and filter it.
45. Cut the tip of the wash bottle to increase the flow and to avoid
that food gets stuck inside the outflow tube.
46. The food tends to sink; thus, shake the wash bottle before adding the food into every tank.
47. Closing the opening of the pipette will prevent the larvae to
escape from the pipette and end up on the floor or another dry
surface. If they escape, try to save the larvae using a net or a
wet piece of paper. Avoid catching them with the fingers or
forceps.
48. If the tank is equipped with aeration, partial cleaning can be
done every second day (see Subheading 3.4), followed by total
cleaning after the next 2 days. Do not vacuum the larvae, and
always filter the discarded water through a net before disposal
to ensure that no larvae are being lost in the process.
49. Try to avoid touching the new clean water with the net;
instead, let the larvae gently drop down at a short distance of
the water surface. This procedure will prevent transfer of any
debris from the net to the water.
50. The Caudata Culture website (www.caudata.org) provides
information on the common symptoms and diseases in urodeles [58].
51. You may need to contact the university veterinarian for the
treatment.
68
Acknowledgments
This work could not have been possible without the opportunities
of working with various salamander species along the years, under
different group leaders: Paschalina Kiriakopoulou-Sklavounou
(AUTH, Greece), Agustin Gonzlez (UCM, Spain), and Elly M.
Tanaka (MPI-CBG, Germany). Also, many thanks to all the people
ever involved in the caring of the different colonies and for interesting conversations that resulted in improvements of the protocols, with special thanks to Heino Andreas, Shahul Hameed, Heng
Wang, Laure Belnoue, Tiago Pinheiro, Ivanna Mayorenko, Jorge
Perlado, Patricia Fernndez, Joana Branco, Laura Domnguez,
Sandra Bandn, Ruth Morona, and Nerea Moreno. This work was
supported by a postdoctoral fellowship to A.J. from the WennerGren-Foundation and by the Swedish Research Council to A.S.
References
1. Duprat AM, Gualandris L, Kan P, Pituello F,
Saint-Jeannet JP, Boudannaoui S (1986)
Archives danatomie microscopique et de morphologie exprimentale. Arch Anat Microsc
Morphol Exp 75:211227
2. Eagleson GW (1996) Developmental neurobiology of the anterior areas in amphibians: urodele perspectives. Int J Dev Biol 40:735743
3. Nieuwkoop PD (1996) What are the key
advantages and disadvantages of urodele species compared to anurans as a model system for
experimental analysis of early development? Int
J Dev Biol 40:617619
4. Gualandris-Parisot L, Husson D, Foulquier F,
Kan P, Davet J, Aimar C, Dournon C, Duprat
AM (2001) Pleurodeles waltl, amphibian,
Urodele, is a suitable biological model for
embryological and physiological space experiments on a vertebrate. Adv Space Res 28:
569578
5. Spemann H (1921) Die Erzeugung tierischer
Chimren durch heteroplastische embryonale
Transplantation zwischen Triton cristatus und
taeniatus. Wilhelm Roux Arch Entwickl Mech
Org 120:374706
6. Spemann H, Mangold H (1924) ber
Induktion von Embryonanlage durch implantation artfremder Organisatoren. Wilhelm
Roux Arch Entwickl Mech Org 100:599638
7. Holtfreter J (1933) Der Einfluss von
Wirtsalter und verschiedenen Orgenbezirken
auf die Differenzierung von angelagertem
Gastrulaektoderm. Wilhelm Roux Arch
Entwickl Mech Org 127:610775
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
69
70
52.
53.
54.
55.
56.
57.
58.
Chapter 5
Maintaining Plethodontid Salamanders in the Laboratory
for Regeneration Studies
Claudia Marcela Arenas, Andrea Gmez-Molina, and Jean Paul Delgado
Abstract
Limb regeneration studies have been extensively carried out in species of Ambystomatidae and
Salamandridae families. So far limited research has been conducted in species belonging to the
Plethodontidae family, where some of the species differs from other salamander families due to their direct
development, thus absence of a larval life. Here, we describe a protocol to maintain the plethodontid
salamanders of genus Bolitoglossa species under laboratory conditions to perform regeneration studies.
Key words Plethodontidae, Bolitoglossa sp., Husbandry, Limb regeneration
Introduction
Ambystomatidae and Salamandridae family species are the most
common salamanders that have been used as models in the development and regenerative biology research [1, 2]. There are about
668 species of salamanders that are distributed in ten families [3].
In most of the species, the biological growth is termed indirect
development, where a larval stage occurs during development and
an ontogenic transition is attained by a process of metamorphosis.
However, in the family Plethodontidae, some species have direct
development [4]. Besides this, they also show some biological
adaptive differences such as anucleate red blood cells, projectile
tongue, tail autotomy, and the absence of lungs [5, 6].
South American salamanders are represented only by the family Plethodontidae [7], mainly by the genus Bolitoglossa, and about
28 species have been recognized [8]. Bolitoglossa species undergo
direct development; they are terrestrial animals distributed in
diverse habitats and elevations, from high-elevation grasslands to
lowland rainforest [9].
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_5, Springer Science+Business Media New York 2015
71
72
Materials
2.2
Reagents
73
Fig. 2 Plastic container to maintain the salamanders in captivity (a) damp paper
tissues to keep the humidity. (b) Environment enrichment with objects where the
animal can climb, hide, or explore
Methods
3.1 Collection
of Salamanders
1. The salamanders are collected by the nighttime visual encounter method [11], which is based in the visual registration of the
animals in a limited time and area.
2. Once the animal is visualized, collect it carefully with both
hands, avoiding taking it by the tail that would lead to caudal
autotomy.
3. The specimens are kept in plastic bags and sprayed with filtered
tap water, and local vegetation is placed inside the bags to prevent desiccation.
74
4. Keep the plastic bags inside a polystyrene ice box with an ice
pack to maintain low temperature and humidity. Avoid direct
contact of the animal with the ice pack.
3.2 Housing
and Environmental
Conditions
1. The animals are placed inside the plastic container (see Note 6).
2. Maintain housing temperature at 1921 C throughout the
period.
3. Use a 12L:12D photoperiodic cycle for the animals.
4. To keep the humidity inside the container, place two layers of
tissue paper and spray them with filtered tap water (see Note 7).
5. Place environmental enrichment objects inside the container
to provide the animals with place to hide and to climb.
6. Replace the damp tissues and wash the container with filtered
tap water twice a week (see Note 8).
3.3
Feeding
1. Place the flask with flies inside the freezer 2030 min
(see Note 9).
2. Place about 20 flies and or larvae per each animal inside the
container.
3. Feed the animals three times a week.
Behavior
Plethodontids move slower and are more docile than newts or axolotls. When handled for long period of time, they can become
stressed; therefore, handle them only when necessary (see Note 10).
In the wild, they can be found in solitary, but in captivity, they
can be kept with other plethodontids in the same container.
3.5 Common
Diseases and Health
Monitoring
3.6 Surgical
Manipulation
3.4
75
Notes
1. In the Andean Community Nations, the capture of wild animals is tightly regulated by the Department of Environment of
the respective country. For experimental studies requiring wild
caught salamanders, a specific license is required to collect the
animals. It is important to know the local regulations regarding the collection of wild animals in each country and, if
required, obtain the respective permits.
2. The plastic containers can be a kitchenware type with rubber
seal. It is important to place items that enrich the environment
so that the animal can hide from the light. Besides this, sticks
or artificial plants can help to increase the area where the animal can climb or explore (Fig. 3).
3. The quality of tap water in our city has a pH of 6.4, which
appears to be easily tolerated by the animals; only filtration is
needed it to remove the chlorine. A standard kitchen carbon
filter is sufficient for this purpose. Another option is to allow
chlorine evaporation overnight.
4. When you collect wild animals, it is important to standardize
the amount of food that is needed to keep the initial weight of
the animal. To do this, measure the weight of the animals once
a week. In our experience, 50 flies and/or larvae a week are
enough to keep the weight of these species. An important
aspect to take into account is that the animals need to hunt for
flies when they are alive, the animals do not eat dead flies.
5. It is critical to adjust the pH of the anesthetic before use; use a
buffered solution (Holtfreters salt solution) and adjust the pH
76
Fig. 3 Limb regeneration Bolitoglossa ramosi. Note the regeneration blastema on the left limb
to 7.4. When a low pH is used, animals could experience bleeding from the skin.
6. Handle the salamanders with gloves to avoid transmission of
any diseases between animals. In order to avoid stress and
induction of tail autotomy, handle the animals with a pencil,
brush, or stick. Because they like to hang on sticks by using the
prehensile tail, reach the tip of the pen or stick to the animal
and allow to climb on it (Fig. 4).
7. The quality of tissue paper is important because some brands
available from the market are acidic and, in contact with water,
will decrease the pH of water. In our experience, the recycled
tissue paper works fine.
8. It is important to keep the plastic containers with low microbial density to prevent diseases in the animals. However,
amphibians are reported to have antimicrobial peptides that
could help them to prevent diseases [15].
9. Flies can be obtained from genetic laboratory facilities.
Otherwise, insects can be purchased from local pet suppliers.
77
Acknowledgments
Our work is funded by a program grant COLCIENCIAS 569 and
Programa-Sostenibilidad 20132014 of the University of
Antioquia. Claudia Arenas is funded by a fellowship from
COLCIENCIAS 567. We thank Juan Manuel Daza for the taxonomic identification of the animals and Melisa Hincapie, Carlos
Muoz, Jose Fang, and Santiago Cuartas for their help during the
field work and maintenance of the animals. The animals used to
establish this protocol were collected under the environmental
78
9.
10.
11.
12.
13.
14.
15.
Part II
Experimental Manipulation in Salamanders
Chapter 6
Newt Lens Transdifferentiation: From Lentectomy
to Immuno-FISH
Nobuyasu Maki
Abstract
Newt lens regeneration is achieved by a unique cellular regulation of transdifferentiation where the dorsal
iris pigmented epithelial cells (PECs) dedifferentiate and redifferentiate into lens cells. Recent studies have
shown that nuclear architecture of PECs is dynamically changed and unique epigenetic regulation in
somatic nucleus is crucial in the lens transdifferentiation. Immuno-FISH, detection of protein and gene
loci in nucleus, is one of the effective tools to analyze nuclear architecture of PECs. In this chapter a whole
process from lentectomy to immuno-FISH is described.
Key words Lens regeneration, Transdifferentiation, Dedifferentiation, Epigenetics, Reprogramming,
Salamander, Immuno-FISH
Introduction
It is a generalized conception that differentiation is an irreversible
event and once cells are terminally differentiated, they lose their
multipotency and proliferation ability so that the differentiated
cells never change into other types of cells in vivo. An apparent
counterexample to this concept is transdifferentiation of iris pigmented epithelial cells (PECs) during newt lens regeneration
(Fig. 1) [1]. After lens removal, the PECs lose their original characteristics by shedding their pigment granules (about day 4).
At almost the same time, an initial cell cycle reentry of PECs occurs.
PECs continue depigmentation and proliferation and finally change
to transparent cells, called as dedifferentiated cells, by day 8 and
form a lens vesicle by day 1012. After day 14, lens differentiation
occurs from dorsal iris. Even though the dedifferentiation and proliferation take place in both dorsal and ventral irises, lens differentiation occurs only in dorsal iris. Lens transdifferentiation has been
demonstrated by clonal culture experiment using newt PECs [1].
Nuclear architecture of PECs dynamically changes during
transdifferentiation [13]. The original PECs have a small and
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_6, Springer Science+Business Media New York 2015
81
82
Nobuyasu Maki
Fig. 1 Illustration of lens regeneration process and changes in nuclear architecture. (a) About 4 days after
lentectomy, PECs begin to shed their pigments and reenter cell cycle. The transparent dedifferentiated cells are
found by day 8. After day 14, the dorsal dedifferentiated cells redifferentiate into lens cells. However, the ventral PECs never differentiate into lens even though these cells show depigmentation and cell cycle reentry. (b)
Structural change in the nucleus during lens transdifferentiation. The nucleus of original PECs is small and
shrunken in morphology. During dedifferentiation, the PEC nucleus swells and its nucleoli become large. Blue,
nucleus. Red, nucleolus. Illustration in (b) is reproduced from [9] with permission (Color figure online)
83
Fig. 2 Immuno-FISH using NS antibody and 18S rDNA probe against PECs before (a) and 6 days after (b)
lentectomy. NS is accumulated in nucleoli of dedifferentiating PECs. Nuclei were counterstained with Hoechst
33258. High-power view of the nucleolus indicated by arrowheads is shown in the inset. Outline of nucleus is
indicated by dashed line. Reproduced with permission from Developmental Dynamics [2]
Materials
2.1 Animal
Maintenance
2.2
Lentectomy
84
Nobuyasu Maki
Fig. 3 Process of lentectomy. (a) The special forceps for lentectomy. The tip of the
forceps is rounded and smoothen with a whetstone. Upper, side view. Lower, top
view. (b) Newt held between the thumb and forefinger. (c) Illustration of the process of lentectomy. Incision of the cornea (13). Holding the lens with forceps
(4 ). Removing of lens (5)
2.3 Sampling
and Tissue Sectioning
85
2.4
Probe Labeling
2.5
FISH Procedure
86
Nobuyasu Maki
Methods
3.1 Animal
Maintenance
3.2
Lentectomy
1. Keep newts in the anesthetic solution until they are anesthetized (about 10 min).
2. Hold the newt neck between the thumb and forefinger.
3. Put the surgical blade into the cornea with the edge side up
(Fig. 3c-1).
4. Carefully insert the blade between the cornea and iris until the
tip of the blade reaches the opposite side (Fig. 3c-2).
5. Incision the cornea by drawing the blade upward. About
two-thirds of the cornea diameter is cut in this operation
(Fig. 3c-3).
6. Insert the forceps through the opening of the cornea (Fig. 3c-4,
see Note 3).
7. Hold the surface of the lens with the forceps (see Note 4).
8. Carefully pull the lens out from the eyeball (Fig. 3c-5).
9. Return the newts to the rearing water. About 1020 min after
returning to the water, newts begin to awake from anesthesia.
3.3 Sampling
and Tissue Sectioning
87
Probe Labeling
3.5
Immuno-FISH
88
Nobuyasu Maki
Notes
1. During lentectomy, a lens epithelial cell layer is held with forceps. To hold it firmly and to avoid tissue damage, the tip of
the forceps should not be sharp. With a whetstone, the tip of
the forceps (e.g., INOX No. 5) is rounded and smoothen
(Fig. 3a). When the forceps is closed, both ends of the tips
should be attached together.
2. To insert a surgical blade into the eyeball through the cornea,
the tip of the blade should be sharp. For this purpose Feather
surgical blade #11 is suitable (the blade which has a rounded
tip such as Kai #11 cannot be used).
3. Lens can be extruded by pushing the eyeball with forceps.
However, this method may cause a damage on the iris. So the
method presented in this chapter is highly recommended.
Because of its transparency, the position of the lens in the eyeball
is visually indistinguishable. Thus, the position of the lens must
be realized based on the structure of the newt eye during the
operation (see Fig. 3). Too much insertion of the forceps causes
lens surface damage and makes it difficult to hold the lens.
89
4. At first, touch the eyeball with both tips of the forceps, and
then close the forceps very carefully without any other
movement.
5. The lens-regenerating eye is very fragile. To avoid tissue damage during its collection, eyeballs are fixed before dissection.
Because a paraformaldehyde fixation causes strong signal
reduction in newt FISH and immunohistochemical analysis, a
methanolacetic acid (3:1) solution is used.
6. This opening in the ventral side is necessary to avoid a crush of
eyeballs during the xyleneparaffin exchange and serve as a
mark indicating a dorsoventral orientation in sections.
7. The paraffin-embedded samples can be stored at 4 C.
8. After drying the slides can be stored at 4 C.
9. This probe solution can be stored at 20 C for several months.
10. Because of the pigment granules in PECs, this treatment is
performed.
11. To obtain FISH signals, the optimum temperature is
70 C. However, to retain an antigenicity of NS, this denaturing treatment is performed at 60 C. Denaturing condition
must be determined for each antigen.
12. Because the 2 hybridization solution is viscous, widening the
tip by cutting it is recommended.
Acknowledgments
This work is supported by JST PRESTO and a grant, KAKENHI
(14514364), to N.M. I would like to thank Panagiotis A. Tsonis for
critical reading and Rinako Maki for producing the illustrations.
References
1. Maki N, Kimura H (2013) Epigenetics and
regeneration. Curr Top Microbiol Immunol
367:237252
2. Maki N et al (2007) Rapid accumulation of
nucleostemin in nucleolus during newt regeneration. Dev Dyn 236:941950
3. Maki N, Tsonis PA, Agata K (2010) Changes in
global histone modifications during dedifferentiation in newt lens regeneration. Mol Vis
16:18931897
4. Jullien J et al (2010) Characterization of somatic
cell nuclear reprogramming by oocytes in which
a linker histone is required for pluripotency gene
reactivation. Proc Natl Acad Sci U S A 107:
54835488
5. Takahashi K, Yamanaka S (2006) Induction of
pluripotent stem cells from mouse embryonic
6.
7.
8.
9.
Chapter 7
Studying Newt Brain Regeneration Following Subtype
Specific Neuronal Ablation
Matthew Kirkham and Alberto Joven
Abstract
The realization that neuronal injury does not result in permanent functional or cellular loss in all
vertebrates has fascinated regenerative biologists. Neuronal regeneration occurs in a subset of species,
including lizards, teleost fish, axolotls, and newts. One tool for studying neuronal regeneration in the
adult brain is intraventricular injection of selective neuronal toxins, which leads to loss of subpopulations
of neurons. To trace cells involved in the regeneration process, plasmids encoding reporter proteins can
be electroporated in vivo into the cells of interest. This protocol describes methods to label the ependymoglial cells of the brain of the red spotted newt Notophthalmus viridescens and follow their response after
ablation of dopaminergic neurons.
Key words Neurogenesis, In vivo electroporation, 6-OHDA, Dopaminergic neurons, Neuronal
regeneration
Introduction
A resurgence of interest in vertebrate brain regeneration has
occurred over recent years, which has coincided with increased
understanding of adult neurogenesis and stem cell biology within
mammals [1]. The mammalian brain regenerates poorly after neuronal loss, but the persistent presence of neural stem cells and
ongoing adult neurogenesis has offered hope that cell replacement treatments for neural injury and disease could be developed
either via transplantation of cells or endogenous regeneration [2].
Hence, an increased understanding of how successful neuronal
regeneration occurs could lead to improvements in cell regenerative therapies in humans.
Injury in adult neuronal regenerative models has been caused
through two main methods. Mechanical injuries for example have
included the removal of part of the optic tectum in Notophthalmus
viridescens, severing the retinal axon in the lizard Gallotia galloti,
cerebellar lesion paradigm in teleost fish and traumatic stab lesion
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_7, Springer Science+Business Media New York 2015
91
92
93
Fig. 1 Illustrations of the applications and equipment needed for intraventricular injection. (a and b) Midbrain
dopaminergic neurons in a control newt, Notophthalmus viridescens (a) and 5 days after injection of 6-OHDA
(b). The neurons are labeled with an antibody against Tyrosine hydroxylase (red ) and the nuclei are labeled
with the DNA dye DAPI (blue). (c and d) GFAP-positive ependymoglial cells (red) lining the third ventricle of a
newt that has been electroporated with a plasmid encoding GFP (green) under a CMV promoter. (e) Overview
of the equipment. (1) Stereomicroscope on bloom arm, (2) FemtoJet, (3) Stereotactic instrument, (4) Quick
drying dental cement, and (5) Micro drill. (f) The universal capillary holder arm of the FemtoJet is attached to
stereotactic instrument using a V clamp. (g) Glass needle in the universal capillary holder ready for injection.
The barrel of the needle is marked with a marker pen. Scale bars 50 M
94
Materials
2.1 Equipment
for intraventricular
Injection
or
micro-drill
maximum
speed
2.2 Reagents
for intraventricular
Injection
95
1. Tank water. This can either be tap water or the salt solution
used to house the animals.
2. 0.1 % MS-222 (Sigma). Dissolve 1 g MS-222 in 1,000 mL of
tank water.
3. Quick drying dental cement (Harvard Dental).
2.3 Equipment
for In Vivo
Electroporation
2.4 Reagents
Required for In Vivo
Electroporation
2.5 Reagents
for 6-OHDA Neuronal
Ablation
Methods
3.1 Intraventricular
Injection of Plasmids
96
1. Check that the electroporator is set up correctly and the electrodes are clean. The newt is electroporated in unidirectional
manner at 150 mV/cm pore forming pulse and 110 mV/cm
transfer pulse with a decay of 5 % between the pulses. The newt
head is approximately 0.9 cm in width. To electroporate horizontally the machine is set to give one pore forming pulse at
135 mV 50 ms on 950 ms off, followed by five 50 ms pulses at
99 mV with 950 ms gap between each pulse.
2. Place the plastic support holding the newt on the bench. Dip
the electrodes and the head of the newt into tank water. Place
a lid of a 5 cm petri dish on top of the body of the newt to
secure it during the electroporation.
3. Use the hole in the skull as a guide to position the electrodes
on the side of the newt head, so that the current will pass
through this region of the brain. Once the head is gripped
firmly with the electrodes, start the electroporator.
4. Mix the fast drying cement and cover the drilled hole. The
cement takes about 10 min to dry.
5. Allow the newt to recover in tap water or tank solution. Place
the newt in a container with shallow water (15 cm in depth).
97
Notes
1. The micropipette puller is used to manufacture glass needles.
Mark the barrel of the needles every 2 mm with a marker pen.
These marks can be used to gauge the volume of the fluid
injected. The glass capillaries from Harvard Apparatus reference GC100F-10 have an inner diameter of 0.58 mm. Thus,
the volume of fluid held in 1 mm of the barrel part of the
needle is equal approximately to 200300 nL.
2. The purity of the DNA is critical to the success of the electroporation. Plasmid preparation using other kits can have reduced
efficiency and may have contaminates which can kill the ependymoglial cells.
3. Plasmid DNA with a concentration above 5 mg/mL can be
very viscous and difficult to pipette. Patience is required, but
the electroporation efficiency is substantially increased with
high DNA concentration.
98
4. Once the needle is loaded test to see if any fluid comes out.
If not, first check that the tubing is connected correctly. Next,
try increasing the injection pressure and thirdly use scissors to
cut the tip increasing the gauge of the needle. The injection
pressure is dependent on how viscous the solution is and the
gauge of the needle and can range from 100 to 2,000 hPa. The
fluid should come out at a slow and steady rate, and to inject
500 nL can take up to 30 s.
5. To drill the hole in the skull maintain a continuous rpm and
gently press the drill tip down on to the skull. Raise the drill tip
at 12 s intervals to check the progress of the drilling.
6. The penetrance to the meninges is determined by the size and
shape of the needle. Try several small sharp movements up and
down with the needle. If this does not work, try a different
needle. Care is needed not to break the needle at this point.
7. In some cases neuronal ablation is performed without electroporation. If this is the case, a hole is drilled in the skull as
described in Subheading 3.1.
Acknowledgement
This work was supported by a grant from the Swedish Research
Council to MK.
References
1. Tanaka EM, Ferretti P (2009) Considering the
evolution of regeneration in the central nervous system. Nat Rev Neurosci 10:713723
2. Kokaia Z, Martino G, Schwartz M, Lindvall O
(2012) Cross-talk between neural stem cells
and immune cells: the key to better brain
repair? Nat Neurosci 15:10781087
3. Zupanc GKH, Srbulescu RF (2011) Adult
neurogenesis and neuronal regeneration in the
central nervous system of teleost fish. Eur J
Neurosci 34:917929
4. Lang DM, Monzn-Mayor M, Bandtlow CE,
Stuermer CA (1998) Retinal axon regeneration in the lizard Gallotia galloti in the presence of CNS myelin and oligodendrocytes.
Glia 23:6174
5. Okamoto M, Ohsawa H, Hayashi T et al
(2007) Regeneration of retinotectal projections after optic tectum removal in adult newts.
Mol Vis 13:21122118
6. Kroehne V, Freudenreich D, Hans S et al
(2011) Regeneration of the adult zebrafish
7.
8.
9.
10.
11.
99
Chapter 8
The Accessory Limb Model: An Alternative Experimental
System of Limb Regeneration
Tetsuya Endo, David M. Gardiner, Aki Makanae, and Akira Satoh
Abstract
Accessory limb model (ALM) was developed as an experimental model and functional assay for limb
regeneration. The ALM provides several ways to identify pathways and test for signaling molecules that
regulate limb regeneration. Here, we summarize the history of the ALM and describe the specific details
involved in inducing ectopic blastemas and limbs from a skin wound on the side of the arm.
Key words Salamander, Axolotl, Nerve, Limb regeneration, Slow-release sustained bead
Introduction
Limb regeneration is one of the impressive biological phenomena
that have been studied in two urodele amphibians, axolotls and
newts. Limb amputation results in formation of a regenerationspecific structure called the blastema, which consists of undifferentiated cells. Blastema cells are derived from stump tissues and
appear to be lineage restricted such that regenerated muscle is
derived from myogenic progenitors, and connective tissue cells are
derived from cells in the connective tissues of the stump. Once a
blastema has formed, it starts restoring the original structure [1].
It is the early stages of blastema induction that are unique to limb
regeneration. Therefore, understanding early phases of limb regeneration that lead to blastema formation is important in order to
understand the regeneration potency in axolotls.
Historically, studies of limb regeneration were based on an
amputation model, and the progression through the stages of blastema formation, growth, and differentiation was described.
However, amputation causes wide-ranging injuries to the tissues at
the amputation plane, and it is not possible to determine which of
the multitude of responses that are triggered are required to induce
limb regeneration. In addition, since regeneration is a default
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_8, Springer Science+Business Media New York 2015
101
102
103
Materials
2.1 Obtaining
Axolotls
and Husbandry
104
Fig. 1 The axolotl and the anatomy of the forelimb. (ac) A juvenile leucistic axolotl. Front view (a) and dorsal
view (b). (c) Left side view of the animal at the forelimb level. The left forelimb is seen from the anterior side.
(d) The anatomy of the left forelimb at the middle of the forearm level as it would appear in transverse section
at the level of the black circle indicated in (c) (modified from [20])
limb relatively fast and are easy to handle. Leucistic and albino
mutants are useful since it is easier to visualize tissues through
their unpigmented skin.
2.2 Axolotl
Maintenance
and Surgery
2.3
Bead Preparation
105
Methods
The procedure described here induces an accessory limb on
the anterior side of the left upper arm (for limb orientation, see
Fig. 1c, d). An accessory limb can be induced at other proximaldistal levels or from any position around the circumference of the
limb with appropriate modification of the procedure. Overview of
the ALM surgeries and resulting phenotypes are shown in
Fig. 2ad.
Fig. 2 Induction of an accessory limb in axolotls. (a) The surgical procedure that induces an ectopic blastema
from an anterior skin wound by deviating a nerve. (b) An induced blastema by the (a) surgery. (c) The surgical
procedure that induces an accessory limb from an anterior skin wound by deviating a nerve and grafting
posterior skin. (d) An accessory limb induced by the (c) surgery. Host limbs in (a)(c) and the accessory limb
in (d) are seen from the ventral side
106
3.1
Skin Wounding
3.2
Nerve Deviation
1. Raise up the left arm so that the ventral side of the limb is up
(Fig. 3c). If the animals are leucistic or albino, two flexor nerve
bundles are visible through the skin (see Fig. 1d).
2. Make an incision on the ventroposterior side from the shoulder level to the elbow level (Fig. 3d).
3. Bisect the flexor nerves distally (at the elbow level).
4. Pull out the free tip of the nerves carefully with forceps (Fig. 3e).
Pay attention not to rupture the ventral brachial artery that is
attached to the flexor nerves by connective tissues.
5. Pass the cut end of the nerve underneath the ventroanterior
skin to the wound (Fig. 3f). Fix the skin back to the original
position to close the ventroposterior incision.
6. Turn the left arm back to see the anterior wound. Position the
cut edge of the nerve in the middle of the wound along
the transverse plane of the limb (Fig. 3g). Trim the distal tip of the
nerve, if it is too long.
7. These procedures (Subheadings 3.1 and 3.2) are sufficient to
induce a blastema (Fig. 2a, b) but insufficient to induce a limb.
To obtain a patterned limb, steps described in Subheading 3.3
are necessary.
3.3
Skin Grafting
1. To obtain a piece of skin for grafting, turn the animal left side
down.
2. Hold the right arm up so that the posterior side of the limb
is visible.
107
Fig. 3 A series of pictures of the surgery that induces an accessory limb. (a) For
left forelimb surgery, the animal is laid down, turning the left side up. A thick blood
vessel is visible on the anterior side of the forearm. (b) A square skin wound is
made on the anterior side of the middle of the forearm. (c) The limb is turned up
to expose the ventral side. (d) An incision is made in the ventroposterior skin to
access the ventral nerve bundle. (e) The ventral nerve bundle is cut at the elbow
level and pulled free of the connective tissues. (f) The cut end of the nerve bundle
is passed underneath the ventroanterior skin to the wound. (g) The limb is turned
down to see the anterior wound. The cut end of the nerve bundle is trimmed and
placed in the middle of the wound. (h) A piece of skin from the opposite side of the
limb (posterior skin is grafted to the anterior wound) is removed from the contralateral limb and grafted to the skin wound. g grafted skin
108
109
Fig. 4 Steps involved in making gelatin beads. (a) Gelatin solution is poured into warmed olive oil. (b) Acetone
is added into the gelatin-oil emulsion. (c) Beads are washed by isopropanol. (d) Beads can be observed in the
bottom of the tubes
13. Add deionized water and wash the beads by inverting few
times.
14. Centrifuge again at 250 g to remove the supernatant.
15. Transfer the beads in a glass beaker with 100 mL of 10 mM
glycine/DDW solution to neutralize the remaining
glutaraldehyde.
16. Stir the solution at room temperature for 1 h.
17. Centrifuge the solution in 50 mL tube at 250 g for 5 min and
discard the supernatant.
18. Wash the beads with DDW (shake well) and centrifuge as
described in step 17. Repeat this procedure three times.
19. Store the beads in DDW at 4 C (Fig. 4d). The beads are stable
for 1 month.
3.5 Protein
Absorption into
the Beads
110
Fig. 5 Equipment and instruments required for grafting gelatin beads. (a) Surgical tools. Sizes of forceps and
scissors depend on animal size. (b) A tungsten needle is melted into a glass Pasteur pipette. Tip of the needle
is sharpened by electrolysis. (c) A bead is poked by the tungsten needle. (d) Beads are placed in the center of
a dish and allowed to dry out, at which point they are shrunken and usually have a yellow color. (e) Proteins
are dropped on the beads and absorbed by the beads. A damp wipe is placed along a margin of the dish to
prevent dehydration of the beads
111
Fig. 6 A series of pictures of the surgery to graft a gelatin bead. (a) Skin is excised from a limb and a small
piece of skin is grafted from the contralateral side of a limb. g grafted skin. Boxes indicate the border of the
wound and the graft. (b) A small tunnel is created by forceps. (c) A bead is inserted underneath the wound
epithelium through the tunnel using a tungsten needle. A black arrow indicates the direction of the tunnel. (df)
The bead is left in the tunnel by removing the tungsten needle gently. White arrows indicate the grafted beads.
(d) Bright field image. (e) Fluorescence image of (d) showing the autofluorescence of the grafted bead. (f)
Merged image of (d) and (e)
112
Notes
1. We prefer to use Trizma preset crystals, pH 7.2 (Sigma-Aldrich
#T8508).
2. It might be appropriate to use type A gelatin depending on the
electrical charge of the protein being used. For example, type
A gelatin is for acidic FGF (FGF1).
3. This fixative needs to determine the stiffness of the beads.
Usually, 2 mM of glutaraldehyde is sufficient for experiments
in axolotls.
4. Proteins are purchased and reconstituted in accordance with
the manufacturers protocol. Usually the proteins are aliquoted
into 1 L each in siliconized microtubes. The aliquoted proteins are stored at 80 C.
5. The size of the wound should be adjusted to approximately the
same size as the transverse dimension of the limb (i.e., larger
wounds on larger animals).
6. It is important not to disorient the skin graft when it is moved
to the wound. To prevent that, prepare 2 510 mm fine lens
paper and place it next to the skin graft. Pick up the skin graft
and flip it onto the lens paper, so that the epidermal side is facing the lens paper. When leucistic or albino mutants are used,
put some dye (e.g., carbon ink) on the skin graft (the dermal
side) to facilitate visualization.
7. The diameter of the expected beads cannot be predicted
because of variability between researchers making the beads.
The size of the bead depends upon the stirring speed and temperature of gelatin solution. Gelatin prepared at higher temperature appears to produce smaller beads. Therefore, the
optimal speed of a stirrer to obtain the desired diameter should
be determined empirically in each laboratory.
8. Proteins typically maintain activity for over 1 week, but usually
maximum activities can be expected within the first couple of
113
Acknowledgments
This work was supported by Grant-in-Aid for Scientific Research
on Innovative Areas (KAKENHI #23124508 to AS and
#22124006 to TE).
References
1. Bryant SV, Endo T, Gardiner DM (2002)
Vertebrate limb regeneration and the origin of
limb stem cells. Int J Dev Biol 46:887896
2. Endo T, Bryant SV, Gardiner DM (2004) A
stepwise model system for limb regeneration.
Dev Biol 270:135145
3. Seifert AW, Kiama SG, Seifert MG, Goheen
JR, Palmer TM, Maden M (2012) Skin shedding and tissue regeneration in African spiny
mice (Acomys). Nature 489:561565
4. Levesque M, Villiard E, Roy S (2010) Skin
wound healing in axolotls: a scarless process.
J Exp Zool B Mol Dev Evol 314:684697
5. Goss RJ (1969) Principles of regeneration.
Academic, New York
6. Mescher AL (1976) Effects on adult newt limb
regeneration of partial and complete skin flaps
over the amputation surface. J Exp Zool 195:
117128
7. Bodemer CW (1958) The development of
nerve-induced supernumerary limbs in the
adult newt, Triturus viridescens. J Morphol
102:555581
8. Bodemer CW (1959) Observations on the
mechanism of induction of supernumerary
limbs in adult Triturus viridescens. J Exp Zool
140:7999
9. Egar MW (1988) Accessory limb production
by nerve-induced cell proliferation. Anat Rec
221:550564
10. Lheureux E (1977) Importance of limb tissue
associations in the development of nerveinduced supernumerary limbs in the newt
Pleurodeles waltlii Michah (authors transl.).
J Embryol Exp Morphol 38:151173
11. Maden M, Holder N (1984) Axial characteristics of nerve induced supernumerary limbs
12.
13.
14.
15.
16.
17.
18.
19.
20.
Chapter 9
High-Efficiency Electroporation of the Spinal Cord
in Larval Axolotl
Aida Rodrigo Albors and Elly M. Tanaka
Abstract
Axolotls are well known for their remarkable ability to regenerate complex body parts and structures
throughout life, including the entire limb and tail. Particularly fascinating is their ability to regenerate a
fully functional spinal cord after losing the tail. Electroporation of DNA plasmids or morpholinos is a valuable tool to gain mechanistic insight into the cellular and molecular basis of regeneration. It provides among
other advantages a simple and fast method to test gene function in a temporally and spatially controlled
manner. Some classic drawbacks of the method, such as low transfection efficiency and damage to the tissue,
had hindered our understanding of the contribution of different signaling pathways to regeneration. Here,
we describe a comprehensive protocol for electroporation of the axolotl spinal cord that overcomes this
limitations using a combination of high-voltage and short-length pulses followed by lower-voltage and
longer-length pulses. Our approach yields highly efficient transfection of spinal cord cells with minimal tissue
damage, which now allows the molecular dissection of spinal cord regeneration.
Key words Electroporation, Spinal cord, Regeneration, Axolotl
Introduction
In the late 1700s, Lazzaro Spallanzani found the salamander tail to
be an eminent model to study regeneration. Due to the transparency of their regenerating tail tissue Spallanzani could describe for
the first time the remarkable process of spinal cord regeneration
[1]. The behavior of the cells lining the central canal of the spinal
cord in response to injury is an integral aspect of spinal cord regeneration in salamanders. Following amputation of the tail, the spinal
cord grows posteriorly as a simple neuroepithelial tube of selfrenewing stem/progenitor cells. After a period of rapid expansion,
the tube undergoes renewed neurogenesis to faithfully reconstruct
tissue architecture and function [25]. Thus, in the view of current
interest in regenerative medicine, salamanders are an unparalleled
system to elucidate cellular and molecular mechanisms involved in
spontaneous recruitment and expansion of neural stem/progenitor
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_9, Springer Science+Business Media New York 2015
115
116
117
Materials
1. Axolotls, 2.22.5 cm snout to tail tip (see Note 1).
2. 10 Phosphate buffered saline (PBS) stock: to prepare 1 L of
10 PBS, dissolve the following reagents in 800 mL of deionized water: 88 g of NaCl, 7 g of NaH2PO4, and 2.3 g of
KH2PO4. Use HCl to adjust the pH to 7.4 and add deionized
water up to 1 L. For 1 PBS solution, dilute 10 stock in
deionized water. Autoclave to sterilize. Store PBS at room
temperature.
3. DNA plasmid: we use the pCAGGs-Cherry-nls plasmid (ubiquitous chicken -actin promoter with a CMV enhancer driving
nuclear Cherry expression) diluted in PBS at a concentration
of 1.5 g/L (see Note 2).
4. DNA Plasmid Maxi Kit (see Note 3).
5. Fast Green solution: make 5 Fast Green solution by dissolving 12.5 mg of Fast Green FCF powder in 10 mL of 1 PBS.
6. 10 TBS: to make 1 L, add 24.2 g of TRIS and 90 g of NaCl
in 990 mL of distilled water. Mix well and adjust the pH to 8.0
by adding 10 mL of HCl 37 %. Store at room temperature for
up to 12 months.
7. Holtfreters solution 400 % (wt/vol): Mix 2.875 g of KCl,
5.36 g of CaCl22H2O, 11.125 g of MgSO47H2O, and
158.4 g of NaCl. Fill up to 10 L with distilled water. Store at
room temperature for up to 6 months.
8. Benzocaine 10 % (wt/vol): To make benzocaine stock solution, dissolve 50 g of benzocaine in 500 mL of 100 % ethanol.
Store at room temperature up to 12 months.
9. Benzocaine 0.03 % (vol/vol): To make 10 L, mix 500 mL of
10 TBS, 500 mL of 400 % Holtfreters, and 30 mL of 10 %
benzocaine. To anesthetize the animals, dilute the 0.03 % solution to 0.008 % in tap water.
10. Polymethyl methacrylate platform and SYLGARD silicone
elastomer (Dow Corning), for stably holding the animal during microinjection (see Note 4).
11. Microloader tips, for loading the microinjection capillaries.
118
12. Sharp and ring forceps, for breaking the microinjection needles
and handling the animals.
13. Borosilicate glass capillaries (1.2 mm O.D., 0.94 mm I.D.).
14. Pressure injector (e.g., PV830 Pneumatic PicoPump, World
Precision Instruments).
15. A stereomicroscope with 3-axis micromanipulator attachment.
16. Electroporation plate: petri dish, UltraPure Agarose, PBS,
scalpel.
17. Tweezer electrodes with round 10 mm platinum plate electrodes (CUY650P10, Nepa Gene).
18. NEPA21 electroporator (Nepa Gene).
19. Cups to house the animals.
Methods
3.1 Preparation
for Microinjection
and Electroporation
1. Make a 2 % (wt/vol) agarose solution in PBS. Heat in a microwave oven to dissolve, swirling to ensure proper mixing.
2. Pour the gel in a 60 mm petri dish and allow to cool down at
room temperature. When the gel solidifies, cut out three wells
with a surgical scalpel to make an electroporation plate
(Fig. 1a). The two parallel wells will hold the electrodes for
electroporating the tail; the third well, the body of the axolotl
(Fig. 1b). Cut a thin line from the third well passing through
the agarose between the two parallel wells. This cut will keep
the tail straight and parallel to the electrodes during electroporation (Fig. 1c) (see Note 5).
Fig. 1 Making of the electroporation plate for holding and protecting the tail during electroporation. (a) Cartoon
illustrating the top view of the electroporation plate. Two wells, 40 mm spaced apart, will hold the electrodes.
The thin cut, 20 mm away from each of these wells, will keep the tail of the axolotl in place and protected from
the electric current, as shown in (b) and, in a lateral view, in (c)
119
3. Dilute DNA plasmid to 1.5 g/L in PBS (see Notes 1 and 6).
Mix the diluted plasmid with Fast Green FCF solution at a
30:1 ratio (see Note 7).
4. Generate microinjection needles on a standard Flaming/
Brown micropipette puller or similar instrument (see Note 8).
5. Set the pressure injector at 2 psi eject pressure and the duration
switch on gated mode (see Note 9).
6. Set up the electroporation parameters. The NEPA21 electroporator allows the configuration of two electrical pulses in one
single electroporation: the poring pulse and transfer pulse. Use
one bipolar poring pulse of 70 V and 5 ms length with a 50 ms
interval and no voltage decay (total two pulses) followed by four
bipolar transfer pulses of 40 V and 50 ms with 999 ms interval
and 10 % voltage decay (total of eight pulses) (see Note 10).
7. Fix the electrode tweezers 7 mm apart (see Note 11). Connect
the lead wires from the electrode tweezers to the electroporator power supply. Place a 100 mL glass bottle or dish filled
with PBS near the work area to submerge the electrodes before
and between each electroporation.
8. Fill the electroporation plate with ice-cold PBS before the start
of the procedure (see Note 12).
9. Anesthetize axolotls in 0.008 % benzocaine. 2.22.5 cm axolotls will take 510 min to fall asleep.
10. Prepare individual cups with tap water.
3.2 Microinjection
into the Spinal Cord
Fig. 2 Setup for microinjection and electroporation of the spinal cord of larval axolotls. (a) Microinjection (1)
and electroporation stations (2) showing the pressure injector (PI), micromanipulator (MN), NEPA21 electroporator (EP), the custom-made electroporation plate (Ep) and commercial electrode tweezers (ET). (b) Lateral
view of the manipulator arm showing the 60 angle respect to the stage plate of the microscope. (c) Image of
the microinjection needle superposed to the spinal cord in the axolotl tail. The opening of the microinjection
121
1. Using round forceps, transfer the axolotl to the electroporation plate filled with ice-cold PBS (Fig. 2f) (see Note 18).
2. Electroporate the tail by applying one bipolar square-wave
pulse of 70 V and 5 ms with 50 ms interval followed by four
bipolar square-wave pulses of 40 V and 50 ms with 999 ms
interval and 10 % voltage decay (Fig. 2g).
3. After electroporation, place the axolotl in its individual cup
with tap water and allow it to recover (see Note 19).
3.4 Confirming
Electroporation
Efficiency
Notes
1. This protocol has been carefully optimized to 2.22.5 cm axolotls. In these animals the electroporation efficiency is the
highest, with the least tissue damage. The same procedure can
be used in larger axolotls, although electroporation parameters
will need to be modified to achieve the best results. Below we
recommend some modifications to our original electroporation conditions that can be used as a starting point.
Fig. 2 (continued) needle should not be bigger than approximately half the thickness of the spinal cord. (d)
View of an axolotl after a successful microinjection into the central canal of the spinal cord. The brain ventricles
and spinal cord are filled with Fast Green. (e) View of the caudal end of the spinal cord after microinjection. The
central canal must be filled with green dye. (f) Top view of an axolotl in the electroporation plate prior to the
procedure. Importantly, the animal is immersed in ice-cold PBS and the tail protected by the agarose. (g) Image
showing tweezer electrodes during the electroporation of the axolotl tail
122
Fig. 3 Highly efficient spinal cord electroporation in a larval axolotl. (a) Schema placing the procedure of spinal
cord electroporation in the context of a typical regeneration experiment in axolotl. (b) Combined bright-field
and fluorescent image of the tail of an axolotl 4 days post-electroporation. The expression of the fluorescent
reporter plasmid pCAGGS-Cherry-nls in the spinal cord suggests a high-efficient transfection of spinal cord
cells. The tail has been amputated at the level of the myotome 12. (c) Cross-section of the electroporated
spinal cord shown in (a), 4 days post-electroporation. (d) Combined bright-field and fluorescent image of the
same animal depicted in (a) after 4 days of regenerative outgrowth. Scale bars in (a) and (c) are 0.5 mm;
50 m in (b)
123
but do not fill it to the brim. For best results, change the electroporation plate after 6080 electroporations.
6. Deionized water does not conduct the electrical pulses efficiently, therefore, dilute the DNA in PBS. For each 2.22.5
axolotl, calculate on 1.5 L of DNA solution per spinal cord.
7. Fast Green FCF is an organic dye that is not taken up by living
cells. Here it is used to visualize the injection of DNA into the
central canal and gauge the amount of solution injected.
8. It is key to produce a microinjection needle tip thin enough to
slip into the central canal of the spinal cord but sturdy enough
to penetrate through the skin and muscle of the tail. Perform a
ramp test as instructed by the manufacturer to determine the
heat value inherent to the glass capillary tube. In our hands, the
following pull cycle parameters (Heat: (ramp test value), Pull:
100, Velocity: 100, Time: 100) provide thinly tapered tips that
will accommodate DNA concentrations up to 2 g/L.
9. Using gentle pressure and manually determining pulse
length provides the best control over the injection.
10. The poring pulse is aimed to open pores in the cell membrane
with minimum damage, and the transfer pulse to deliver
charged DNA into the cells. Based on our experience, the
above-mentioned parameters are the best compromise between
electroporation efficiency and tissue damage in larval axolotls
(2.22.5 cm snout-to-tail length). For larger animals, increase
the voltage to 50 V and the number of pulses to 5.
11. For larger animals, start with a 6 mm gap between electrodes.
If the tail tissue gets damaged, increase the distance to 7 mm.
12. The ice-cold PBS helps preventing excessive heating of the
electroporated tail. Change PBS regularly, so it stays cold.
13. Bring the microloader tip as deep as possible into the microinjection needle, and then eject/push the DNA solution to avoid
the formation of air bubbles. If bubbles form, gently tap the
needle to get rid of them. The animals will not like an air bubble into the spinal cord or brain ventricles. If it happens, sacrifice the animal.
14. Bring the micromanipulator next to the stereomicroscope so
that the tip of the microinjection needle appears in the middle
of the field of view under low magnification. This will help to
operate comfortably during microinjection.
15. In small and transparent axolotls, the spinal cord can be clearly
identified as the brown tubular structure above the notochord.
The opening of the tip should be in a 45 angle and be not
bigger than half the thickness of the spinal cord at the level of
injection (Fig. 2c). To avoid disrupting the spinal cord as much
as possible, we recommend to inject the DNA solution 23 mm
posterior to the planned amputation or injury site.
124
16. This is a critical step. The spinal cord sits only a few micrometers
under the tail muscles, and a fine move of the fine drive unit
of the micromanipulator will bring the tip to the central canal.
Confirm this by gently depressing the footswitch and observing how the DNA solution spreads out. If the tip of the microinjection needle is inside the central canal the solution will
flow inside the spinal cord. If it is not, make tiny movements
bringing the needle tip back and forth while keeping the
footswitch pressed. Stop when you see the green dye flowing
inside the tube. If the DNA solution does not flow, dip the
ring forces in PBS and bring the tip of the microinjection
needle in the PBS film that forms on those. Then, activate
the footswitch to unclog the needle without the need of breaking the tip.
17. Sometimes the DNA solution spreads out in a tube-like manner and gives the false impression that is inside the central canal
of the spinal cord but is, instead, in the meningeal space. This
will result in only the outer neuronal cells being electroporated. Inspect the caudal-most end of the spinal cord. It will be
filled up with the green solution if the injection was successful
(Fig. 2e).
18. It is important to carry out this step as quickly as possible to
avoid the diffusion of injected DNA solution.
19. Examine the animals few hours after electroporation. If the tail
appears visibly curled or burned, sacrifice the animal. We recommend not to change the water of the animals during the
next 34 days to avoid damaging the tails.
20. The expression of the reporter plasmid can be readily observed
2 days post-electroporation. However, we recommend to wait
4 days before proceeding with the experiment for maximum
expression, and to allow the animals to fully recover from the
electroporation.
21. During the initial optimization/training electroporations, we
recommend to fix the piece of amputated tail to quantify the
efficiency of electroporation in tissue sections (Fig. 3c).
References
1. Spallanzani L (1766) Lettre to C. Bonnet
dated A Bomporto, prs de Modene ce 21
Septembre 1766. Edizione Nazionale delle
opere di Lazzaro Spallanzani edn.
2. Holtzer SW (1956) The inductive activity of
the spinal cord in urodele tail regeneration.
J Morphol 99:140
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
125
Chapter 10
Pseudotyped Retroviruses for Infecting Axolotl
Tzu-Hsing Kuo and Jessica L. Whited
Abstract
The ability to introduce DNA elements into host cells and analyze the effects has revolutionized modern
biology. Here we describe a protocol to generate Moloney murine leukemia virus (MMLV)-based,
replication-incompetent pseudotyped retrovirus capable of infecting axolotls and incorporating genetic
information into their genome. When pseudotyped with vesicular stomatitis virus (VSV)-G glycoprotein,
the retroviruses can infect a broad range of proliferative axolotl cell types. However, if the retrovirus is
pseudotyped with an avian sarcoma leukosis virus (ASLV)-A envelope protein, only axolotl cells experimentally manipulated to express the cognate tumor virus A (TVA) receptor can be targeted by infections.
These strategies enable robust transgene expression over many cell divisions, cell lineage tracing, and cell
subtype targeting for gene expression.
Key words Axolotls, Salamander, MMLV, VSV-G, ASLV-A, Limb regeneration
Introduction
The astonishing regenerative abilities of axolotls and other
salamanders have been the subject of intense experimentation
over several centuries. However, a researchers ability to unlock
biological secrets is direct function of the power of the tools
operational in the experimental system. In axolotls, a major hurdle has been the lack of a robust gene expression tool. Recent
advances have permitted the creation of transgenic axolotls [1]
and even transgenic axolotls with inducible gene expression [2, 3].
However, obtaining stable lines of transgenic animals is a lengthy
endeavor perhaps best reserved for genes of special and intense
interest. In other model systems, transgenesis has been complemented with faster methods for examining gene function. Axolotl
researchers have long used electroporation to introduce exogenous
DNA to cells in vivo. However, this method is transient as rapidly
dividing cells, such as those in the regenerating limb blastema,
partition the introduced DNA into daughter cells, resulting in an
eventual diminishing of the expression across the tissue.
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_10, Springer Science+Business Media New York 2015
127
128
129
Materials
Before starting this protocol, please consult the safety department
of your institution. Each institution/country may have specific
regulations for handling pseudotyped retroviruses.
2.1 Plasmid
Components
2.3 Injection/
Infection Components
130
Methods
3.1 Preparation
of Plasmids
3.2 Maintaining/
Splitting 293T Cell
Line (See Note 5)
3.3
Virus Preparation
131
DAY 1: Split 293T cells in the ratio of 1:20 from one confluent
plate. For each gene of interest, six 10 cm culture plates are used
for Day 1.
1. Aspirate and discard old media.
2. Add 10 mL fresh complete media to old plate.
3. Aspirate media against cell surface to detach cells.
4. Add 0.5 mL of cells to each new plate containing 10 mL fresh
media.
5. Place all plates in a humidified incubator at 37 C with 5 %
CO2.
DAY 4: Split one 10 cm plate from Day 1 to two PDL-coated
10 cm plates. For each gene of interest, twelve 10 cm plates are
used for Day 4 (see Note 6).
6. Aspirate old media against cell surface to detach cells.
7. Split media to two PDL-coated (see Note 7) plates containing
5 mL fresh media.
8. Place all plates in a humidified incubator at 37 C with 5 % CO2.
DAY 5: Transfect 293T cells via PEI.
9. Mix plasmid DNA and PEI in serum-free DMEM in 15 mL
conical tubes and vortex briefly. For each plate, 6 g of retroviral vector, 3 g of retro gag pol (helper) plasmid, 1 g of coat
protein plasmid (VSV-G or ASLV-A), 25 L of PEI (1 mg/mL),
and 250 L of serum-free DMEM are used. We use 12 plates
in this protocol; therefore, multiply the components by 12.5.
Mix serum-free DMEM and DNA first, and then add PEI
dropwise to the premixed DNA/DMEM solution.
10. Incubate the DNA/PEI mixture for 1015 min.
11. Meanwhile, replace old media with 10 mL fresh complete
medium for each plate.
12. Evenly apply DNA/PEI mixture dropwise onto the surface of
cells.
13. Swirl plates gently and place plates back to incubator.
DAY 6: Replace media with 5 mL 10 % NuSerum medium for each
plate.
14. Aspirate and discard old media (see Notes 8 and 9).
15. Add 5 mL fresh media containing 10 % NuSerum for each
plate.
16. Place all plates in a humidified incubator at 37 C with 5 %
CO2.
132
133
134
Fig. 1 Infecting axolotls with retrovirus. (a) Arrangement of unopened glass capillary needles in a 15 cm petri
dish. Note that the needles are suspended from the surface by a strip of modeling clay. Lidding the dish keeps
the needles from collecting dust. (b) Unopened needle filled with retrovirus and fast green, which allows for
visualization during injection. Note the long taper of the needle. Filled needles can be broken with forceps at
the tip to open. (c) Juvenile axolotl with a right upper forelimb blastema immediately following infection.
Ventral side is up. (c) Higher magnification of (c). Note that injection of the virus has been confined to the
blastema and does not extend into the stump
135
Fig. 2 In vivo expression of EGFP following infection with QC-EGFP retrovirus. (a, a) Bright-field and fluorescent images of a right forelimb at 5 days postinfection, 19 days post-amputation. (b, b) Bright-field and
fluorescent images of same limb on live animal after full regeneration. Amputation plane was mid-humerus.
Scale bar is 1 mm
Notes
1. The ORF of a gene of interest should be cloned after an appropriate promoter (for example, for ubiquitous expression, CMV
can be used). For a single gene without a marker, a stop codon
is included. However, we recommend using the 2A system to
simultaneously allow for visualizing cells. In this strategy, the
ORF can be cloned without a stop codon and instead followed
directly by 2A and a fluorophore sequence. 2A encodes
18-amino acid self-cleaving peptide that is operational in axolotl [6]; hence, the engineered virus will express one transcript
and a single polypeptide that is then cleaved into the protein of
interest and the fluorophore.
2. A control virus should be prepared in parallel. For example,
QCMV-EGFP, which expresses EGFP alone under the CMV
promoter, can be used as a negative control.
3. To heat inactivate serum, incubate thawed FBS in 56 C water
bath for 30 min. Shake the bottle every 10 min.
136
137
138
139
32. Place the animal on a silicone pad. For blastemas, the animal
can be placed on its ventral side and the limb pulled away from
the animal atop the pad. Alternatively, animals can be placed
on their sides with limbs resting atop the bodies. Be sure to
keep axolotls moist throughout the procedure. This can be
accomplished by performing the procedure quickly or by dabbing the body with axolotl water/tricaine while on the silicone pad.
33. Practice with your specific pneumatic device will be necessary
to become adept at delivering virus. The idea is to fill the area
of interest with the virus-containing fast green solution and to
do it with minimal damage to the animals tissue, minimal
waste/leakage, and minimal carryover to other tissues. Low
pressure is advisable, but too low can result in clogged needles
from slimy tissue becoming impacted at the ejection site or
from virus clogging the needle. Too high will result in premature ejection of the virus, too much virus, and possibly in the
injection of air (which is not good for the animals).
34. Normally, axolotls can be immediately returned to housing
containers and fresh axolotl water. There is no worry that virus
will leak out provided the needles and injection entries were
quite small. However, when first learning the procedure, axolotls can be kept asleep postinfection for 1 h to ensure the
injection site is not disrupted. To do this, gently transfer the
injected animals to a prepared hydration chamber (for example, a large petri dish with tricaine-soaked filter paper).
Following the resting period, transfer the axolotls to fresh
water in normal housing. Throughout the procedures, proceed in compliance with your institutions biosafety guidelines.
This may involve temporarily housing the infected animals in a
BL2-area until the virus is no longer infectious or until a
predetermined time dependent upon your institution.
Decontaminate all areas exposed to the virus. Wear appropriate
personal protective gear (PPE) as VSV-G-coated retroviruses
are capable of infecting human cells.
Acknowledgements
This work was supported by start-up funds from Brigham and
Womens Hospital (J.L.W.). The authors would like to thank
Christina DeMaso in Dr. Connie Cepkos lab (Harvard Medical
School) for sharing similar protocols used in infecting mammalian cells.
140
References
1. Sobkow L et al (2006) A germline GFP transgenic axolotl and its use to track cell fate: dual
origin of the fin mesenchyme during development and the fate of blood cells during regeneration. Dev Biol 290:386397
2. Whited JL, Lehoczky JA, Tabin CJ (2012)
Inducible genetic system for the axolotl. Proc
Natl Acad Sci U S A 109:1366213667
3. Khattak S et al (2013) Germline transgenic
methods for tracking cells and testing gene
function during regeneration in the Axolotl.
Stem Cell Reports 1:90103
4. Atkinson DL et al (2006) Cellular electroporation induces dedifferentiation in intact newt
limbs. Dev Biol 299:257271
5. Cone RD, Mulligan RC (1984) Highefficiency gene transfer into mammalian cells:
generation of helper-free recombinant retrovirus with broad mammalian host range. Proc
Natl Acad Sci U S A 81:63496353
6. Whited JL et al (2013) Pseudotyped retroviruses for infecting axolotl in vivo and in vitro.
Development 140:11371146
7. Echeverri
K,
Tanaka
EM
(2003)
Electroporation as a tool to study in vivo spinal
cord regeneration. Dev Dyn 226:418425
Chapter 11
Thyroxine-Induced Metamorphosis in the Axolotl
(Ambystoma mexicanum)
Peggy S. Coots and Ashley W. Seifert
Abstract
The axolotl (Ambystoma mexicanum) has remained an important model for regeneration and developmental
biology for over a century. Although axolotls in captive-bred colonies usually exist in an aquatic form, they
retain the ability to undergo metamorphosis following exposure to thyroid hormone. Here we present a
robust method for inducing metamorphosis in adult axolotls that results in high survivability and produces
terrestrial animals that can be maintained in long-term captivity.
Key words Metamorphosis, Thyroxine, T4, Thyroid hormone, Axolotl, Ambystoma, Salamander,
Regeneration
Introduction
Amphibian metamorphosis produces a suite of behavioral,
morphological, physiological, biochemical, and genetic changes
associated with the transition from a larval to adult form [13].
These changes are mediated by alterations in circulating thyroid
hormones (T3triiodothyronine and T4thyroxine) and their
interaction with thyroid hormone receptors in target cells [3, 4].
Among urodeles, however, paedomorphosis has evolved in several lineages to produce species that fail to undergo an obvious
metamorphosis in natural populations [5]. Instead, these species become sexually mature while retaining many larval traits.
Although the biochemical changes that mediate the failure to
undergo metamorphosis in these paedomorphic lineages have not
been identified, some urodeles, including Ambystoma mexicanum
(axolotl) and Ambystoma tigrinum (tiger salamander), retain the
ability to undergo metamorphosis following exposure to thyroidstimulating hormone (TSH), T3 and T4 [2, 3, 69].
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_11, Springer Science+Business Media New York 2015
141
142
Materials
1. Axolotls (see Note 1).
2. Round plastic containers (diameter = 21.5 cm, height = 15 cm).
3. Plastic shoeboxes with lids (width = 33.5 cm, depth = 19.5 cm,
height = 9 cm).
4. L-Thyroxine (100 M) stock T4 solution: Add 100 mg
L-thyroxine (T4) (Sigma; Cat. no: T2376) in 10 mL 0.4 M
NaOH in a 15-mL conical tube. Vortex the tube for 30 s and
incubate the tubes in a water bath maintained at 37 C for
5 min. Repeat vortex and heat cycle until L-thyroxine is completely dissolved. This solution is now 13 mM T4. Take 5 mL
of T4 solution and add to 645 mL of sterile water to make
100 M stock solution. The remaining 5 mL of 13 mM T4 can
be wrapped in foil and stored at 4 C.
5. L-Thyroxine (T4) working solution (50 nM): Make
50 nM T4 as needed by mixing 100 M T4 stock solution
with 40 % modified Holtfreters solution at 1:2,000 dilution
(see Note 2).
6. 40 % modified Holtfreters solution: For a 1,136-L
(300-gallon) container, add 1,589.7 g NaCl, 90.8 g NaHCO3,
22.7 g KCl, 181.7 g MgSO47H2O, and 90.8 g
CaCl42H2O. Add all the dry components into the container
and fill to volume with sterile water. Mix with circulation until
all components have gone into solution. Add 75.7 mL Amquel
and 75.7 mL Novaqua to this solution. Solution is now ready
to use (see Note 3).
7. California blackworms (Lumbriculus species).
8. Night crawlers (earthworms).
143
Methods
1. To facilitate successful metamorphic transition, feed animals
California blackworms every day for 1 week prior to the first
addition of T4 working solution (normal feeding schedule is
every 3 days).
2. After 1 week, transfer axolotls into plastic containers with
working T4 solution (one animal per container). The amount
of T4 solution should fill the container (see Note 4).
3. Remove and replace working T4 solution every 3 days.
4. Feed animals after each changing with a generous portion of
California blackworms (approximately 2025 worms). Since
they will stop eating during metamorphosis, the goal is to get
them to eat as much as possible.
5. Obvious morphological changes are rarely visible until approximately 2 weeks after T4 exposure. The first visible changes
occur in the third week and include noticeable weight loss,
dorsal ridge recession, tail fin recession, and eye protrusion
(Fig. 1) (see Note 5).
6. Concomitant with the above morphological changes, the gills
begin to disappear. It is critical to begin lowering the water
level as this happens. The water level should be reduced an inch
per water changing. When the gills have nearly disappeared,
144
Notes
1. In our experience, wild type and albino color morphs exhibit
higher survivability than white mutants. We have used this
method successfully on animals ranging in size from 7 cm TL
to >20 cm TL adults.
2. We typically add 50 mL of T4 stock solution to a 44-gallon
container (e.g., large Rubbermaid trash bin) to make 100 L
working solution (with 40 % modified Holtfreters solution).
The working T4 solution can be used for several days when
morphing a large number of animals.
3. Although we maintain our colony of axolotls at a relatively
constant water temperature of 1820 C, for metamorphosis we
use Holtfreters solution that is at room temp (~2325 C).
4. It is imperative that plastic containers used for induced metamorphosis are kept separately from normal housing containers
because T4 can induce metamorphosis at extremely low concentrations. Once containers are exposed to L-thyroxine, they
should be clearly labeled.
5. As the skin matures to an adult phenotype, it is shed frequently
and this will cause the water to appear dirty. In our experience,
this does not require an increase in water changing frequency.
6. Paper towels provide a surface to help the axolotls reach the
surface and acquire oxygen along with helping to keep the
containers moist. Axolotls will eat significantly less at this point
during metamorphosis and may refuse to eat at all. To help
encourage consumption, place 510 blackworms directly in
front of their mouth.
7. Resumption of feeding is critical for metamorphs to survive in
captivity. A behavioral change is associated with transition to a
terrestrial mode of feeding. Every animal must be offered food,
although every axolotl may not eat at every feeding. This is
normal. Patience is required while feeding metamorphs.
145
Acknowledgments
This work was supported by the Office of the Vice President of
Research at the University of Kentucky.
References
1. Dodd M, Dodd J (1976) The biology of metamorphosis. In: Lofts B (ed) Physiology of the
Amphibia, vol 3. Academic, New York,
pp 467599
2. Page RB, Monaghan JR, Samuels AK, Smith JJ,
Beachy CK, Voss SR (2007) Microarray analysis
identifies keratin loci as sensitive biomarkers for
thyroid hormone disruption in the salamander
Ambystoma mexicanum. Comp Biochem
Physiol C Toxicol Pharmacol 145:1527
3. Rosenkilde P, Ussing AP (1996) What mechanisms control neoteny and regulate induced
metamorphosis in urodeles? Int J Dev Biol
40:665673
4. Brown DD (1997) The role of thyroid hormone in zebrafish and axolotl development.
Proc Natl Acad Sci U S A 94:1301113016
5. Shaffer HB, Voss SR (1996) Phylogenetic and
mechanistic analysis of a developmentally integrated character complex: alternate life history
modes in ambystomatid salamanders. Am Zool
36:2435
6. Norris DO, Platt JE (1974) T3-induced and
T4-induced rates of metamorphosis in immature and sexually mature larvae of Ambystoma
7.
8.
9.
10.
11.
Chapter 12
Generation of Aneurogenic Larvae by Parabiosis
of Salamander Embryos
Anoop Kumar and Jean Paul Delgado
Abstract
Limb regeneration of salamanders is nerve dependent, and the removal of the nerves in early stages of limb
regeneration severely curtails the proliferation of the blastemal cells and growth of the regenerate. The
removal of the neural tube from a developing salamander embryo results in an aneurogenic larva and the
aneurogenic limb (ANL) develops independently without innervation. Paradoxically, the limb in an ANL
is capable of regeneration in a nerve-independent manner. Here, we describe a detailed method for the
generation of ANL in the spotted salamander, Ambystoma maculatum, for regeneration studies.
Key words Newt, Axolotl, Limb regeneration, Nerve dependence
Introduction
Innervation of tissues and organs is a fundamental requirement in
vertebrates as well as invertebrates. In a classical perspective, the
nerves are associated with neurotransmission, whereas in a context
like organ regeneration, the nerves exert dependence on the tissues. The nerve dependence is established during embryonic development and it persists throughout adult life. In tissue regeneration
of mammals, the recent findings indicate that regeneration of hair
follicle [1], digit tip [2], bone marrow [3], and earlobe [4, 5]
requires the presence of nerves. The nerve dependence of tissues
was discovered in relation to salamander limb regeneration [6] and
has been subjected to extensive experimental studies spanning
several decades. These studies revealed several salient features of
nerve dependence during salamander limb regeneration [79].
Transection of the salamander limb in an experimental context
invokes rapid wound healing, followed by dedifferentiation and
proliferation of progenitor cells from the local stump tissues, leading to formation of a limb blastema. The limb blastema is a heterogeneous collection of cells and the origin and nature of these cells
varies among salamander families [1012]. Despite these
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_12, Springer Science+Business Media New York 2015
147
148
149
Materials
2.1 Obtaining
and Maintenance
of Embryos
2.2 Reagents
for Embryo Storage
and Manipulation
2.3 Microsurgery
Tools
150
Fig. 1 Equipment and tools required for microsurgical methods. (a) Custom-built power supply unit with a 4.5
and 9 V D.C. supply. (b) Electrodes with connecting leads. A custom-made electrode holder for multiple tungsten needles (right) and single electrode holder (left upper panel) and negative electrode with platinum wire for
immersion in the electrolyte solution (left, bottom panel). (c) Electrolysis setup. The platinum electrode is
immersed in the beaker containing saturated solution of potassium nitrate, and the terminal is connected to
0 V connector. The tungsten wire is held by the crocodile plug, and the end is connected to desired voltage
terminal. Supply of higher voltage results in faster electrolysis. The low voltage is desirable for fine sharpening
of the needles. (d) Microsurgery tools: tungsten electrode holder, spatula for reshaping the FIMO clay, tweezers, and sharpened tungsten needles in its storage container
151
Methods
3.1 Maintenance
of Embryos
152
153
154
The host larvae are able to feed on brine shrimp during larval
growth (Fig. 2c). The brine shrimps can cause contamination and
fungal infections in the larvae if left for longer periods. Therefore,
it is essential to change the Stenberg solution few hours after feeding. Change the multi-well plates every 45 days.
Notes
1. The rate of limb development varies among different amphibian taxa. The limb development from bud stage to digit stage
is very slow in axolotl larvae compared to A. maculatum.
Therefore, in aneurogenic axolotl larvae, reinnervation of the
limb is possible due to slow growth.
2. In our experience, the modeling clay from FIMO works well.
Other brands which we tested were non-malleable or leaches
color and appear to be toxic to the developing larvae. It is also
possible to make parabiosis chambers from agar, gelatine, or
Dow Corning 184 elastomer. However, these chambers are
not as flexible as FIMO clay to hold the embryos together.
This drawback can be ameliorated by making small bridging
plates made of the same components or thin glass cover slips.
3. The negative platinum electrode is immersed in the electrolyte.
Platinum helps to prevent corrosion and prolong the longevity
of the electrode.
4. The voltage can be reduced to 4.5 V to create a sharp needle
with 23 m tip diameter. It takes longer to produce a fine tip
with reduced voltage, but the low voltage gives better control
in shaping the needle.
5. Long-term storage of the embryos at lower temperature is not
recommended. The embryos develop deformity, and many
embryos in the inner mass of the clutch eventually perish. In
our experience, the length of storage also depends on the stage
of development. Early stage embryos (stage 1015) withstand
lower temperature (710 C), whereas preferred temperature
for late stage embryos is above 10 C. At this storage, developmental progression occurs in the embryos.
6. This step is critical; do not prolong incubation in the KOH
solution. The embryos will perish or deformity may occur during development. If the clutch of eggs contains late stage
155
156
Acknowledgements
We thank Professor Jeremy Brockes for advice and support. Our
work is funded by a Program Grant and MRC Research
Professorship to Jeremy Brockes. JP Delgado is funded by
Programa-Sostenibilidad 20132014, University of Antioquia,
Colombia.
References
1. Brownell I, Guevara E, Bai CB, Loomis CA,
Joyner AL (2011) Nerve-derived sonic hedgehog defines a niche for hair follicle stem cells
capable of becoming epidermal stem cells. Cell
Stem Cell 8:552565
2. Takeo M, Chou WC, Sun Q, Lee W, Rabbani
P, Loomis C, Taketo MM, Ito M (2013) Wnt
activation in nail epithelium couples nail
growth to digit regeneration. Nature 499:
228232
3. Yamazaki S, Ema H, Karlsson G, Yamaguchi T,
Miyoshi H, Shioda S, Taketo MM, Karlsson S,
Iwama A, Nakauchi H (2011) Nonmyelinating
Schwann cells maintain hematopoietic stem
cell hibernation in the bone marrow niche.
Cell 147:11461158
4. Buckley G, Metcalfe AD, Ferguson MW
(2011) Peripheral nerve regeneration in the
MRL/MpJ ear wound model. J Anat 218:
163172
5. Buckley G, Wong J, Metcalfe AD, Ferguson
MW (2012) Denervation affects regenerative
responses in MRL/MpJ and repair in
C57BL/6 ear wounds. J Anat 220:312
6. Todd TJ (1823) On the process of reproduction of the members of the aquatic salamander.
Q J Sci Lit Arts 16:8496
7. Kumar A, Brockes JP (2012) Nerve dependence in tissue, organ, and appendage regeneration. Trends Neurosci 35:691699
8. Singer M (1952) The influence of the nerve
in regeneration of the amphibian extremity.
Q Rev Biol 27:169200
9. Stocum DL (2011) The role of peripheral
nerves in urodele limb regeneration. Eur J
Neurosci 34:908916
10. Brockes JP, Kumar A (2002) Plasticity and
reprogramming of differentiated cells in
amphibian regeneration. Nat Rev Mol Cell
Biol 3:566574
11. Simon A, Tanaka EM (2013) Limb regeneration. Wiley Interdiscip Rev Dev Biol 2:
291300
12. Sandoval-Guzman T, Wang H, Khattak S,
Schuez M, Roensch K, Nacu E, Tazaki A,
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
157
Chapter 13
In Vivo Modulation and Quantification of microRNAs
During Axolotl Tail Regeneration
Jami R. Erickson and Karen Echeverri
Abstract
The ability to regenerate diseased, injured, or missing complex tissue is widespread throughout lower
vertebrates and invertebrates; however, our knowledge of the molecular mechanisms that regulate this
amazing ability is still in its infancy. Many recent papers have shown important roles for microRNAs in
regulating regeneration in a number of species. The ability to detect and quantify miRNA expression fluctuations at a single cell level in vivo in different cell types during processes like regeneration is very informative. In this chapter, we describe how to use a dual-fluorescent green fluorescent protein (GFP)-reporter/
monomeric red fluorescent protein (mRFP)-sensor (DFRS) plasmid to quantitate the dynamics of specific
miRNAs over time following miRNA mimic injection as well as during regeneration. In this bicistronic
vector, the mRFP allows for verification of miRNA expression, while the GFP functions as an internal
control to normalize miRNA expression and thus obtain quantitative results. In addition, we demonstrate
how this technique revealed dynamic miR-23a expression and function during tail regeneration.
Key words microRNAs, Axolotl, Sensor plasmids
Introduction
The ability to regenerate multiple body parts is a fascinating phenomenon that remains poorly understood at a molecular level [1
12]. While model systems like axolotls are desirable since they can
regenerate these tissues, lack of a full axolotl genome sequence
hinders classical genetic studies. Recent studies have shown that
small noncoding microRNAs (miRNAs) are highly conserved and
may play an important role in regulating gene expression after
injury in many species including axolotls [1319].
miRNAs are small noncoding RNAs that are approximately 22
nucleotides long [2022]. The major function of miRNAs is posttranscriptional regulation of gene expression. The canonical mechanism of action for miRNAs is to bind to a region of complementary
sequence in the 3 untranslated region (UTR) of the target
messenger RNA (mRNA). The binding of the miRNA to its target
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_13, Springer Science+Business Media New York 2015
159
160
Fig. 1 Schematic of DFRS plasmid. This dual miRNA reporter construct can be
used to analyze and quantify expression of endogenous miRNAs and to quantitatively evaluate the effect of modulators of microRNAs, like inhibitors or mimics.
The DFRS plasmid contains both GFP (control) and mRFP (reporter), each under
the control of their own SV40 promoter. Following the open read frame of mRFP
is a multiple cloning site (MCS) to allow addition of seed sequences for an miRNA
of interest
161
Materials
1. Fluorescent dissecting scope.
2. Pressure injector.
3. Micromanipulator.
4. Electroporator.
5. Electrodes.
6. Needle puller.
7. Axolotls.
8. miRNA DFRS plasmids.
9. miRNA mimic.
10. Anesthesia (0.01 % p-aminobenzene).
11. Fast Green.
12. Glass capillaries.
13. Sylgard-dissecting pad.
14. Insect pins.
15. Microloader pipette tips.
16. 1 PBS.
17. Tweezers.
18. Image J.
Methods
3.1 Preparation
of Plasmid Solution
for Injection
1. To prepare the plasmid solution for injection, dilute the dualfluorescent green fluorescent protein (GFP)-reporter/monomeric red fluorescent protein (mRFP)-sensor (DFRS) plasmid
to a final concentration of 0.5 g/L with 0.2 mg/mL Fast
Green and 1 phosphate buffered saline (PBS) (see Note 1).
3.2 Preparation
of Needles
162
3. Insert and secure the loaded injection needle into the pressure
injector nozzle attached to the micromanipulator.
4. To open the needle, very gently break the very tip using a forceps. To ensure the needle is open, press the injection pedal to
observe release of the plasmid solution.
3.3 Injection
and Electroporation
of Animals
3.4
Tail Amputation
3.5
Mimic Injection
163
Quantification
164
Fig. 2 Quantifying miR-23a dynamics in single cells during tail regeneration. (a) Control GFP expression (green)
and reporter mRFP expression (red) shown at 0, 24, 48, and 96 hour post amputation (hpa). Arrowheads indicate examples of cells that are expressing the DFRS plasmid and are used for quantification. Scale bar is
200 m. (b) Graphical representation of mRFP expression modulated by miR-23a in the spinal cord during
regeneration. mRFP expression is dynamic during the regenerative process, revealing that miR-23a expression is increased at 24 and 96 hpa, while miR-23a levels at 48 hpa are similar to steady state levels of miR23a (***P < 0.001, NS not significant)
Fig. 3 Determining the effect of microRNA mimic injection using miR-141a sensor. (a) Control GFP expression
(green) and reporter mRFP expression (red) shown at 0, 24, 48, and 96 hour post mimic injection (hpi).
Arrowheads indicate examples of cells that are expressing the DFRS plasmid and are used for quantification.
Scale bar is 200 m. (b) Graphical representation of mRFP expression modulated by miR-141a in the spinal
cord following mimic injection. Overall there is a general decrease in the amount of mRFP expression until
96 hpi, indicating that most DFRS plasmid-expressing cells also took up and processed injected miR-141
mimics (***P < 0.001, **P < 0.01, NS not significant)
165
Notes
1. Adding Fast Green will allow visualization of the injection
under visible light and is an ideal way to localize injections to
the correct tissue site.
2. Needles can also be pulled manually or pre-pulled needles can
be ordered.
3. Animals used here are 23 cm in length, injection volumes and
electroporation conditions may need to be varied depending on
the size of animals used. For further details on how microinjection and electroporation are performed, refer to refs. 29 and 30.
4. The injection is successful if the Fast Green color can be visualized in the tissue of interest. If not, adjust the injection needle
using the micromanipulator and reinject.
5. The orientation of the electric field can be used to direct plasmid uptake to specific regions, or the electrodes can be inverted
and the axolotl can be electroporated a second time for bidirectional plasmid uptake [29, 30].
6. Fluorescence of mRFP and GFP can be visualized within 24 h
and can confirm a successful injection.
7. After visualization of mRFP and GFP, the tail can be amputated adjacent to the DFRS-expressing cells to examine the
expression patterns of the miRNA of interest (Fig. 2a).
8. If the expression of the miRNA of interest increases during the
regenerative process, a decrease in mRFP expression relative to
GFP will be observed (Fig. 2a, b).
9. Here we have demonstrated the effect of a mimic on microRNA
levels, and this same technique can also be used to assay the
quantitative effect of microRNA inhibitors in vivo.
10. For each individual microRNA mimic or inhibitor, the optimum concentration must be determined by testing different
concentrations.
11. After visualization of mRFP and GFP, an miRNA mimic can be
injected and targeted to the DFRS-expressing cells.
12. If Fast Green color appears in the desired location, remove the
needle. If the injection was not administered in the correct
location, adjust the position of the injection using the
micromanipulator and reinject.
13. Be sure to use the same exposure time as well as other microscope
settings as the first imaged time point. We have found that a significant decrease in relative mRFP fluorescence can be observed at
24 h after mimic injection. If the injected mimic is targeting the
binding site at the 3 UTR of mRFP, a decrease in the mRFP
expression relative to GFP can be observed (Fig. 3a, b).
14. Synchronizing the windows will allow the same area to be
measured in each channel.
166
Acknowledgements
We thank Davide De Pietri Tonelli and Antonio Giraldez for the
kind gift of the DFRS plasmids.
The authors are grateful for support from the Stem Cell
Training Grant (T32 HD060536 04) for this project as well as
funding from the Department of Genetics, Cell Biology and
Development at the University of Minnesota.
References
1. Kumar A, Brockes JP (2012) Nerve dependence in tissue, organ, and appendage regeneration. Trends Neurosci 35:691699
167
Part III
Salamander Cells in Culture
Chapter 14
Derivation and Long-Term Culture of Cells
from Newt Adult Limbs and Limb Blastemas
Patrizia Ferretti and Anoop Kumar
Abstract
Notwithstanding the key importance of in vivo models for understanding patterning and cellular
interactions in the regenerating tailed amphibian (salamander) limb, dissection of molecular mechanisms,
as in other species, can be greatly aided by robust in vitro models. This chapter focuses on derivation and
maintenance of cell lines from adult post-metamorphic salamanders and in particular cells derived from
normal and regenerating limbs. We also describe a protocol for nucleofecting newt cells that can be used
both to investigate the gene function in short-term studies and to establish stable cell lines.
Key words Axolotl, Blastema, Cell culture, Limb, Nucleofection, Regeneration, Transfection,
Salamander
Introduction
Cultures of mesenchymal cells from the late-bud stage blastemas
of 915-month-old axolotls were used by Boilly and Albert [1],
but they were not established as lines available to the scientific
community.
Long-term cultures of cells derived either from limb blastemas
or normal limbs from adult urodeles (tailed amphibian) with the
ability to undergo myogenic differentiation were first established
in 1988 [2]. The cell lines derived from the newt, Notophthalmus
viridescens, that have been more extensively used are a limb blastema cell line, originally named as BlH1 (blastema hind limb 1)
and more recently called B1H1, and two lines from normal hind
limbs derived either from ankle (A1) or thigh (TH4B) tissue [3, 4].
Both cell lines derived from normal limbs and limb blastemas
contain thin, well-spread, and large cells as well as smaller, thicker,
and often bipolar cells (Fig. 1a, b). All cells present in these cultures are much larger than mammalian cells, with nuclei up to
50 m in diameter. They have an average duplication time of
2 days, which can vary depending on the line and culture
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_14, Springer Science+Business Media New York 2015
171
172
Fig. 1 Newt cells cultured from normal limb and limb blastema maintained either at low/medium density (ac)
or at high density to induce myogenic differentiation (e, f ) express blastemal (c, d) and muscle markers (f),
respectively. (a, b) Live images of BlH1 and TH4 cells. (c, d) Newt cells stained for blastemal markers, 22/18
(c) and K8 (d). (e) Histological staining of cells grown at high density; some myotubes are indicated by arrowheads. (f) Myotube stained with the muscle marker, 12/101
173
174
Fig. 2 A1 and BlH1 newt cells nucleofected with either RFP (a) or GFP (b) constructs. Note that the high levels
of expression of the nucleofected gene are observed in both cell lines
Materials
Here we indicate the materials required and the sources we use,
but unless otherwise specified, equivalent reagents obtained from
other suppliers will work as well. Tissue culture flasks from Corning,
Nunc, or BD Falcon have all proven suitable for culturing newt
cells (plastic from other sources has given variable results).
2.1 Sources
of Adult Newts
175
3. 12 cm spring scissors.
4. Bone cutter.
5. Dissection board.
6. Sterile water: Aliquot commercially available HPLC grade
water in 250 mL bottles and autoclave. Store at room temperature (see Note 1).
7. Sterile glass Pasteur pipettes.
8. Sterile Petri dishes and 50 mL tubes.
9. Sodium hypochlorite solution: Prepare 1 % sodium hypochlorite solution in sterile water and adjust the pH to 8.8 with
1 N HCL.
10. Tricaine (ethyl 3-aminobenzoate methanesulfonate) (MS222): Prepare a 0.1 % solution using filtered water available in
the amphibian facility. Adjust the pH between 7.2 and 7.5
using 1 N NaOH. Prepare fresh before use.
11. Recovery solution for newts: Dissolve 10 g sulfamerazine
(Sigma, Cat. no. S0800) in 2 L of filtered water available in an
amphibian facility. Add 0.1 mL analgesic solution (Alvegesic
Vet, stock 10 mg/mL) and adjust the pH between 7 and 7.5
with 5 N NaOH.
2.3 Reagents
and Equipment
for Newt Cell Isolation
and Maintenance
176
177
178
2.4 Equipment
and Reagents
for Nucleofection
of Newt Cells
Methods
3.2 Tissue
Preparation
for Culturing
3.3 Dissociated
Cultures from Newt
Limb Blastemas
179
180
181
3.7 Thawing
Newt Cells
The procedure for thawing newt cells is similar to that used for
mammalian cells, with much care needed to rapidly dilute and
eliminate the DMSO present in the freezing medium in order to
avoid toxic effects and maximize cell recovery.
1. Dispense 8 mL of cold AMEM or L15 in a conical centrifuge
tube.
2. Rapidly thaw the cells by immersing the vial in a water bath
at 37 C.
3. As soon as the cell suspension has thawed and is still cold,
dilute it in the 8 mL of medium in the centrifuge tube.
4. Spin the cell suspension at 62 g for 5 min. Decant the supernatant and resuspend the pellet in 5 mL of AMEM.
5. Transfer the cell suspension to a 75 cm2 (or smaller) gelatinecoated flask and place it in the amphibian cell incubator.
3.8 Nucleofection
of Newt Cells
182
183
Notes
1. Sterile water bottles should be designated for amphibian cell
culture only and prepared as follows: use new bottles, fill them
with distilled water, and autoclave; discard the autoclaved
water and then fill the bottles with HPLC grade water and
autoclave. Do not wash the bottles with detergents.
2. Amphibian cells do not grow equally well in all fetal calf sera
(FCS). Therefore, batch screening is essential. Usually testing
34 samples of serum from a supplier is sufficient for identifying a good growth promoting FCS. Once a suitable batch has
been identified, an appropriate number of 500 mL bottles
(e.g., 2040 depending on the volume of work anticipated) of
that FCS batch should be reserved. The FCS can be stored at
20 C for several years.
3. The culture medium can be stored at 4 C for a couple of
weeks. The medium should be brought to room temperature
before use. Do not warm it in a water bath at 37 C.
4. As L-15 medium is amino acid rather sodium bicarbonate
buffered, it effectively maintains a physiological pH when handling tissue and cells outside the CO2 incubator. Cells can be
cultured long-term in this medium if an incubator with 2 %
CO2 is not available.
5. Gentamicin is used at the dissection stage as it is rather effective in reducing contaminations, but it was found that its prolonged use affects cell growth. Hence, it should not be included
in the growth medium.
6. All of the lines currently in use were started by plating cells or
explants onto collagen-coated plastic, but as the newt limb cell
lines attach well also on gelatine, it is expected this would be a
suitable substrate for starting new lines.
7. Normal limb tissue is very difficult to dissociate, and only occasionally live cells are obtained following enzymatic digestion.
In contrast, blastemas are more easily dissociated but more
prone to contaminations.
8. If antibody staining and high-resolution imaging under oil
objective is anticipated, culturing of newt cells on Nunc 35 mm
Petri dishes is recommended. This will allow mounting of the
dishes with a round cover slip and easy trimming of the edges
of the plate with a pair of sharp scissors to accommodate the oil
objectives. Newt cells usually do not grow optimally on glass
184
coverslips. If plating the cells on a coverslip, coating with polylysine before gelatine coating helps cell attachment.
9. The presence of antibiotics in the freezing medium does not
appear to cause any adverse effect.
10. The cells can be kept at 70 C for a few days but ideally should
be transferred into liquid nitrogen within 24 h.
11. Efficiency of transfection using the Lonza kit and Mirus electroporation solution was assessed in parallel using various plasmid and comparable results obtained. Cuvettes with larger
volumes can also be used for nucleofection of cells.
12. The nucleofection approach described here is also applicable
to cells derived from the limb of axolotl, a paedomorphic
salamander.
References
1. Boilly B, Albert P (1988) Blastema cell proliferation in vitro: effects of limb amputation on
the mitogenic activity of spinal cord extracts.
Biol Cell 62:183187
2. Ferretti P, Brockes JP (1988) Culture of newt
cells from different tissues and their expression
of a regeneration-associated antigen. J Exp
Zool 247:7791
3. Corcoran JP, Ferretti P (1997) Keratin 8 and
18 expression in mesenchymal progenitor cells
of regenerating limbs is associated with cell
proliferation and differentiation. Dev Dyn
210:355370
4. Corcoran JP, Ferretti P (1999) RA regulation
of keratin expression and myogenesis suggests
different ways of regenerating muscle in adult
amphibian limbs. J Cell Sci 112:13851394
5. Sessions SK (2008) Evolutionary cytogenetics
in salamanders. Chromosome Res 16:
183201
6. Morrison JI, Borg P, Simon A (2010) Plasticity
and recovery of skeletal muscle satellite cells
during limb regeneration. FASEB J 24:
750756
7. Ferretti P, Fekete DM, Patterson M, Lane EB
(1989) Transient expression of simple epithelial keratins by mesenchymal cells of regenerating newt limb. Dev Biol 133:415424
8. Casimir CM, Gates PB, Ross-Macdonald PB,
Jackson JF, Patient RK, Brockes JP (1988)
Structure and expression of a newt cardioskeletal myosin gene. Implications for the C
value paradox. J Mol Biol 202:287296
9. Kosaka M, Kodama R, Eguchi G (1998) In
vitro culture system for iris-pigmented
10.
11.
12.
13.
14.
15.
16.
17.
epithelial cells for molecular analysis of transdifferentiation. Exp Cell Res 245:245251
Simon A, Brockes JP (2002) Thrombin activation of S-phase reentry by cultured pigmented
epithelial cells of adult newt iris. Exp Cell Res
281:101106
Bettencourt-Dias M, Mittnacht S, Brockes JP
(2003) Heterogeneous proliferative potential
in regenerative adult newt cardiomyocytes.
J Cell Sci 116:40014009
Lo DC, Allen F, Brockes JP (1993) Reversal of
muscle differentiation during urodele limb
regeneration. Proc Natl Acad Sci U S A 90:
72307234
Tanaka EM, Drechsel DN, Brockes JP (1999)
Thrombin regulates S-phase re-entry by cultured newt myotubes. Curr Biol 9:792799
Tanaka EM, Gann AA, Gates PB, Brockes JP
(1997) Newt myotubes reenter the cell cycle
by phosphorylation of the retinoblastoma protein. J Cell Biol 136:155165
Beug S, Vascotto SG, Tsilfidis C (2006) Newt
orthologue of Growth arrest-specific 6
(NvGas6) is implicated in stress response during newt forelimb regeneration. Dev Dyn
235:711722
Giampaoli S, Bucci S, Ragghianti M, Mancino
G, Zhang F, Ferretti P (2003) Expression of
FGF2 in the limb blastema of two
Salamandridae correlates with their regenerative capability. Proc Biol Sci 270:21972205
Kumar A, Velloso CP, Imokawa Y, Brockes JP
(2000) Plasticity of retrovirus-labelled myotubes in the newt limb regeneration blastema.
Dev Biol 218:125136
185
Chapter 15
Culture and Transfection of Axolotl Cells
Jean-Franois Denis, Fadi Sader, Patrizia Ferretti, and Stphane Roy
Abstract
The use of cells grown in vitro has been instrumental for multiple aspects of biomedical research and especially
molecular and cellular biology. The ability to grow cells from multicellular organisms like humans, squids,
or salamanders is important to simplify the analyses and experimental designs to help understand the biology of these organisms. The advent of the first cell culture has allowed scientists to tease apart the cellular
functions, and in many situations these experiments help understand what is happening in the whole
organism. In this chapter, we describe techniques for the culture and genetic manipulation of an established cell line from axolotl, a species widely used for studying epimorphic regeneration.
Key words Axolotl, Cell culture, Electroporation, Limb regeneration, Lipofection, Salamander,
Transfection
Introduction
The ability to grow cells from multicellular organisms in vitro was
invented by R.G. Harrison at the beginning of the twentieth century. Over a century after this development, tissue culture has
become an intrinsic part of the experimental arsenal used in every
modern laboratory studying molecular and cellular biology, biochemistry, and genetics whether for biomedical or fundamental
reasons. The ability to grow cells in culture is important to help
our understanding of how different cell types proliferate or respond
to specific signals. Tissue culture is therefore an important complement to studying processes that occur in vivo. We are particularly
interested in the process of epimorphic regeneration in vertebrates
and the animals of choice for this are the urodele amphibians (the
most commonly used models are the axolotl and the newt).
Although cultures from human and mouse cells have been established with tremendous success, it is often a challenge to set up cell
cultures from lower vertebrates like salamanders.
The first, and presently the only, cell line available from an
Axolotl Limb is the AL-1 cell line (Fig. 1). It was established in the
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_15, Springer Science+Business Media New York 2015
187
188
Fig. 1 AL-1 cells transfected with plasmid containing the CMV-mCherry expression cassette. (ac) Transfection
with 0.5 g of plasmid using Lipofectamine 3000 at 37 C for 4 h. (a) Bright-field view of transfected cells
indicates that cells are attached and normal looking. (b) Same cells as (a) visualized with the mCherry filter.
(c) Overlay of bright-field and mCherry from (a) and (b) indicates that about 40 % of cells are transfected. (df)
Transfection using square wave electroporation of 5 g of plasmid. (d) Bright-field view of transfected cells,
less cells survive electroporation which explains the lower number of cells in the frame. (e) Same cells as (d)
visualized with the mCherry filter. (f) Overlay of bright-field and mCherry from (d) and (e) shows that about
50 % of cells are transfected. The cells were photographed with a 20 objective
2
2.1
189
Materials
AL-1 Culture
190
2.3 Equipment
and Reagents
for Nucleofection
of AL-1 Cells
Methods
3.1 Thawing
AL-1 Cells
191
6. Spin the cell suspension at 62 g for 5 min. Decant the supernatant and resuspend the pellet in 5 mL of AMEM.
7. Transfer the cell suspension to a 75 cm2 (or smaller) flask and
place it in the amphibian cell incubator.
8. Change the medium on the following day.
3.2 Maintenance
of AL-1 Cells
192
3.3 Cryopreservation
of AL-1 Cells
3.4 Electroporation
of AL-1 Cells
3.5 Transfection
of AL-1 Cells
with Lipofectamine
3000
193
3.6 Transfection
of AL-1 Cells by
Nucleofection
194
Notes
1. Originally, AL-1 cells were grown in 10 % FBS with 1 ITS,
but protocol optimization experiments showed that no difference in cell growth is observed by lowering FBS to 5 % and ITS
concentrations to 0.2.
2. AL-1 cells need to be maintained at 4090 % confluence for
optimal cell growth. If the density is too low, the cells will
195
Table 1
Methods of transfection tested on AL-1 cells
Method of transfection
Result of transfection
Toxicity
Electroporation (square
wave is necessary)
Cytofectamine (Bio-Rad)
0 % (done at 26 C)
None observed
Fugene-HD (Roche)
None observed
Lipofectin (Invitrogena)
0 % (done at 26 C)
0 % (done at 26 C)
Lipofectamine 2000
~20 % if done at 37 C
4h
Lipofectamine 3000a
0 % (done at 26 C)
None observed
Lipofectamine 3000a
~50 % if done at 37 C
4h
0 % (done at 26 C) 4
and 16 h
Magnetofection PolyMag
(Oz Biosc)
Less than 5 % at 26 C
Nucleofection
6080 %
3050 % survival
Ca-Pi precipitation
0 % (done at 26 C)
None observed
Lipofectamine 2000
N.B. for each method listed, various ratios of plasmid to reagents were tested and only the summary is presented in this
table. The most efficient methods are the ones described in detail (i.e., electroporation, Lipofectamine 3000, and
nucleofection)
196
7. Use about 5 105 cells per 0.2 cm cuvette. Due to the large
size of amphibian cells, manufacturers recommended number
of cells is not required. Nucleofection can be successfully carried
out also using a lower number of cells. However, under these
conditions cell death is higher. Cuvettes with larger volumes
can also be used for nucleofection of cells.
8. Efficiency of transfection using the Lonza kit and Mirus electroporation solution was assessed in parallel using various plasmids and comparable results were obtained.
References
1. Roy S, Gardiner DM, Bryant SV (2000)
Vaccinia as a tool for functional analysis in
regenerating limbs: ectopic expression of Shh.
Dev Biol 218:199205
2. Levesque M, Gatien S, Finnson K, Desmeules
S, Villiard E, Pilote M, Philip A, Roy S (2007)
Transforming growth factor: beta signaling is
essential for limb regeneration in axolotls. PLoS
One 2:e1227. doi:10.1371/journal.pone.
0001227
3. Shaikh N, Gates PB, Brockes JP (2011) The
Meis homeoprotein regulates the axolotl Prod
1 promoter during limb regeneration. Gene
484:6974
4. Villiard E, Brinkmann H, Moiseeva O, Mallette
FA, Ferbeyre G, Roy S (2007) Urodele p53 tolerates amino acid changes found in p53 variants
linked to human cancer. BMC Evol Biol 7:180.
Chapter 16
Isolation and Culture of Neurospheres
from the Adult Newt Brain
Liyakath Ali Shahul Hameed and Andrs Simon
Abstract
Neural stem cells (NSCs) give rise to neurons in the adult brain and are possible targets in regenerative
therapies. In vitro cultures of NSCs as neurospheres have been established from cells isolated from diverse
species. Newts are exceptional regenerators among vertebrates. These animals are able to efficiently
replace neurons following ablation of those by activation and subsequent differentiation of NSCs. Here
we describe the method for isolating and culturing of NSCs from the newt brain both during self-renewing and differentiating conditions. Newt NSC culture provides a useful tool for functional studies of NSC
fate with the potential of resulting in novel regenerative strategies.
Key words Primary culture, Neurosphere, Neural stem cells, Newt
Introduction
Newts have remarkable regeneration capacities among adult vertebrates [1], which extend also to the brain. In a series of experiments, it has been shown that the newt brain is able to activate
sufficient cues to direct activated NSCs toward specific neuronal
subtypes within an existing brain tissue [24]. As a complement to
in vivo studies, in vitro cultures of NSCs offer additional opportunities to reveal regulatory mechanisms that control NSCs fate.
A hallmark of NSCs is their potential to form neurospheres in
culture. In cultures, NSC fate is easy to manipulate by over- and
under-expression approaches and by treatment with small molecules. Such manipulations can be used to uncover critical regulatory mechanisms underlying proliferation and differentiation
toward neurons. Recently, we used this approach to characterize
the heterogeneity of newt NSCs isolated from defined brain regions
at the molecular level and carry out functional manipulations of
neurogenesis [5].
Here, we describe a detailed protocol for the formation and
propagation of neurospheres from the adult newt brain (Fig. 1).
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_16, Springer Science+Business Media New York 2015
197
198
Fig. 1 Formation and differentiation of neurospheres. (a) Small spheres appearing 4 days after plating.
(b) Spheres typically reach maximum size around day 14. (c) Plating neurospheres on PDL plates leads to
attachment of the neurospheres to the substrate. (d) By day 15, differentiating neurospheres are apparent by
bright-field microscopy
Materials
Prepare all solutions fresh before use. Dilute all culture medium to
66 % with Milli-Q water to adjust amphibian osmolarity (see Note 1),
and filter using 0.22 m syringe filter.
199
Fig. 2 Immunocytochemical characterization of neurospheres. (ad) Growing neurospheres stained with stem
cell marker GFAP and proliferation marker, PCNA. Most of the GFAP-expressing cells are marked by the proliferation marker, PCNA. (eh) Differentiating neurospheres stained with the neuronal marker, Tuj-1, and the glial
marker, GFAP. This image illustrates that most of the progeny are neurons and some progeny are glial cells
200
Methods
All the cell culture procedures are carried out under aseptic
conditions.
3.1 Isolation
of Brain Cells
201
2. Sacrifice the animal and dissect out the brain under a dissection
microscope.
3. Drop the extracted brain in to a 60 15 mm Petri dish containing dissection solution.
4. Remove carefully the meninges and blood vessels off the brain
with fine forceps. Wash the tissue by transferring it to a new
60 15 mm Petri dish containing dissection solution (see Note 7).
5. Place the brain into fresh dissection medium and take the brain
into culture hood. Use a scalpel to cut the brain into small
(approximately 1 mm3) pieces (see Note 8).
6. Collect the pieces of brain using a 1,000 L pipette and transfer to the fresh 1.5 mL microtube. Allow the tissue to sink to
the bottom by gravity, and remove the supernatant solution.
7. Add 500 L freshly made enzyme digestion solution and incubate at room temperature for 1 h. During the incubation, stir
the tube gently once every 15 min (see Note 9).
8. Add equal volume of enzyme inhibitor to stop the enzyme activity.
Incubate 5 min at RT. Transfer the incubation mixture to 15 mL
falcon tube and centrifuge for 5 min at 80 g (see Note 10).
9. Remove supernatant and add 1 mL of fresh dissection medium
to the precipitate.
10. Dissociate the newt brain cells by triturating with 1,000 L
pipette for 2 min slowly up and down (see Note 11).
11. Filter cell suspension through a 40 m tissue strainer to remove
any undissociated cells or tissue clumps.
12. Collect the filtered cells and centrifuge at 80 g for 5 min.
13. Remove most of the supernatant, leaving 300 L in the tube to
make sure that cells are not lost.
14. Resuspend the cells in 4 mL of expansion medium and transfer
them to a 25 cm2 cell culture flask (see Note 12).
3.2 Neurosphere
Formation
and Differentiation
202
3.3 Immunostaining
of Neurospheres
and Differentiated
Neurospheres
Notes
1. Amphibian osmolarity is 225 5 mOsmol. Therefore, the culture medium is diluted to 66 % with Milli-Q water to match
the osmolarity of the newt cells.
2. It is important to remove any remaining ethanol from the surgical tools by rinsing them with sterile PBS before touching
the tissue. Traces of ethanol will act as a fixative for the tissue.
203
204
Acknowledgment
This work was supported by a grant from the Swedish Research
Council to AS.
References
1. Brockes JP, Kumar A (2008) Comparative
aspects of animal regeneration. Annu Rev Cell
Dev Biol 24:525549
2. Berg DA, Kirkham M, Beljajeva A, Knapp D,
Habermann B, Ryge J, Tanaka EM, Simon A
(2010) Efficient regeneration by activation of
neurogenesis in homeostatically quiescent
regions of the adult vertebrate brain. Development 137:41274134
3. Berg DA, Kirkham M, Wang H, Frisen J, Simon
A (2011) Dopamine controls neurogenesis in
the adult salamander midbrain in homeostasis
and during regeneration of dopamine neurons.
Cell Stem Cell 8:426433
4. Parish CL, Beljajeva A, Arenas E, Simon A
(2007) Midbrain dopaminergic neurogenesis
and behavioural recovery in a salamander lesion-
Chapter 17
Methods for Axolotl Blood Collection, Intravenous
Injection, and Efficient Leukocyte Isolation from Peripheral
Blood and the Regenerating Limb
Ryan J. Debuque and James W. Godwin
Abstract
The vertebrate immune system comprises both adaptive and innate immune cells with distinct functions
during the resolution of inflammation and wound healing after tissue injury. Recent evidence implicates a
requirement for innate immune cells from the myeloid lineage during the early stages of limb regeneration in
the Mexican axolotl. Understanding the functions of innate and adaptive immune cells in the axolotl has been
hampered by a lack of approaches to isolate and analyze these cells. Here we describe a protocol to isolate
myeloid cells from the regenerating axolotl limb that incorporates intravenous delivery of physiological labels.
In addition we provide a protocol to enrich for leukocytes in the peripheral blood. These protocols produce
single-cell suspensions that can be analyzed using flow cytometry or sorted into specific subsets using fluorescent-activated cell sorting (FACS). FACS is a routine approach to sort cells based on their physical characteristics as well as their cell surface antigen repertoire. Isolated cell populations can then be analyzed in a
wide range of downstream assays to facilitate a greater understanding of leukocyte biology in the axolotl.
Key words Salamander, Leukocyte biology, Myeloid cell, Flow cytometry and regeneration
Introduction
Urodele amphibians possess the remarkable capacity to resolve
injury in a range of clinically relevant organs such as the heart,
brain, and spinal cord as well as regenerate structures such as limbs
and tails [1]. Resolution of tissue injury triggers an inflammatory
response that is mediated by cells of the immune system, a mechanism conserved between mammals and vertebrates with robust
regenerative abilities [26]. Cells of the immune system can be
divided into two separate branches that include the evolutionarily
ancient innate immune cell populations and the more recently
evolved adaptive cell types. Adaptive immune cells comprise of T
and B lymphocytes. Innate immune cells include phagocytic cells
(monocyte, macrophage, and neutrophils), natural killer (NK)
cells, basophils, eosinophils, mast cells, and platelets.
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_17, Springer Science+Business Media New York 2015
205
206
Materials
2.1 General
Reagents, Equipment,
and Consumables
207
Axolotl Surgery
2.4 Antibody
Staining and Flow
Cytometry
2.5 Modified
Giemsa-Wright Stain
208
Methods
It is recommended to prepare the necessary equipment and
reagents prior to performing any of the methods described in this
section. Working dilutions of any of the reagents described should
be prepared in advance unless indicated otherwise.
3.1 Time
Considerations
209
Fig. 1 Flowchart of the key steps performed to isolate and validate macrophages in the regenerating axolotl
limb. The time taken between the limb amputation and FACS is 5 days with surgery occurring on the first and
third days and tissue harvesting, processing, staining, and FACS all occurring on the fifth day
Ethics Approval
3.3 Animal
Maintenance
and Usage
210
3.5 Intravenous
Injection of High
Molecular Weight
Dextran
211
6. Load capillary glass filament with 5 mg/mL 2 MDa dextranrhodamine using a 10 L pipette tip. Ensure that no air bubbles are present within the injection volume (see Note 4).
7. Load the capillary glass filament into the microinjector.
8. Perform intravenous injection into the afferent vessel under a
dissection microscope. Set injection pressure to 20 psi and
slowly eject 5 mg/mL 2 MDa dextran-rhodamine from the
glass needle into the target vessel using small pulses (510 ms)
(see Note 5).
9. Slowly withdraw capillary glass filament from afferent vessel
minimizing any damage to the vessel or surrounding tissue.
10. Slide a gloved finger over the injection site or hold it there for
up to 30 s to accelerate clotting. This will aid in sealing the
vessel at the entry point with skin mucus and minimize additional blood loss.
11. Using a fluorescent stereomicroscope, visualize a blood vessel
in the tail to confirm delivery of the fluorescent 5 mg/mL
2 MDa dextran-rhodamine. Cells lining the vessels and within
the bloodstream will be fluorescent for approximately 46 h
postinjection.
12. Place the axolotl into a fresh tank of 20 % Holtfreters solution
for recovery in quiet individual housing for at least 24 h.
3.6 Blastema
Isolation and Tissue
Dissociation
212
10. Rinse the tissue culture dish with an additional 3 mL of collagenase II. Transfer into the C-tube to ensure that all cells are
collected.
11. Add enough collagenase II to the C-tube to a final volume of
10 mL.
12. Add 100 L of collagenase/dispase (100 mg/mL) to the
10 mL 2 mg/mL collagenase II solution for a final concentration of 1 mg/mL (see Note 7).
13. Cap tube with lid, invert, and swirl gently to align the tissue to
the plastic blades of the C-tube.
14. Place the inverted C-tube onto the gentleMacs apparatus
ensuring that the C-tube is locked in place.
15. Run C-tube Heart 1 protocol. Maintain inverted position and
place on ice. Process additional C-tubes if required (see Note 8).
16. Wrap C-tubes with foil and place on a roller at room temperature. Leave for 1 h to digest (see Note 9).
17. Run C-tube Heart 2 protocol and place tubes on ice with the
lid facing upward. Process additional C-tubes if required.
18. Transfer the tissue suspension within each C-tube into a fresh
50 mL tube by passing through a 100 m cell strainer (see
Note 10).
19. Rinse the C-tube twice with 10 mL HBSS-EDTA solution and
pass through a 100 m cell strainer. The final volume should
be 30 mL.
20. Centrifuge the cell suspension at 350 g for 15 min at 4 C.
21. Aspirate supernatant and resuspend the pelleted cells with
blocking buffer solution at a final concentration of no more
than 1 106 cells/100 L.
3.7 Live-Cell
Antibody Staining
for Myeloid Cells
from the Limb
Blastema
1. All staining reactions are conducted in 1.7 mL microcentrifuge tubes (see Note 11).
2. Block cells on ice away from light for at least 20 min.
3. Prepare 1.7 mL microcentrifuge tubes with the correct dilution of primary antibody: 0.5 mg/mL biotinylated Isolectin
B4 (use at a 1:50 dilution).
4. Add 100 L to each individual 1.7 mL microcentrifuge tube.
Mix by flicking or pipetting up and down.
5. Incubate on ice away from light for at least 20 min (see Note 12).
6. Wash any unbound primary antibodies with 900 L of wash
solution (HBSS-EDTA) and mix thoroughly and centrifuge at
350 g at 4 C for 5 min (see Note 13).
7. Prepare a working dilution of combined secondary antibodies:
2 mg/mL Streptavidin Alexa Fluor 488 (1:200).
213
Cytospin
Fig. 3 FACS gating strategy on single-cell suspensions of blastemas obtained at 4 days postamputation. (a)
Forward scatter profile of crude blastema cell suspension analyzed by flow cytometry. Single cells are gated
on for downstream analysis using forward scatter area (FSC-A) and forward scatter (FSC). (b) Scatter profile of
single cells and tissue debris. Tissue debris are gated out using side scatter area (SSC-A) and forward scatter
area (FSC-A). (c) Scatter profile of nonviable cells and non-debris. Nonviable cells are excluded by DAPI staining as dead cells become permeable allowing DAPI to bind to the cells nucleus. (dg) Single-stain controls for
dextran-rhodamine (DEX) and Isolectin B4 (IB4). (d) Detection of DEX+ cells in unlabeled (negative control). (e)
Detection of DEX+ cells in positive control. (f) Detection of IB4+ cells in isotype (negative control). (g) Detection
of IB4+ cells in positive control
214
Fig. 4 Detection of myeloid cells in the regenerating axolotl limb at 4 days postamputation by flow cytometry.
(a) Four distinct populations can be detected when cells are co-labeled with DEX and IB4: DEX IB4, DEX
IB4+, DEX+ IB4+, and DEX+ IB4+. (b) Side scatter area (SSC-A) and forward scatter area (FSC-A) profiles of
each of the four individual populations are shown individually as indicated
215
Fig. 5 Modified Giemsa-Wright stains on sorted cell populations. (a) IB4 Dex cells consist predominantly of
red blood cells and nonmyeloid cells. (b) IB4+ Dex cells consist of a heterogeneous population of myeloid
cells including cells with mononuclear phagocyte morphology (monocytes and macrophages) as well as cells
with polymorphonuclear morphology (neutrophils). (c) IB4+ Dex+ cells are a homogenous population of cells
displaying clear mononuclear phagocyte morphology. (d) IB4 Dex+ cells comprise of a heterogeneous population of cells displaying nonmyeloid cell morphology. This includes cells with dark round nuclei with small
spherical cytoplasm and cells with dendritic/fibroblast-like morphology displaying light colored large elliptical
nuclei with large elongated multipolarized cytoplasm. Scale bars indicate 50 m
4. Transfer slides into the slide mailer containing a working dilution of Giemsa staining solution. Leave for 15 min at room
temperature.
5. Wash by transferring slides into a slide mailer containing fresh
distilled water. Ensure that any precipitation is removed.
6. Air dry slides (see Note 16).
7. Mount slides using mounting media and coverslip.
8. Slides can then be stored or imaged using a color microscope.
Examples of the expected results for each of the FACS-isolated
cell populations in the regenerating limb are shown in Fig. 5.
3.10 Isolation of RNA
from FACS-Sorted
Cells
216
3.11 DNase1
Treatment and DNase
Inactivation
3.12
cDNA Synthesis
Volume
4 L
10 SuperScript Enzyme
Mix
2 L
X L
DEPC-treated water
To 20 L
1. Quantitative polymerase chain reaction (qPCR) assays are performed using SYBR green and analyzed using a thermal cycler
instrument.
2. cDNA samples synthesized from the RNA of cell populations
isolated by FACS at 4 days postamputation can be analyzed for
gene expression relative to whole blastema tissue at 4 days
postamputation.
3. Gene expression levels are calculated using the Pfaffl method
and is normalized using the geometric mean of at least two
housekeeping (normalizer) genes.
4. Repeat experiments at least three times from two biological replicates containing cDNA pooled from eight individual blastemas.
5. Primer sequences used in qPCR gene expression analysis are
listed in Table 1.
217
Table 1
Primer sequences used for qPCR
Primer
Forward
Reverse
RBL27
CATCAGATCAAGCAAGCAGTA
CCAATGCAGCAGTTTAGATG
B-actin
TCCATGAAGGCTGCCCAACT
TGGCGCCACATCTGATTGAT
CD11b
CTCGCCCTACTGTGGATTGT
AAGGCCACCTTCTCCAGATT
Neutrophil
elastase
CGCCTCCCTACAGTTCAGAG
ATCGGACTCATTCCATCCAA
Myeloperoxidase
TCAACAGCTGGAGAATCGTG
ATGTTGATGGCGGCTAAATC
CSF1R
CTCCAGGATGGGACTGTCAT
CCGCTTGGAGGTAGAGTCTG
MPEG1
CTTGCAAGACAGTTCGTCCA
TGGTTCGGTTTCCCAGATAG
MMP9
GCATCGTAGGATTCTCCATCA
ACCAGTGAAGGCCGTTCCGAT
VEGFR2
ACTGAAAACGCTGGGAAATG
AGAAGGTGGTGGTGTCCAAG
VCAM1
AAGACGACCGAAGTGTACGT
CCTGGTGAGTTTCAAGGTGC
ICAM1
AGCATAACGGCAAGATGGTC
GGATTTTCCCTCAATCAGCA
EpCAM
TAATGGCACAGACACCTGCT
TCTGCAACGTCATTCTCAGG
Collagen A2
ACCTGGTGGAAAGGGAGACT
GAAGATCCAGGTTCCCCATT
Fibronectin
ATTGCGTACTCCCAACTTCG
ACATGCTTCTCCCACGAGTC
3.14 Isolation
of Peripheral Blood
in the Axolotl
218
Fig. 6 Isolation of peripheral blood in the adult axolotl. Deoxygenated blood vessels in the gills can be exposed
after a pad of lint-free tissue is placed under the ventral side of the head. This allows easy access to insert the
needle of the SURFLO winged infusion set into the vessels shown in blue (color figure online)
9. Transfer blood into a 15 mL tube containing cold HBSSEDTA solution (see Note 23).
10. Slide a gloved finger over the injection site or hold it there for
up to 30 s to accelerate clotting. This will aid in sealing the
vessel at the entry point with skin mucus and minimize additional blood loss.
11. Place the axolotl into a fresh tank of 20 % Holtfreters solution, for recovery in quiet individual housing for at least 24 h.
3.15 Magnetic
Depletion of Red Blood
Cells Using EasySep
Magnets
219
220
105
10
105
IB4
0.076%
10
103
IB4
9.27%
103
102
Pan Ig
0.027%
102
Pan Ig
9.44%
0
-102
-102
0
102
103
104
105
102
103
104
105
Fig. 7 Leukocytes can be enriched from total peripheral blood following magnetic depletion of red blood cells
using the EasySep system. A comparison between normal peripheral blood (left) and leukocyte-enriched (right)
samples using flow cytometric analysis. Treatment with sodium dithionite allows for red blood cells to be depleted.
This results in an over 100-fold increase in the detection of innate (IB4) and adaptive (Pan Ig) immune cells
221
Fig. 8 Gene expression analysis on sorted cell populations using qPCR. Gene expression is relative to a whole
blastema at 4 days postamputation. DEX IB4+ and DEX+ IB4+ cells are enriched for genes restricted to the
myeloid lineage in comparison to DEX IB4 and DEX+ IB4 cells. DEX IB4+ cells are a heterogeneous
population of myeloid cells indicated by high enrichment of myeloid cells (CD11b), neutrophils (neutrophil
elastase and myeloperoxidase), and monocytes/macrophages (CSF1R). In contrast DEX+ IB4+ cells are predominantly enriched for monocyte and macrophage genes (CSF1R, MPEG1, and MRC-1)
222
Fig. 9 Gene expression analysis on sorted cell populations using qPCR. Gene expression is relative to a whole
blastema at 4 days postamputation. DEX IB4+ and DEX+ IB4+ cells are not enriched for genes commonly
expressed by endothelial cells (VEGFR2, VCAM1, and ICAM), epithelial cells (EpCAM), and fibroblasts (Col1a2
and fibronectin) in comparison to DEX IB4 and DEX+ IB4 cells
223
Notes
1. The percentage of Tricaine used to anesthetize the axolotl is
dependent on the size of the animal. It is necessary to adjust
this percentage to avoid any potential lethal doses. For
example, animals that are 3 cm (snout to tail) in size are
routinely anesthetized using 0.05 % Tricaine, whereas 0.2 %
Tricaine is used in animals that are greater than 15 cm
(snout to tail) in size.
2. It is recommended that surgeries are performed under a dissection microscope. Following amputation of the limb (proximal or distal), it is common to find that the bone/s have not
been completely removed. To prevent any bone/cartilage
protruding into the regenerating wound epithelium, it is recommended that bones be trimmed to fall in line with the plane
of amputation.
3. Choosing an injection point in an afferent vessel upstream of
branch points will assist in maintaining a continuous mixture
of the reagent into the oxygenated blood flow back to the
heart. This is especially important for fine vessels where blood
flow from other vessels downstream will help pull the blood
flow toward the heart.
4. Capillary glass filaments of 0.94 mm diameter are most suitable in our hands but other diameters can also be used.
Optimizing the parameters for pulling glass filaments should
be determined empirically and long fine needles can have their
tips snapped to an appropriate size.
5. Injection of air into the bloodstream should be avoided in all
instances.
224
6. Alternatively, isolated blastemas may be placed into microcentrifuge tubes containing 500 L of 2 mg/mL collagenase II. Using a pair of surgical scissors, users can take
advantage of the microcentrifuge tubes curved bottom to
mince the blastema into fine pieces. It is important that all
tissue pieces are cut very finely and there are no large chunks
remaining when using this method to avoid any potential
jamming of the C-tube during tissue processing stage using
the gentleMacs.
7. In addition, it is recommended that 2 L of DNase1 (1 mg/
mL) is added to the final solution to prevent any clumping
of cells.
8. Additional protocols on the gentleMacs can be utilized;
however, we have found that the two-step heart isolation protocol gives the best ratio of total viable cells.
9. The minimum time C-tubes can be placed on a roller at room
temperature is 40 min; lower incubation time decreases yield
of cells. Incubation times spanning greater than 2 h results in
a decrease in cell viability.
10. This protocol does not yield cells from the epidermal layer
(skin). Therefore, it is expected that there will be tissue that is
not digested. This will appear as small white pieces and will
not pass through the 100 m cell strainer.
11. In order to maximize cell viability, it is recommended that all
steps in this section be carried out on ice.
12. Normal time for primary antibody incubation is approximately
4060 min. We have found that this can be decreased to a
minimum of 20 min without any decrease in efficiency of antibody binding capacity.
13. More than 900 L of wash solution can be used. Additional
washes can be performed but are not necessary.
14. Normal time for secondary antibody incubation is approximately 30 min. We have found that this can be decreased to a
minimum of 10 min without any decrease in secondary antibody binding capacity to the primary antibody.
15. Cells can be fixed onto the side using 4 % paraformaldehyde.
16. Slides can be left to dry overnight and mounted the next day.
17. For this protocol we have chosen to use the commercially
available RNA isolation kit from Life Technologies. Other
RNA isolation kits may be used in this method.
18. The exact volume of lysis/binding solution used is not critical.
A general tip is that low-end volumes (100 L) be used for
fewer cells (<1,000 cells) and high-end volumes (500 L) used
for large amounts of cells (1 107 cells).
225
Acknowledgments
We thank Nadia Rosenthal and the members of her laboratory for
the advice and helpful discussions. We thank members of the
FlowCore facility at Monash University for performing FACS
experiments. This work was supported by grants from Stem Cells
Australia (Australian Research Council). The Australian
Regenerative Medicine Institute is supported by grants from the
State Government of Victoria and the Australian Government.
References
1. Tanaka EM, Reddien PW (2011) The cellular
basis for animal regeneration. Dev Cell 21:
172185
2. Harty M, Neff AW, King MW, Mescher AL
(2003) Regeneration or scarring: an immunologic perspective. Dev Dyn 226:268279
226
14.
15.
16.
17.
18.
19.
Chapter 18
Assessing Cardiomyocyte Proliferative Capacity
in the Newt Heart and Primary Culture
Hans-Georg Simon and Shannon Odelberg
Abstract
Unlike humans, adult newts possess extraordinary abilities to functionally regenerate lost and injured
organs, including cardiac muscle. The most remarkable feature of mature newt cardiomyocytes is their
ability to reenter the cell cycle, undergo cell division, and serve as a reservoir for progenitor cells. There
are, however, a number of unsolved questions concerning the cellular and molecular mechanisms that
underlie this plasticity; for example, we still lack a deeper understanding of the cell-inherent properties of
newt cardiomyocytes and to what degree they differ from their mammalian counterparts. Along with considerable morphological changes at the wound site, a striking feature shared by different regenerating tissues in the newt is an extensive and dynamic remodeling of the extracellular environment. The dynamic
signaling between cardiomyocytes and extracellular environment is of eminent importance in the control
of the differentiated state of the cell, but the molecular details remain elusive. In this chapter, we describe
methods to assess cardiomyocyte proliferation in vivo and enrich primary cardiomyocytes from newt hearts
to study their behavior, taking extracellular matrix components into consideration.
Key words Heart regeneration, Cardiomyocyte proliferation, EdU, Cardiomyocyte culture,
Extracellular environment, ECM
Introduction
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_18, Springer Science+Business Media New York 2015
227
228
Fig. 1 Time course of cardiac regeneration. Cardiac injury induces the deposition of a tenascin-C (TNC)-rich
transitional extracellular matrix (green). While the unamputated heart contains only a few cells actively synthesizing DNA (EdU = white), there is a substantial proliferative response throughout the myocardium between 7
and 35 dpa. Cardiac muscle is labeled with MF20 (red). White dotted lines demarcate the amputation site.
Scale bar: 400 m; dpa days postamputation
Long-term cultures revealed that the majority of newt cardiomyocytes can enter into S phase. Surprisingly, while all cells were
exposed to serum-containing medium, only about a third of newt
cardiomyocytes went through more than one complete cycle of
cell division [2]. Interestingly, the picture from in vivo labeling
studies after injury of the newt ventricle also indicates that only a
subset of differentiated cardiomyocytes seem to be responsible for
the increase in cell number observed upon regeneration [1]. It is
229
Materials
Prepare all solutions using ultrapure water and analytical-grade
reagents. Prepare and store all reagents at room temperature
(unless indicated otherwise). Diligently follow all waste disposal
regulations when disposing waste materials. The use of animals
must follow the guidelines of and be approved by the Institutional
Animal Care and Use Committee.
2.1 Surgical
Procedures
230
2.2 Collection
of Regenerating
Tissue
2.3 Monitoring
Proliferative
Progenitor Cells
(HABP,
231
Table 1
Primary detection reagents used for immunohistochemistry
Probe
Company
Dilution
Antigen/source
Tenascin-C
Millipore
AB19013
Rabbit
1:500
Embryonic chick
brain
Fibronectin
Sigma
F0791
Mouse
IgG1
1:200
IST-3 hybridoma
HABPbiotinylated
Calbiochem
385911
Bovine
1:300
Bovine nasal
cartilage
MF20
DSHB
MF20
Mouse
IgG2b
1:2,000 ascites;
Light meromyosin
1:300 Bioreactor
supernatant
Wilms Tumor 1
Calbiochem
CA1026
Rabbit
1:150
Human WT1
3377
Rabbit
1:200
Ser10 of human
histone H3
Table 2
Secondary detection reagents used for immunohistochemistry
Fluorophore
Company
Product no.
Secondary
Ab/target
Dilution or
concentration
Imaging
channel
AF488
Invitrogen
A11034
Anti-rabbit
1:500
Green
AF488
Invitrogen
A11029
Anti-mouse
1:500
Green
AF546
Invitrogen
A21123
Anti-rabbit
1:500
Near red
AF594
Invitrogen
A11005
Anti-mouse
1:500
Near red
AF647
Invitrogen
A21242
Anti-mouse
IgG2b
1:300
Far red
AF546
Invitrogen
S11225
Streptavidin
1:300
Near red
AF594 conjugated
azide
Invitrogen
A10270
EdU
1 ng/mL
Near red
DAPI
Roche
10236276001
Nuclear DNA
1:1,000
Blue
4. 100 % ethanol.
5. Forceps and iridectomy scissors. Sterilize in autoclave and keep
stored in 70 % ethanol during procedure.
6. 20 mL glass scintillation vials and caps. To sterilize expose to
UV light for at least 30 min.
232
Methods
Carry out all procedures at room temperature unless otherwise
specified. The use of animals must follow the guidelines of and be
approved by the Institutional Animal Care and Use Committee.
3.1 Surgical
Procedures
for Ventricular
Resection
233
Fig. 2 Newt ventricular resection surgery. (a) Cartoon demonstrating how anesthetized newts are held in place
on a perfboard with elastic cord. (b) Vertical line indicates position of ventral near-midline incision. (c) Resection
of cartilaginous sternum and exposure of pericardial sac. (d) Gentle external pressure releases the heart from
the chest cavity. (Note the two darker red atria on top and the single pale red ventricle at the bottom.) (e)
Resection of approximately 20 % of the ventricle. (f) A single suture closes the external chest wound following
ventricular resection
234
3.3 Monitoring
Proliferative
Progenitor Cells
235
236
We prepare primary cultures of newt cardiomyocytes using a modified version of previously described methods [2].
1. For each preparation, six to eight adult newts are anesthetized
as described in Subheading 3.1, step 1.
2. To minimize possible contamination, generously cover the
newt chest and forelimbs in alcohol-free povidone-iodine.
3. Expose hearts as described in Subheading 3.1, steps 3 and 4,
and with iridectomy scissors, cut and remove the single ventricle free of atrial tissue.
4. Place the ventricles quickly and carefully into a sterile 60 mm
cell culture dish with room temperature 70 % L15 media.
5. Store ventricles overnight (1218 h) at 25 C in fresh 70 %
L-15 media supplemented with 1 pen/strep/fungizone.
6. In a designated tissue culture hood, rinse sterile forceps and
iridectomy scissors in 70 % calcium- and magnesium-free PBS
(nPBS) and dissect each ventricle into four to six pieces.
7. Using the forceps, transfer dissected ventricles to a sterile scintillation vial containing 3 mL of digestion mixture [nPBS with
0.5 % trypsin, 400 U/mL type II collagenase, 0.15 % BSA,
0.3 % glucose, 1 pen/strep/fungizone].
8. Place the standing scintillation vial into a shaking water bath at
27 C and digest the heart tissue for 8 h, with a change of
enzyme cocktail every 2 h. Rotate the scintillation vial containing the dissected heart muscle at 90 rpm for the first 2 h and
60 rpm for the final 6 h. To protect cells from light during the
incubation, cover the scintillation vial with aluminum foil.
9. Following the 8-h digestion, triturate the cell suspension with
a sterile fire-polished glass Pasteur pipette for 25 min.
10. To neutralize the digestion mixture, add the cell suspension to
50 mL conical tubes containing 30 mL newt minimum essential
media (70 % MEM) with 10 % FBS and 1 pen/strep/
fungizone.
11. In a low-speed centrifuge with swinging bucket rotor (e.g.,
Beckman Allegra 6KR), centrifuge the suspension for 10 min
at 200 g to collect the cells.
12. Remove the supernatant and resuspend the sedimented cells
with 4 mL newt MEM with 10 % FBS and 1 pen/strep/
fungizone.
3.6 Enrichment
of Cardiomyocytes
and Culture of Primary
Cells
237
238
Fig. 4 Explanted newt cardiomyocytes in culture. (a) Early on in diluted cultures, cardiomyocytes display the
typical bifurcated structure. (b) Picture frame of movie displaying beating cardiomyocytes. Within 2 weeks the
cardiomyocytes reach a considerable density, interconnect, and beat in synchrony. (c and d) Throughout
in vitro culture, the cardiomyocytes maintain their contractile machinery as verified by MF20 staining. (d)
Cardiomyocytes actively synthesize DNA as determined by EdU incorporation. Scale bar for a, c, d: 50 m
Notes
1. The tail pinch test involves pinching the tail with enough force
to normally cause a newt to respond with a jerk. If the newt
fails to respond, this suggests that the newt is sufficiently anesthetized. The supine positioning test involves rolling the newt
onto its back in the supine position. A newt does not appreciate being rolled onto its back and will immediately flip its body
to the prone position if not sufficiently anesthetized.
239
Acknowledgements
We would like to acknowledge both current and former research
laboratory personnel who helped develop the protocols described
in this chapter. These individuals include Claudia Guzman and
Sarah Mercer. We thank Drs. Jeremy Brockes and Anoop Kumar
240
8.
9.
10.
11.
12.
13.
Chapter 19
Long-Term Organ Cultures of Newt Hearts
Tanja Piatkowski and Thomas Braun
Abstract
Adult newts regenerate their hearts after injury by initiating proliferation of cardiac muscle and non-muscle
cells. Mechanistic studies in vivo to analyze heart regeneration are challenging due to the long reproduction cycle of newts and the complexity of the genome. Culture of primary newt cells might offer alternative
experimental approaches, but monolayers of newt cardiomyocytes and slice cultures of newt hearts show
extensive morphological changes during cultivation. Hence, we developed a protocol to culture intact
newt hearts in vitro, avoiding major morphological changes of explanted organs during a 5-week cultivation. The model provides improved accessibility and allows manipulation of cultured organs by small
molecules and viral vectors. We found that dedifferentiation and S-phase entry of cardiomyocytes, which
are hallmarks of cardiac regeneration in vivo, can be recapitulated in cultured hearts in vitro. We reason
that long-term organ cultures of newts are a versatile tool for mechanistic studies on organ regeneration.
Key words Organ culture, Newt heart, 3D culture, S-phase labeling, Dead cell stain
Introduction
The spatial and temporal organization of a tissue is crucial for many
cellular processes such as proliferation, migration, and apoptosis
[1]. Cell-cell and cell-environment interactions play an important role
during tissue morphogenesis, homeostasis, and remodeling [2].
Tissue formation and remodeling depend on expression of receptors
and cell adhesion molecules, intracellular signaling cascades, and
mechanical junctions [3, 4], which are easily disrupted when cells
are separated from their native environment. In addition, the extracellular matrix (ECM) affects cellular behavior and gene expression
by providing a scaffold with a tissue-specific 3D ultrastructure [5]
and allowing transmission of mechanical forces formed by interactions of cells. The ECM consists of fibrous and noncollagen
components, including collagens, elastin, fibronectin, laminin, and
tenascin. Glycosamine and proteoglycans form the hydrous part
of the ECM, in which different cell types such as fibroblasts,
nerve endings, pericytes, vessels, and macrophages are located [6].
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_19, Springer Science+Business Media New York 2015
241
242
243
Fig. 1 Immunofluorescence staining of tissue sections from hearts after 14 and 35 days of ex vivo culture.
(a) Control hearts show a normal morphology after 14 days in culture as indicated by staining for F-actin,
Vimentin, and nuclei (DAPI). (b) Only slight changes in the trabeculated network of the ventricle are visible after
5 weeks of cultivation. (c and d) Thin trabeculae with scattered cardiomyocytes are visible in the damaged
region of hearts cultivated directly after injury. (e and f) Hearts placed in culture 1 day after experimental injury
show trabeculae that contain multiple dedifferentiated cardiomyocytes. Red, Vimentin; green, F-actin; blue, DAPI.
Scale bar: 75 m
Materials
244
Fig. 2 SYTOX Blue staining of cultured newt hearts. The cell membrane-impermeable dye SYTOX Blue marks
nuclei of cells with damaged membranes undergoing cell death. To generate an internal positive control, tissue
damage was induced by squeezing the aortic trunk with forceps immediately before staining (indicated by
arrows). (a, c, and e) Epi-illumination images of hearts after 2 weeks cultivation. (b, d, and f) SYTOX Blue staining of the hearts is shown in the upper panel. Cultured hearts do not show significant signs of cell death after
14 days in culture irrespective of previous experimental damage. Solid lines indicate injured regions of hearts
(c and e). Dashed lines indicate the outer borders of cultured hearts (b, d, and f). Scale bar: 500 m
2.2
Organ Removal
1. 70 % ethanol.
2. L15 Leibovitz medium: To 35 mL L15 Leibovitz medium with
GlutaMAX, add 1 mL penicillin/streptomycin solution, and
make up to 50 mL with sterile cell culture water (see Note 5).
3. Sterile scissors for decapitation, microscissors, and fine forceps
with curved and straight tips.
4. Stereomicroscope.
2.3
Cultivation
245
Fig. 3 EdU labeling of hearts after 14 days in culture. Cardiac cells enter S-phase and proliferate in vivo after
injury. Low-level EdU labeling is also present in uninjured hearts [8]. (af) EdU incorporation of cells in cultured
hearts indicating S-phase entry. (a) Control hearts showing low numbers of EdU-positive cells. (b) EdU-labeled
cardiomyocytes in uninjured control hearts. (cf) After heart injury, EdU-positive cardiac cells increase in number. Red, myosin light chain 3 (MYL3); green, EdU; blue, DAPI. White borders in the upper row indicate magnified areas. Scale bar (a, c, and e), 150 m; (b, d, and f), 25 m
2.4 Tissue Staining
Against Vimentin
and F-Actin
246
2.6 Sample
Preparation
for Cryosections
2.7 Reagents
for S-phase Labeling
and Antibody Staining
Against Myosin Light
Chain 3 (MYL3)
Methods
Two different injury models are possible in newts: (1) an immediate
ex vivo injury model, where the hearts are damaged directly before
cultivation (injury ex vivo) and (2) a delayed ex vivo injury model,
where the injury is applied 24 h before cultivation (1 day after injury).
3.1 Animal
Pretreatment, Heart
Injury, and Organ
Removal
247
3.2 Preparation
and Cultivation of Ex
Vivo Hearts
3.3 Vimentin
and F-Actin Tissue
Staining
248
1. Wash the hearts twice in APBS and fix them for 1 h with 4 %
PFA solution.
2. Wash the tissues 3 for 10 min each in APBS.
3. Incubate the samples in 20 % sucrose overnight at 4 C.
4. Thereafter, transfer the hearts to 30 % sucrose, and incubate
them for the next 48 h at 4 C.
5. For embedding, transfer the heart to a cryomold containing
OCT Tissue-Tek and incubate for 10 min at room temperature.
6. Freeze the cryomold in liquid nitrogen and store the samples
at 80 C (see Note 14).
7. Cut 6 m sections by using a cryostat microtome and mount
the sections on SuperFrost Plus slides.
8. Perform EdU labeling as described for cell culture by using the
protocol provided by the supplier, but without fixation.
9. Afterward, wash the slides 3 for 10 min in 5 % BSA solution.
10. Label the cardiomyocytes by using an antibody against MYL3.
Dilute the antibody 1:500 in 5 % BSA and 0.1 % Triton X-100 in
APBS and incubate the slides therein overnight at 4 C.
11. Wash the samples 3 for 10 min each with APBS.
249
Notes
1. Sulfamerazine inhibits folic acid synthesis and is antibacterial.
It is a fine white powder. Protect yourself and work under a
hood.
2. Alvegesic (butorphanol) solution is given as a pre- and postoperative analgesic to the newts.
3. Histoacryl is a tissue glue, which harden within seconds.
Always wear gloves and avoid skin contact.
4. Dissection instruments are sterilized by autoclaving or by incubation in 70 % ethanol for 15 min.
5. L15 Leibovitz medium is buffered by a combination of salt,
sugar, and amino acids and can be used for cultivation without
CO2/sodium bicarbonate.
6. When using PFA powder protect yourself by working under
the hood. Wear nitrile gloves, a lab coat and protective goggles. Avoid skin contact. To enhance solubility of PFA in
APBS, you might increase the temperature by using a temperature-regulated magnetic mixer, but do not exceed 50 C.
7. Triton X-100 is highly viscous solution and pipetting an exact
volume is difficult. Therefore, weigh out Triton X-100 using a
laboratory balance.
8. After 1520 min, newts should not show any swim or tail
movement, not even when the animals are lying on their backs.
Prolong the incubation time in tricaine solution, if animals
show any movements.
9. The newt skin is contaminated by bacteria, which is only
reduced after disinfection with sulfamerazine. Therefore, first
remove control hearts and hearts subjected to damage after
organ removal. Hearts that are removed 1 day after injury
should be dissected last. Separate the hearts in single tubes
during transfer to cell culture and always disinfect the dissecting stools between the procedures.
250
Acknowledgments
This work was supported by the Max-Planck-Society, the Excellence
Cluster Cardiopulmonary System (ECCPS), the University of
Giessen-Marburg Lung Center (UGMLC), the Cell and Gene
Therapy Center (CGT) of the University of Frankfurt, and the
DFG (SFB TRR81).
References
1. Haycock JW (2011) 3D cell culture: a review
of current approaches and techniques. Methods
Mol Biol 695:115
2. Pampaloni F, Reynaud EG, Stelzer EH (2007)
The third dimension bridges the gap between
cell culture and live tissue. Nat Rev Mol Cell
Biol 8:839845
3. Green JB (2008) Sophistications of cell sorting. Nat Cell Biol 10:375377
4. Griffith LG, Swartz MA (2006) Capturing
complex 3D tissue physiology in vitro. Nat Rev
Mol Cell Biol 7:211224
5. Badylak SF (2002) The extracellular matrix as a
scaffold for tissue reconstruction. Semin Cell
Dev Biol 13:377383
6. Hein S, Schaper J (2001) The extracellular
matrix in normal and diseased myocardium.
J Nucl Cardiol 8:188196
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
251
Chapter 20
In Vitro Preparation of Newt Inner Ear Sensory
Epithelia as a Model for Repair and Regeneration
Ruth R. Taylor
Abstract
The sensory hair cells of the inner ear transform sound energy into electrical signals, but are readily lost
through aging, excessive noise, and ototoxic agents. The newt provides an excellent model in which to
explore regeneration and whilst loss of hair cells from inner ear epithelia does not require whole organ
regeneration, new hair cells are generated from differentiated supporting cells that transdifferentiate without an intervening mitotic event. Here we describe the methods for maintaining the sensory epithelia in
long term culture; for the use of the aminoglycoside, gentamicin, to kill the hair cells; and for the examination of the tissue by electron microscopy or fluorescence microscopy. Demembranation of the epithelium
reveals the underlying ultrastructure of the tissue for examination by scanning electron microscopy (SEM)
and is a technique that can be utilized with immunogold labelling.
Key words Hair cell, Salamander, Notophthalmus viridescens, Stereocilia, Utricle, Saccule
Introduction
The inner ear contains the mechanosensitive sensory epithelia that
detect and transduce sound and those in the vestibular portion that
detect changes in head position and are involved in maintaining
balance and posture. The inner ear of the adult newt, Notophthalmus
viridescens comprises seven sensory epithelia: the three cristae
located within the ampullae of the semicircular canals that detect
angular motion of the head; the utricle which detects gravity and
acceleration; the amphibian papilla and saccule which are auditory
end organs and the lagena which performs mostly a vestibular
function. In some other newt species there is also a basilar papilla
but this has been lost from Notophthalmus viridescens.
The mechanosensory hair cells of the inner ear epithelia
transform sound waves and vibrations into electrical signals that
ultimately are detected and interpreted by the cortex of the brain
[1]. In mammals, the loss of these cells from the auditory system
results in a progressive and permanent deafness. Ototoxic agents,
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_20, Springer Science+Business Media New York 2015
253
254
Ruth R. Taylor
excessive noise, and aging can trigger this loss and new hair cells
are not generated. Following damage to the mammalian vestibular
system there is a very limited level of hair cell replacement [2].
These new hair cells are thought to arise from the non-sensory
supporting cells that surround each hair cell via direct phenotypic
conversion (transdifferentiation) without a mitotic event [3]. The
capacity of urodele amphibians, such as Notophthalmus viridescens,
to regenerate various body parts and tissues such as limb and the
lens has lead us to investigate the generation of new hair cells arising spontaneously following their ablation in the inner ear of this
vertebrate [4].
It has long been known that aminoglycosides, used to treat
tuberculosis, gram-negative sepsis, and many other infections, are
ototoxic and can cause deafness through the loss of hair cells and
consequent loss of spiral ganglion cells [5]. They have been used in
many inner ear studies both in lower vertebrates and in mammals.
In mammals administration of aminoglycoside such as gentamicin
by a series of systemic injections will cause permanent damage to
the cochleae and some aminoglycosides will affect the vestibular
sensory epithelia. In the newt, attempts to systemically deliver aminoglycosides to ablate hair cells have proved to be difficult as sufficiently high dosage results in death of the animal due to kidney
damage. However, in vitro organotypic long term cultures provide
a suitable preparation to examine recovery and regeneration of the
inner ear epithelia.
Materials
Adult newts are available from animal suppliers from the USA
(e.g., Charles D. Sullivan & Co.). Maintain the animals according
to the regulations of the institution and/or the guidelines of the
authorities.
2.1 Culture
of Sensory Epithelia
255
256
Ruth R. Taylor
12. Specimen support stubs for SEM. Use appropriate size for the
instrument according to manufacturers recommendations for
their particular holders.
13. Silver Paint: Electrolube Silver Conductive paint or Electrodag
Fast drying conductive paint (provides a conductive adhesive
for attaching specimens to support stub).
14. Sputter coater: platinum foil.
2.3 Demembranation
of Tissue
2.4 Immunogold
Labelling
2.5 Transmission
Electron
Microscopy (TEM)
257
Methods
258
Ruth R. Taylor
1. Fix the inner ear tissues in situ in the otic capsule. For this,
place the otic capsule in a 35 mm Petri dish and immobilize
using tweezers. Gently perfuse 2.5 % glutaraldehyde using a
syringe at the edge of the oval window taking care not to damage the labyrinth.
2. Immerse the entire otic capsule in glutaraldehyde solution for
at least 90 min at room temperature on a mechanical rotator,
or overnight at 4 C.
3. Rinse the samples three times for 10 min in 0.1 M cacodylate
buffer,
4. Post-fix the tissues in OsO4 for 2 h at room temperature.
5. To dissect the inner ear epithelial tissues from the otic capsule,
place the tissue in cacodylate buffer under a stereo microscope.
6. Gently crack open the otic capsule using a limited amount of
force at the edge of the oval window.
259
Fig. 1 (a) The bony otic capsule with oval window opened (arrow). (b) Labyrinth removed from otic capsule
with underside of the utricle shown to the left-hand side (arrow) and saccule uppermost (arrowhead). One
ampulla with crista, right-hand side. (c and d) Scanning electron micrographs of saccule. Hair bundles of the
saccular macula are partially covered with otoconial membrane (arrow) and extramacular hair bundles at the
very outer edge of the saccule are present (arrowhead). (d) Higher magnification of hair cell bundle showing
the graduated length of stereocilia and long kinocilium. (e and f) Transmission electron micrograph of hair cell
and cross section of bundle. (g) Extrastriolar region of utricular macula after demembranation: small hair
bundles are present. One bundle retains its long kinocilium. Its microtubular filaments have begun to splay.
Adjacent hair bundle has recess only at position of kinocilium. (h) Immunofluorescence of saccule.
Neurofilaments are labelled with antibody to neurofilament (green) and extend from the central macular hair
cell region out to extramacular hair cells at the edge of the saccule (arrows). Phalloidin labels the f-actin at cell
junctions and the actin-rich hair bundles (arrowhead, red) (color figure online)
260
Ruth R. Taylor
261
paraformaldehyde
with
0.05
For complete sections of the inner ear labyrinth, intact otic capsules can be sectioned following glutaraldehyde fixation and decalcification (see Note 12). However, end organs can be dissected
from the otic capsule following fixation and post-fixation with
osmium and processed for TEM as for intact otic capsules.
262
Ruth R. Taylor
After fixation and opening of the otic capsule from culture, tissues
can be processed for immunofluorescence either as an intact labyrinth or the maculae and cristae can be dissected out at the onset.
To prevent loss of the samples processing is carried out using 72 or
96 well plates. During change of solutions damage to the epithelium should be avoided by gently aspirating fluid from around the
tissue or if tissue is manipulated from one well to the next a fine
spatula is used to lift the sample from underneath.
1. Fix inner ear epithelia in situ within the otic capsule using 4 %
paraformaldehyde for 90 min. Initially, a small amount of fixative is gently perfused through the oval window using a syringe
to ensure that the fixative reaches the entire labyrinth rapidly.
2. Rinse the otic capsule 3 for 5 min each with PBS.
3. Transfer the otic capsule to a 35 mm petri dish with PBS and
place under a dissecting microscope.
263
Notes
1. Osmium tetroxide is volatile and a hazard to human health.
Handle OsO4 inside a fume hood (waste OsO4 must be collected in a marked bottle for suitable disposal).
2. Demembranation with immunogold label: following demembranation in buffer at pH 6.0 and fixation the tissue needs to
be processed in cytoskeletal buffer at pH 7.0.
3. Care should be taken to wear gloves when handling the newts
to prevent Salmonella transmission or fungal contamination.
264
Ruth R. Taylor
Both ears can be used for all experiments but it makes good
practice to use one ear as a control without ablating the hair
cells so that any effects of culturing can be considered. When
both ears are used for regeneration study, it is useful to fix one
for immunohistochemical analysis and the other for electron
microscopy. Incubators for amphibian cell culture should be
set at 25 C with 2 % CO2.
4. All further procedures should be carried out in a laminar flow
hood with sterile tools and medium to prevent infection.
5. Extensive hair cell loss is seen at 2 days post-gentamicin treatment and hair cell regeneration begins around 12 days
post-treatment.
6. As an alternative to specimen holders specifically produced for
electron microscopy, we have found that plastic capsules with
an open lattice base (such as those used to hold desiccant) suitable as they allow fluids to be drained from the samples without damage or loss of tissue. To prevent loss during the critical
point drying stage cover the capsules with copper mesh or lids
if available.
7. If required, the process can be halted at 70 % ethanol stage and
left overnight at 4 C.
8. Care should be taken that sufficient wet paint is applied to
attach the sample but that paint does not coat any of the upper
surface of the sample.
9. It is not always easy to determine orientation for mounting
specimens. Both saccule and utricle tend to be concave and on
careful inspection of the saccule the opening to the amphibian
papilla can be seen towards one edge of the epithelium. Nerve
tracts are often visible on the underneath of both end organs.
10. Biotinylated phalloidin and gold-conjugated streptavidin can
be used in conjunction with the demembranation protocol as
per immunogold labelling.
11. Carbon coat tissue by either using appropriately equipped
sputter coater or by evaporation using appropriate device.
12. We have found that otic capsules need to be incubated for several days (up to 5) in 4.13 % EDTA to decalcify sufficiently for
sectioning. They should become more translucent and when
pressed with forceps be soft and pliable.
13. Uranyl acetate is mildly radioactive and should be handled
with care. Waste uranyl acetate should be retained in a dedicated bottle for specialist disposal.
14. This infiltration technique allows for the plastic to be initially
introduced in a less viscous form and eventually all of the sample will be infiltrated with viscous plastic support material.
265
Acknowledgement
Many thanks to Professor Andrew Forge for sharing his extensive
knowledge of electron microscopy techniques. This work was supported by Action on Hearing Loss (formerly RNID and Deafness
Research, UK).
References
1. Forge A, Wright T (2002) The molecular architecture of the inner ear. Brit Med Bull 63:524
2. Li L, Forge A (1997) Morphological evidence
for supporting cell to hair cell conversion in the
mammalian utricular macula. Int J Dev Neurosci
15:433446
3. Forge A, Li L, Nevill G (1998) Hair cell recovery in the vestibular sensory epithelia of mature
guinea pigs. J Comp Neurol 397:6988
4. Taylor RR, Forge A (2005) Hair cell regeneration in sensory epithelia from the inner ear of a
urodele amphibian. J Comp Neurol 484:
105120
5. Forge A, Schacht J (2000) Aminoglycoside antibiotics. Audiol Neuro-otol 5:322
6. Davies S, Forge A (1987) Preparation of the
mammalian organ of Corti for scanning electron
microscopy. J Microsc 147:89101
Part IV
Transgenesis in Salamanders
Chapter 21
Transgenesis in Axolotl (Ambystoma mexicanum)
Shahryar Khattak and Elly M. Tanaka
Abstract
Transgenic animals have been indispensable in elucidating and deciphering mechanisms underlying various
biological phenomena. In regeneration, transgenic animals expressing fluorescent protein genes have been
crucial for identifying the source cells for regeneration and the mechanism of blastema formation. Animals
are usually generated by manipulating their genome using various techniques at/in one cell embryo/fertilized
egg stage. Here, we describe the generation of germline transgenic axolotls (Ambystoma mexicanum) using
the I-SceI meganuclease and Tol2 transposase.
Key words Salamander, I-SceI meganuclease, Tol2 transposase, Transgenics
Introduction
The ability to generate transgenic vertebrate animals has transformed
the molecular analysis of development, adult physiology, and disease. Recently, generation of transgenic animals via the random
integration of plasmid or BAC constructs into the genome has
been applied to zebrafish and axolotl systems for the purposes of
studying regeneration. The implementation of these animals has
had a fundamental role in elucidating the cell sources of regenerating
structures, the mechanisms by which regenerating tissue forms, as
well as analysis of molecular pathways operating during regeneration [18].
Such insights have been gained in salamander limb and spinal
cord regeneration via the generation of transgenic axolotls. Axolotls
are the most easily bred salamander species in laboratory setting
throughout the year with an average spawn size of 500 eggs per
mating. Adult axolotls are kept in water with an average temperature of 1820 C (0.5 C) with a daynight cycle of 12 h. The
conductivity of water is in the range of 500750 microSiemens
(S)/cm with a pH between 7.0 and 8.0. The level of NO2 in water
should be zero and that of NO3 should be kept below 100 mg/l.
The water should be dechlorinated by overnight evaporation,
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_21, Springer Science+Business Media New York 2015
269
270
2
2.1
Materials
Equipment
Axolotl Transgenesis
271
Fig. 1 Shows the plastic mold for making the injection plate. The mold shown
here is for making a 10 cm injection plate. Reproduced from ref. [11]
272
2.2
Solutions
Methods
Axolotl Transgenesis
273
274
Fig. 3 Depicts a 10 cm injection plate and the mold used to make this injection
plate. Axolotl eggs are aligned in the grooves/channels of the injection plate to be
injected with DNA injection mix
Fig. 4 The injection needle is made by pulling a glass capillary with a micropipette puller
Axolotl Transgenesis
275
1. Collect the eggs from the plastic leaves by gently scraping them
off with a scalpel and place them in a metal sieve.
2. Rinse the eggs with 70 % Ethanol for approximately 1520 s
followed by rinsing thoroughly (but gently) with sterile water.
3. Place the eggs in a petri dish containing 1 MMR/pen-strep
solution using a transfer pipette.
4. Under stereomicroscope, manually dejelly the eggs using sharp
forceps/tweezers.
Transfer the dejellyed eggs into a clean petri dish containing
1 MMR/pen-strep solution (see Note 2).
276
Fig. 5 A 5 nL drop size is measured using the DNA injection mixed filled injection
needle using the micrometric scale under the stereomicroscope. If the drop size
is not optimal, break the needle more or adjust the pressure of the pneumatic
injection pump to obtain the 5 nL volume drop size
10. Transfer the healthy eggs into a sterile 24-well plate (one egg per
well) containing 0.1 MMR/ pen-strep solution. Let the eggs
develop for 78 days in the 24-well plate at room temperature.
11. Using a transfer pipette, gently transfer (one by one) the normally developed larvae from the wells of the 24-well plate to a
145 mm petri dish containing 0.1 MMR/pen-strep solution.
12. The larvae will start swimming after 23 days.
3.5 Transgenic
Larvae Screening
Notes
1. The in vitro transcription of Tol2 transposase should be performed in a RNAse free environment. Wipe the bench with
RNAse away or similar products. Use only DNAse-, RNAse-free
Axolotl Transgenesis
277
Chapter 22
Generating and Identifying Axolotls with Targeted
Mutations Using Cas9 RNAGuided Nuclease
G. Parker Flowers and Craig M. Crews
Abstract
The CRISPR/Cas9 RNA-guided nuclease now enables a reverse genetics approach to investigate the function
of genes of interest during regeneration in the axolotl. The process of generating the constructs necessary
for targeting a gene of interest is considerably less labor intensive than for other methods of targeted mutagenesis such as Zinc finger nucleases or Transcription activator-like effector nucleases. Here, we describe
the identification of targetable sequences in the gene of interest, the construction of unique guide RNAs,
the microinjection of these RNAs with Cas9-encoding mRNA, the selection of well-injected animals, and
an inexpensive, PCR-based method for identifying highly mutagenized animals.
Key words Transgenesis, Salamander, CRISPR, Limb regeneration, Guide RNA
Introduction
The axolotl has a long history of use as a genetic model organism,
but in recent decades it has been surpassed in use by other amphibian and aquatic model systems such as Xenopus species and zebrafish
[1]. While a number of mutant axolotl lines have been identified,
the comparatively long generation time and exceptionally large,
currently unsequenced, genome hinder genetic studies in the
axolotl. Nonetheless, better-characterized genetic model organisms lack the remarkable adult regenerative ability of the axolotl,
and because of this fascinating ability the axolotl continues to be a
premiere vertebrate model organism for regeneration research [2].
Numerous recent studies identifying transcripts and proteins
specifically upregulated in various tissues during regeneration are
providing greater insight into the molecular mechanisms governing regeneration in the axolotl [39], though the tools for investigating the function of these genes in vivo have been relatively
blunt, with ectopic overexpression and transient knockdown via
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_22, Springer Science+Business Media New York 2015
279
280
281
Materials
2.1 sgDNA
Construction
Components
2.3 Mating
Components
1. 10 L plastic tub.
2. Novaqua (Kordon).
3. Amquel (Kordon).
4. Modified 40 % Holtfreters solution (20 mM NaCl, 0.2 mM
KCl, 0.8 mM NaHCO3, 0.2 mM CaCl2, 4 mM MgSO4,
0.1 mL/L Amquel, 0.1 mL/L Novaquel, pH to 7.5).
5. Freezer bags.
6. Sealable plastic bags.
282
7. Rounded stones.
8. Plastic Foliage.
9. Male and female axolotl.
10. Fish nets.
2.4 RNA
Microinjection
Components
1. 25 75 1 mm microscope slides.
2. 100 mm 15 mm petri dishes.
3. Agarose.
4. 10 Marcs Modified Ringers (1 M NaCl, 20 mM KCl, 10 mM
MgS04, 20 mM CaCl2, 50 mM HEPES, pH 7.4, autoclaved)
use to make 1 MMR with 50 U/L PenicillinStreptomycin,
0.1 MMR with 50 U/L PenicillinStreptomycin, 1 MMR
with 20 % Ficoll and 50 U/L PenicillinStreptomycin, and
0.1 MMR with 50 U/L PenicillinStreptomycin.
5. Ficoll PM 400 (Sigma-Aldrich).
6. PenicillinStreptomycin.
7. Fine forceps (e.g., Dumont #55 Fine Forceps, Fine Science
Tools).
8. Micropipette Puller (e.g., Flaming/Brown P-87, Sutter).
9. 1 mm glass capillary with filament (e.g., TW100F-4, World
Precision Instruments).
10. Microloader tips.
11. Microinjector (e.g., Picospritzer III, Parker-Hannifin).
12. Dissecting microscope with fluorescence.
13. Hemocytometer or scale slide.
2.5 Genotyping
Components
283
Methods
3.1 sgDNA
Construction
Fig. 1 Schematic of genomic RGN target and oligonucleotides used to construct targeting vector. (a) Putative
genomic sequence with target sequence in blue and the protoadaptor motif in red. This sequence conforms to
the GG(N1N18)NGG constraints. (b) The sequence of the two oligonucleotides used to insert the targeting
sequence into the pDR274 vector. (c) After annealing, the oligos will have two 5 overhangs compatible with
the BsaI-cut ends of pDR274. One end will have a TAGG overhang, where the GG corresponds to the first two
bases of the target site. The other end will have an AAAC overhang
284
285
sgRNA Synthesis
286
3.4 Obtaining
and Microinjecting
Embryos (See Note 4)
287
angle along the edge of the dish, to make a ramp in the agarose.
Let the agarose solidify, remove the slide, and cut away any
overhanging agarose with a razor blade.
2. Prepare injection needles. Using a P-87 Flaming/Brown
Needle Puller we use Pressure = 200, Pull = 90, Velocity = 70;
however, these parameters must be optimized for the filament
being used. This produces a needle with a very long, thin tip.
3. 4836 h before start of injection, fill lidded plastic tank with
10 L of 40 % Modified Holtfreters solution.
4. Line bottom of tank with rounded stones to prevent spermatophores from sticking to surface of tank.
5. Place plastic foliage such as leaves and plants in tank (optional).
6. Place two small freezer bags in sealed plastic bags in tank to
reduce water temperature.
7. Place sexually mature female and male in tank (see Note 5).
8. The following morning, check for successful spermatophore
deposition (see Note 6).
9. Remove the male.
10. Place the female in a fresh tub in 40 % Modified Holtfreters
solution. It will typically begin to lay eggs that evening or the
following morning.
11. Remove the tip of a 3 mL transfer pipette with scissors to create an opening larger than an axolotl egg. Collect eggs by placing the female in a fresh tub of 40 % Modified Holtfreters
solution. Scrape eggs from surface and transfer to new container with transfer pipette (see Note 7).
12. Remove excess liquid from collected eggs and place in container of 1 MMR with pen-strep on ice.
13. Transfer a few eggs to a petri dish at a time, leaving the
remaining eggs to soak in 1 MMR.
14. Under a dissecting microscope, use two very sharp forceps to
remove jelly and membrane. Take care to not damage the eggs
with your forceps.
15. Discard damaged eggs, load the dejellied eggs onto the ramp,
and remove all fluid using a transfer pipette. Cover the embryos
with several milliliters of 20 % Ficoll in 1 MMR with PenStrep (see Note 8).
16. Prepare RNA injection mix. As a starting concentration, we
mix together the RNAs to a final concentration of approximately 150 ng/L of cas9 mRNA, 30 ng/L sgRNA, and
1050 ng/L of nls-GFP mRNA just prior to injecting. Store
on ice. Using microloader tips, load an injection needle with
23 L of this mixture (see Note 9).
288
17. Using fine forceps, break the tip of your injection needle to
create an opening. Insert the needle into the microinjector
capillary holder and tightly seal.
18. Using a hemocytometer or other scale bar, inject one droplet
of your injection mix over the scale. Adjust the pulse duration
so that a single pump produces a droplet of a diameter of 180
200 m (see Note 10).
19. Orient the injection ramp so that you insert the needle into the
animal pole of the embryos along the sloped side of the ramp.
A micromanipulator is not necessary for these injections.
To confirm that the needle has not become clogged during the
procedure, occasionally inject a droplet outside of the injection
media. If necessary, adjust the pulse duration and/or clip the
needle tip to produce the proper sized droplet.
20. Very carefully place the injected embryos in an incubator at
1618 C.
21. Two to three hours post-injection gently fill the injection
plate with 0.1 MMR with pen-strep. While some leakage is
normal, remove any damaged or excessively leaking embryos
(see Note 11).
22. The following morning, gently transfer the embryos into fresh
0.1 MMR with pen-strep.
23. Continue to monitor the embryos daily, removing abnormal
embryos, and replacing media (see Note 12).
24. Depending upon the concentration of injected tracer mRNA,
the embryos will be translucent enough to assess the uniformity of injection after about 1 week. Using a fluorescence
microscope, separate embryos displaying ubiquitous fluorescent tracer expression from those displaying little or no expression (see Note 13).
3.5
DNA Extraction
1. If numbers of well-injected embryos are sufficiently large, sacrifice at least two whole embryos to assess background mutation frequency. Extract DNA from individual embryos using
QIAgen DNeasy Blood and Tissue kit.
2. After several weeks, a small quantity of tissue can be removed
from the tail of an axolotl larva without deleterious effects.
Anesthetize the larva by placing it in 0.1 g/L Tricaine in 40 %
Modified Holtfreters solution in a small petri dish.
3. When the larva stops responding to touch, using a scalpel, slice
off transparent tissue in the tail fin beyond the posterior end of
the notochord while the embryo is still submerged in Tricaine.
4. Immediately transfer the larva to a fresh container of 40 %
Modified Holtfreters solution without Tricaine.
289
5. Under the microscope, retrieve the tail clip and using forceps,
place in a 50 L PCR tube.
6. Repeat for as many larvae as necessary.
7. Using a fine pipette tip, remove excess liquid from fin clip.
8. Add 50 L of 100 mM NaOH to fin clip. Vortex well, and
centrifuge briefly.
9. In a thermocycler, heat at 95 C for 20 min.
10. Vortex well.
11. Add 15 L of 1 M TrisHCl pH 7.4 to each tube and mix well
by pipetting.
3.6 Fluorescent
PCR to Detect
Somatic Mutations
(See Note 14)
1. Identify primers around target site that produce a band (excluding primer dimers) of the predicted size by PCR (see Note 15).
2. If primary PCR produces a band of the proper size and additional bands, obtain internal nested primers and confirm that
secondary amplification produces single product of the
intended size. The primer pair producing the single, final
nested product must have melting temperatures greater than
55 C (see Note 16).
3. For one of the primers producing the single nested PCR product, obtain a new copy with forward m13 sequence
(5-TCCCAGTCACGACGT-3) attached to the 5 end.
4. Set up each primary PCR as follows:
(a) 2.5 L 10 Taq buffer.
(b) 0.5 L 10 mM dNTPs.
(c) 0.5 L 10 M forward primer.
(d) 0.5 L 10 M reverse primer.
(e) 0.125 L Taq DNA polymerase.
(f) 1 L extracted axolotl genomic DNA.
(g) To 25 L with nuclease-free water.
In each genotyping run, include at least one PCR using
genomic DNA extracted from an uninjected larva or embryo and
one PCR without DNA template to test for contamination.
5. Run the PCR on a thermocycler as follows:
(a) 95 C for 30 s.
(b) 30 cycles of 95 C for 30 s, 55 C for 20 s, 68 C for 15 s.
(c) 68 C for 1 min (see Note 17).
6. Confirm that the PCRs are successful and that there is no contamination by running 5 L of product on 1 % SB gel (see Note 18).
7. If primary PCR is successful, proceed to secondary PCR. First,
prepare 11,000 dilutions of your primary PCR products
(including no DNA control) in nuclease-free water.
290
Fig. 2 Analysis of fluorescent fragment sizes after PCR of targeted gene from uninjected (a) and RGN-injected
(b) animals. While a PCR of the region of interest from DNA from a control animal produces a single peak of the
expected size (a), PCR of DNA extracted from an RGN-injected animal reveals multiple products indicating a
high deletion frequency in the target site
291
Notes
1. Several online sources provide useful EST sequencing and
cDNA sequences that can be used to identify potential target
sites. These include GenBank, Sal-site (www.ambystoma.org),
and Axolomics (www.axolomics.org). There are several constraints on the selection of genomic targets in the axolotl. First,
the CRISPR-derived RGN system can be used to target any
sequence preceding an NGG protoadaptor motif (PAM); however, the in vitro synthesis of sgRNAs using the T7 polymerase
will result in the attachment of GG to the 5 end of the
sgRNA. Thus, the method described here will allow one to
target any sequence that conforms to the structure of
5GG(N1N18)NGG (Fig. 1a). Such targets occur once in
every 128 bases of random sequence, and they can be identified in a sequence using the online resource http://zifit.partners.
org/ZiFiT/ . If such constraints do not permit the identification of targets, alternative strategies may be used, including,
but not limited to, synthesizing sgRNAs using Sp6 polymerase
which attaches GA to the 5 end of sgRNAs, synthesizing
sgRNAs with shorter targeting sequences [15], or permitting
mismatches between the 5 end of the sgRNA and the target
sequence [16]. Second, RGN targets should be capable of
being amplified for genotyping with available genomic information. Intron-exon boundaries are not clearly delineated in
cDNA-derived sequences and the average intron size in the
axolotl genome is larger than 10 kb [17]; thus, one should first
confirm that a target can be amplified with available sequence
data before proceeding to subsequent steps. Furthermore, targets should be spaced sufficiently far from primers used for
genotyping to detect deletions that may extend beyond the
targeted site.
2. Samples that are not digested by BsaI almost always contain
the correct product.
3. We strongly recommend assessing the quality of injection by
coinjecting an mRNA encoding a fluorescent protein with
Cas9 mRNA and the sgRNA to confirm that injected RNAs do
not undergo degradation and are uniformly distributed
throughout the embryo. A number of plasmids are publically
available from which such mRNAs encoding fluorescent proteins can be easily synthesized. Assuming such plasmids contain a polyadenylation signal sequence, synthesis of such
mRNAs should follow the same protocol as below. Linearize
the plasmid with an enzyme that cuts after the signal sequence
and use a mMessage mMachine kit with the proper polymerase
for synthesis.
292
293
294
19. Note that the ratio of the concentration of the nested, m13tailed primer to the reverse gene specific primer is 1:1,000.
Higher concentrations of the nested, m13-tailed primer can
result in failure of production of sufficient quantities of 6-FAMlabeled product for analysis.
20. Numerous academic research facilities provide fragment analysis for a low cost compared to standard sequencing methods.
These include the DNA Analysis Facility at Yale University
(http://dna-analysis.research.yale.edu).
21. While this method provides a rapid and inexpensive assessment
of successful mutagenesis, definitive confirmation of mutagenesis can only be achieved through cloning and sequencing of
individual PCR products.
22. Several considerations should be made in the development of
mutant lines. As RGNs may have sequence-specific off-target
effects, crossing animals that have been mutagenized using
the same sgRNA may result in illusory, off-target genotype
phenotype associations. When possible, several distinct sgRNAs
should be used to produce mutants for each gene of interest.
References
1. Malacinski GM (1989) Developmental
Genetics. In: Malacinski GM, Armstrong JB
(eds) Dev. Biol. Axolotl. Oxford University
Press, New York, NY, pp 103109
2. Voss SR, Epperlein HH, Tanaka EM (2009)
Ambystoma mexicanum, the axolotl: a versatile
amphibian model for regeneration, development,
and evolution studies. Cold Spring Harb Protoc
2009:pdb.emo128. doi:10.1101/pdb.emo128
3. Monaghan JR, Athippozhy A, Seifert AW,
Putta S, Stromberg AJ, Maden M, Gardiner
DM, Voss SR (2012) Gene expression patterns
specific to the regenerating limb of the Mexican
axolotl. Biol Open 1:937948. doi:10.1242/
bio.20121594
4. Campbell LJ, Surez-Castillo EC, OrtizZuazaga H, Knapp D, Tanaka EM, Crews CM
(2011) Gene expression profile of the regeneration epithelium during axolotl limb regeneration. Dev Dyn 240:18261840
5. Knapp D, Schulz H, Rascon CA, Volkmer M,
Scholz J, Nacu E, Le M, Novozhilov S, Tazaki
A, Protze S, Jacob T, Hubner N, Habermann
B, Tanaka EM (2013) Comparative transcriptional profiling of the axolotl limb identifies a
tripartite regeneration-specific gene program.
PLoS One 8:e61352. doi:10.1371/journal.
pone.0061352
6. Holman EC, Campbell LJ, Hines J, Crews CM
(2012) Microarray analysis of microRNA
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
295
Chapter 23
Gene Manipulation for Regenerative Studies Using
the Iberian Ribbed Newt, Pleurodeles waltl
Toshinori Hayashi and Takashi Takeuchi
Abstract
Newts provide a unique model animal for regenerative studies. An experimental model system for molecular
genetics in newts is needed in order to clarify the mechanisms of regeneration from the perspective of gene
functions. We have identified that Iberian ribbed newt (Pleurodeles waltl) is a suitable model animal for such
studies. Here we describe protocols for gene manipulation using Pleurodeles waltl.
Key words Newt, Salamander, Molecular genetics, Regeneration, Transgenesis, TALENs, Genome
editing
Introduction
Regeneration of a lost tissue in an animal is an important issue.
Urodele amphibians, such as newts have the remarkable capability
to regenerate organs, and various body parts, such as limbs, ocular
tissues, brain, spinal cord, intestine, and these animals have been
used for regeneration studies for centuries [1, 2]. However, the
identities of the genes that regulate regeneration are still unclear.
To elucidate gene function during newt regeneration, it is particularly important to establish transgenic and knockout lines, and
systematically cross these lines to study the functions of genes. For
this purpose, we chose the Iberian ribbed newt (Pleurodeles waltl)
as a model animal. Previously we have shown that P. waltl can
spawn fertilized eggs all year around in the laboratory [3]. Each
female can lay more than 150 eggs per spawning and they spawn
every 24 weeks (Table 1). In this chapter we describe methods for
artificial insemination, I-SceI mediated transgenesis, and genome
editing using P. waltl.
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_23, Springer Science+Business Media New York 2015
297
298
Table 1
Comparison of the breeding properties of P. waltl is comparable
to X. tropicalis
P. waltl
X. tropicalis
6 months ~
5 months ~
9 months ~
6 months ~
12 cm ~
44.5 cm
14 cm ~
56 cm
Egg size
1.21.5 mm
0.70.8 mm
150600
(5,000~/year)
1,0009,000
Maturation
Adult size
2
2.1
Materials
Reagents
Gene Manipulation for Regenerative Studies Using the Iberian Ribbed Newt
2.2 Vectors/
Plasmids
299
2.3
Equipment
Methods
3.1 Maintenance
of a Newt Colony
3.2 Artificial
Insemination
300
Fig. 1 Egg (a) and sperm (b) collection for the artificial insemination. (c and d) De-jellied fertilized eggs.
(c) Good quality eggs with (left )/ without (right ) an eggshell. Arrows indicate white spots on the animal pole.
(d) Examples of low quality eggs. Arrows indicate leak of cytoplasm
Gene Manipulation for Regenerative Studies Using the Iberian Ribbed Newt
3.4 Preparation
of TALEN mRNAs
(See Note 26)
301
3.5 Microinjection
(See Note 29)
302
3.6 Sorting
the Embryos
and Rearing
Notes
1. Sterilize by autoclaving and store at room temperature.
2. After autoclaving, add HEPES, CaCl2, MgSO4, and gentamicin sulfate.
3. Prepare this solution just before use.
4. Store at 20 C. It should be stable after several times of
freeze-thawing.
5. Sterilize by filtration using membrane filter and store in a clean
bottle at 4 C.
6. For efficient integration into the newt genome, sub-clone the
construct between 2 I-SceI recognition sites [4, 5]. The plasmid DNA should be prepared using an ion exchange column
(e.g., NucleoBond, TAKARA Bio or Plasmid Midi kit,
QIAGEN). Do not use silica membrane columns since they
cannot remove endotoxins. The digested plasmids should be
stored on ice and used as soon as possible.
7. Obtain or construct TALEN plasmids for your targeting genes.
Many laboratories and companies provide various types of
TALEN scaffolds. We recommend Platinum Gate TALENs
[6], as high activity had been confirmed in the new embryos
[7]. In order to synthesize the mRNAs by in vitro transcription, the plasmids must contain T7, T3, or SP6 RNA polymerase binding site.
8. We prefer to use a NANOJECT II microinjector (Drummond,
Broomall, PA). As this injector is driven not by compressed air
but by an electric motor, it is compact and easy to operate.
9. The injector head should be mounted in a suitable micromanipulator. Although MM33 would be suitable for NANOJECT
Gene Manipulation for Regenerative Studies Using the Iberian Ribbed Newt
303
304
Acknowledgments
We would like to thank Prof. Takashi Yamamoto and Dr. Tetsushi
Sakuma (Hiroshima University) for providing the TALENs.
We also thank Profs. Andras Simon and Anoop Kumar for their
Gene Manipulation for Regenerative Studies Using the Iberian Ribbed Newt
305
7.
8.
9.
10.
Part V
Gene Expression
Chapter 24
Transcriptomics Using Axolotls
S. Randal Voss, Antony Athippozhy, and M. Ryan Woodcock
Abstract
Microarray and RNA-sequencing technology now exists for the characterization of the Ambystoma mexicanum
transcriptome. With sufficient replication, these tools give the opportunity to truly investigate gene expression in a variety of experimental paradigms. Analysis of data from the Amby002 array and RNA-sequencing
technology can identify genes that change expression levels in concert with each other, which in turn may
reveal mechanisms associated with biological processes and molecular functions.
Key words Microarray, RNA sequencing, Transcriptomics, Statistics, RNA
Introduction
Transcription is a fundamental molecular process that is common to
all organisms. During transcription, an RNA molecule is synthesized from a template DNA sequence (gene). The synthesis of RNA
molecules differs in time, type, and amount across cells. The RNAs
that derive from transcription directly or indirectly orchestrate
development and maintain cellular structures and functions.
At higher levels of biological organization, RNAs regulate physiology
and behavior. Thus, the analysis of transcription is relevant to all life
scientists, and it can be studied in any organism using common
molecular methods, analytical techniques, and instrumentation.
In model genetic organisms, transcription methodologies are
routinely used to explore the functions of genes and phenotypes,
to monitor experiments, and to classify and define biological processes. Such applications will expand as costs decline, and new
approaches are developed to allow greater flexibility of experimental design and greater accuracy of RNA measurement.
In reality, transcription studies are in their infancy, and relatively
few temporal and spatial analyses of gene expression have been
accomplished in model organisms. At the same time, it is now
possible with next-generation nucleotide sequencing technology
to obtain the complete genome or transcriptome of any organism.
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_24, Springer Science+Business Media New York 2015
309
310
Materials
2.1
Animals
2.2
RNA Extraction
2.3 Affymetrix
Microarray Analysis
311
Fig. 1 (a) Query tool within Sal-Site used to search for microarray data for an individual probeset. Probesets
may be queried by Affymetrix probeset ID or gene name. (b) Display of expression profile of a sample probeset.
Data are transformed so that measurements approximate a normal distribution and thus can be analyzed by
parametric analysis. Log2 intensity values from the microarray are plotted as a function of time for probeset
axo05645, which has been annotated as matrix metalloproteinase 2
2.4 RNA-Sequencing
Analysis
312
2.5 Software
for Microarray
and RNA-Sequencing
Analysis
Methods
3.1 Microarray
Sample Preparation
313
314
15. The standard procedure for the Affymetrix 3 IVT Express kit
is used for microarray analysis. A minimum of 50 ng total RNA
is needed per GeneChip.
3.3 Basic Statistical
Analysis
315
Fig. 2 Representative frequency distribution of Log2 transformed intensity values for a single Affymetrix
GeneChip generated in JMP Genomics version 5.0. The bottom quartile is identified at 4.12. The distribution
can be used to define unexpressed and poorly expressed probesets
316
317
3.4.2 Transcript
Expression Analysis Using
RSEM/EBSeq
3.4.3 Sequence
Annotation Aided by
Nucleotide BLAST
It is not straightforward to annotate transcripts from RNAsequencing data because transcripts/isoforms and genes may not
show a 1:1 relationship. To principally address this complexity in
the absence of a sequenced genome, we suggest selecting a representative reference transcript for each component (gene). Reference
transcripts may be identified from either the isoform of greatest
length or from the isoform with highest average expression values
(TPM or FPKM) across all experimental conditions. In practice,
RSEM expression values from the transcriptome assembly may be
imported into a database (such as MySQL) and then queried to
determine the average isoform values across all experimental
conditions.
Reference transcriptome annotation may be facilitated by
means of nucleotide BLAST [24], where the reference transcriptome is queried against a BLAST database of known annotation
(such as sequences corresponding to genes on the Ambystoma
microarray probeset). Annotation can be easily assigned for BLAST
results with perfect 1:1 query-target relationships. However, deviations from this later pattern with redundant query hits may require
manual curation and sorting to determine the best BLAST match.
318
Once gene lists have been generated from statistical analysis of normalized data (e.g., using EBSeq), overrepresentation analysis of
gene lists can be performed in the same manner as in microarray
data analysis. These tools typically perform Fishers exact tests to
identify groups of genes (by assigning them based on involvement
with a specific biological term known as an ontology) that appear
in a list more often than expected by chance if a random selection
of genes of the same size as the gene list was tested. Detection of
changes in large numbers of genes all associated with the same
process can suggest a change in the process itself, which can be
further tested with downstream experiments. Recommended sites
for performing this type of analysis include DAVID [14] and
Panther [2527].
Clustering analysis can be performed on RNA-sequencing data
once care has been taken to normalize the data. A number of packages in R exist for the purpose of investigating RNA-sequencing
data, for example, DESeq [28] and edgeR [29]. The authors of
Trinity have utilized the edgeR package to analyze RNA-sequencing
data and prepare the data for clustering analysis. Note that the
default parameters within Trinity utilize Euclidean distance and
complete linkage clustering [30]. We recommend changing the
parameters to Pearson and average to detect similarities
between expression patterns while overlooking differences in magnitude of expression.
References
1. Page RB et al (2007) Microarray analysis identifies keratin loci as sensitive biomarkers for
thyroid hormone disruption in the salamander
Ambystoma mexicanum. Comp Biochem
Physiol C Toxicol Pharmacol 145:1527
2. Huggins P et al (2012) Identification of differentially expressed thyroid hormone responsive genes from the brain of the Mexican
Axolotl (Ambystoma mexicanum). Comp
Biochem Physiol C Toxicol Pharmacol 155:
128135
3. Stewart R et al (2013) Comparative RNA-seq
analysis in the unsequenced axolotl: the oncogene burst highlights early gene expression in
the blastema. PLoS Comput Biol 9:e1002936.
doi:10.1371/journal.pcbi.1002936
4. Wu CH, Tsai MH, Ho CC, Chen CY, Lee HS
(2013) De novo transcriptome sequencing of
axolotl blastema for identification of differentially expressed genes during limb regeneration. BMC Genomics 14:434. doi:10.1186/
1471-2164-14-434
5. JMP Genomics Version 5.0. (19892007) SAS
Institute, Cary, NC
6. R: a language and environment for statistical
computing. (2012) R Foundation for Statistical
Computing, Vienna, Austria
7. MySQL AB (20082009) Sun Microsystems Inc
8. Irizarry RA et al (2003) Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics
4:249264
9. Holder D, Raubertas RF, Pikounis VB, Svetnik
B, Soper K (2001). Statistical analysis of
high density oligonucleotide arrays: a SAFER
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
319
Chapter 25
Sal-Site: Research Resources for the Mexican Axolotl
Nour W. Al Haj Baddar, M. Ryan Woodcock, Shivam Khatri,
D. Kevin Kump, and S. Randal Voss
Abstract
Sal-Site serves axolotl research efforts by providing Web access to genomic data and information, and
living stocks that are reared and made available by the Ambystoma Genetic Stock Center (AGSC). In this
chapter, we detail how investigators can search for genes of interest among Sal-Site resources to identify
orthologous nucleotide and protein-coding sequences, determine genome positions within the Ambystoma
meiotic map, and obtain estimates of gene expression. In the near future, additional genomic resources will
be made available for the axolotl, including a listing of genes that are partially or wholly contained within
Bacterial Artificial Chromosome (BAC) vectors, a prioritized collection of deeply sequenced BAC clones,
chromosome-specific assemblies of genomic DNA, and transgenic axolotls that are engineered using
TALENs and CRISPRs. Also, services provided by the AGSC will be expanded to include microinjection
of user constructs into single cell embryos and distribution of axolotl tissues, DNA, and RNA. In conclusion, Sal-Site is a useful resource that generates, shares, and evolves Ambystoma associated information and
databases to serve research and education.
Key words Salamander, Limb regeneration, Transcriptome, Analysis, Genomic resources
Introduction
Salamanders are important models in basic and biomedical research.
Over the last 100 years, salamanders have figured prominently in
studies of development, neurophysiology, ecology, evolution, and
tissue regeneration. To enhance efforts in these and other research
areas, genome resource development is underway for the primary
salamander modelthe Mexican axolotl (Ambystoma mexicanum).
A bioinformatics Web-baseSal-Sitewas created to make axolotl
genomic data and information available to the community. Here
we describe Sal-Site content and best practices for extracting useful
information.
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_25, Springer Science+Business Media New York 2015
321
322
2
2.1
Sal-Site Website
Homepage
Fig. 1 The Sal-Site homepage. This hompage (A) has hyperlinks to orientation video (B ) that introduces
Ambystoma mexicanum, general information about Sal-Site funding mechanisms (C ), salamander fun facts
(D ), and the most highly accessed links of Sal-Site (E )
2.2
Axolotl Research
323
Fig. 2 Axolotl research webpage provides hyperlinks to axolotl research projects and selected publications. The
axolotl research projects include the salamander genome project, axolotl Bacterial Artificial Chromosome
(BAC) project, axolotl chromosome sequencing project, and axolotl transgenic project
324
Fig. 3 Axolotl BAC project. Alignment of genomic sequences from frog (Xenopus tropicalus), chick (Gallus
gallus), and human (Homo sapiens) to an axolotl BAC that contains early growth response 1 (egr1). The horizontal black bars show regions of sequence similarity where nucleotide alignments are greater than 50 %
identical. The white and gray bars at the top show untranslated regions and exons (Ex), respectively
325
Fig. 4 Genome resources main webpage. This webpage provides hyperlinks to resources and tools. (A) Gene
and EST Database, (B ) Genetic Map and Marker Information, (C ) NCBI-BLAST tool, and (D ) Gene Expression
326
Fig. 5 Gene and EST database. Genes of interest can be searched using the axolotl V4.0 and V3.0 transcriptome assemblies as reference databases. Searches can be conducted by typing a specific contig (A), isotig (B ),
or gene symbol identifier (C ) in the appropriate field, followed by selecting a reference database from the
drop-down menu (D )
(Fig. 6C). It is important to note that more than one contig hit
may be reported for a query; these may represent split or nonoverlapping contigs for the same gene model, or incomplete transcript
models with low quality sequence. Moreover, when more than one
contig shows sequence identity to the same region of a query, it is
possible that the contigs are paralogous genes. In this example,
two contigs have been assembled for egr1. By clicking on the
hyperlink provided for the first contig (contig316872), a gene card
appears (Fig. 7A). The gene card offers an additional hyperlink to
open a NCBI page for the corresponding orthologous human protein (Fig. 7B). The gene card page also includes a hyperlink to
PubMed that reports some of the scientific articles that are related
to the gene of interest (Fig. 7C). Clicking the contig ID hyperlink
opens a sequence annotation page that provides detailed information about the contig, including the number of ESTs in the contig
(Fig. 7D), contig nucleotide sequence (Fig. 7E), and the longest
computer predicted open reading frame (ORF) with protein
327
Fig. 6 An example search of the Gene and EST database. The gene symbol for early growth responseegr1
is typed in the appropriate field (A) and a reference database is selected; Ambystoma mexicanum V4.0 contigs
in this example (B ). After clicking the submit button, a result webpage appears showing axolotl hits (contigs)
annotated for egr1 (C )
328
Fig. 7 An example gene card for Ambystoma mexicanum erg1. (A) The Sal-Site gene card for egr1 provides a
hyperlink to human EGR1 at NCBI (B ). The gene card page also includes a hyperlink to access related PubMed
scientific articles (C ). The gene card also provides links of the axolotl contigs annotated for erg1. Clicking the
first link (contig316872) accesses an annotation webpage that details the number of ESTs associated with the
contig (D ), contig nucleotide sequence (E ), and the longest computer predicted open reading frame (ORF) with
protein translation (F ). Additionally, this sequence annotation webpage contains data for BLAST hits linked to
human sequences curated by NCBI, including sequence similarity and alignment statistics (G ). In this example,
EST depth and sequence coverage provide very good evidence that contig316872 corresponds to egr1
2.3.3
BLAST
329
Fig. 8 Axolotl genetic map and marker information. This webpage details information about the ordering of
917 genetic markers among 17 linkage groups (LG). A part of LG3 is shown as an example (A ). A conserved
synteny table provides the positions of orthologous loci from human, axolotl, chicken, and Xenopus tropicalis
genomes (B )
The first portion of BLAST output contains header information (Fig. 10 I) that reports the algorithm used for the BLAST
search task (e.g., blastn) and the database that has been searched.
The second portion of BLAST output (Fig. 10 II) displays the
length of the query input sequence and a graphical representation
of the BLAST results summarizing the top hits (i.e., a sequence
330
Fig. 9 The Sal-Site NCBI-BLAST tool is used to identify axolotl contigs that share sequence identity with query
sequences. As an example, the human egr1 (gi|31317226) nucleotide sequence is used as a query and pasted
into the appropriate field (A ). Alternatively, the query sequence can be uploaded from a file (B ). The BLAST can
be performed using one of several algorithms (C ); the BLASTN algorithm is selected here. The reference database is chosen through a drop-down menu (D )
331
Fig. 10 BLAST output. The human egr1 nucleotide sequence (gi|31317226) was used as a query to identify the
most likely axolotl orthologous sequence in the V4 transcript assembly. (I) The report shows the algorithm that
was used for the BLAST search and the database that was searched. (II) A graphical representation of results
that summarizes the top hits to the query sequence. The color bar shows the alignment score. Each of the bars
below the query corresponds to a hit. By moving the cursor over these hit bars, it is possible to display a short
summary of the sequence ID and alignment
Gene expression technologies have advanced nearly all areas of biological research, providing, for example, greater insight into molecular pathways underlying biological processes. A number of
advanced molecular tools for gene expression analysis (including
microarray, quantitative-pcr, RNA-seq, and Nanostring) are being
used in axolotl studies. Sal-Site provides information that is useful
for conducting microarray analyses using a custom Affymetrix
GeneChip that is available for purchase from Affymetrix (part
332
Fig. 11 Continuation of BLAST output from Fig. 10. (III) Summary of information for each hit (contig), including
length, score, and E-value (AD ). The score (C) measures similarity between the query and the hit sequence.
The E-value (D ) is an index of the statistical significance of the hit reported. (IV) A detailed pairwise alignment
of the query to each of the recovered hits. The header (E ) is a repetition of the header information in the previous output section. This final output also provides the identities and the strand directionality of both sequences
for which the alignment was performed (F ). Identities refer to the number and percentage of identical nucleotides in the alignment. The pairwise alignment (G ) provides the coordinates of the query and hit sequences
that are aligned (i.e., nucleotides positions) in addition to a vertical line series that represent identical nucleotides shared between both sequences. As shown, the best hit recovered for human egr1 is contig316872
333
Fig. 12 Gene expression during axolotl limb regeneration. This webpage provides a searchable database of
gene expression profiles during axolotl limb regeneration using gene symbol or Affymetrix probe ID. In addition,
axolotl microarray library, probe annotation, and raw microarray data files are provided
Approximately 150 years ago, Mexican axolotls (Ambystoma mexicanum) were collected from their ancestral aquatic habitats near
present day Mexico City to establish laboratory populations in
Europe and subsequently the US. The AGSC at University of
Kentucky houses a historically significant population of axolotls
that was founded by Rufus R. Humphrey in 1936, and successfully
propagated by the University of Indiana through 2005. Over time,
domestication created homogeneous wild type and mutant stocks
that are amendable to laboratory culture and breeding. These
stocks provide a sustainable resource for research and educational
efforts around the world. The AGSC operates as a premier animal
334
Fig. 13 Comprehensive microarray analysis of gene expression during axolotl limb regeneration is available on
the Sal-Site genome resources page. (I) The cartoon shows regeneration stages after axolotl limb amputation
(A). The stages include pre-bud (PB), early bud (EB), medium bud (MB), late bud (LB), palette (P), digital outgrowth (DO), and completed (C). The vertical line shows the plane of amputation. The DPA (days postamputation) numbers shows when individuals were observed for each regeneration stage. (II) Temporal gene
expression profile for egr1 during the first 28 days of limb regeneration
resource center providing A. mexicanum embryos, larvae, juveniles, adults to laboratories and classrooms nationally and internationally. Essentially all laboratory axolotls in the USA, and the
majority if not all axolotls in labs around the world, descend from
the AGSC collection. The AGSC also supplies animals and display
aquaria to museums, zoos, workshops, and aquariums. Sal-Site
provides contact information for AGSC personnel, pricing for axolotl stocks (embryos, larvae, juveniles, adults, and transgenics),
axolotl care supplies, information about animal care and use, ordering, and answers to frequently asked questions (Fig. 14).
2.5
Education
Fig. 14 The Ambystoma Genetic Stock Center (AGSC). Sal-Site provides a portal to the AGSC, where researchers and educators can purchase axolotls and supplies
Fig. 15 The Education hyperlink. Sal-Site provides useful information for working with axolotls, including information about axolotl care, use, and development
336
References
1. Putta S, Smith JJ, Walker JA, Rondet M,
Weisrock DW, Monaghan JR, Kump DK, King
DC, Maness NJ, Habermann B, Tanaka EM,
Bryant SV, Gardiner DM, Parichy DM, Voss
SR (2004) From biomedicine to natural history research: EST resources for ambystomatid
salamanders. BMC Genomics 5:54
2. Smith JJ, Putta S, Walker JA, Kump DK,
Samuels AK, Monaghan JR, Weisrock DW,
Staben C, Voss SR (2005) Sal-Site: integrating
new and existing ambystomatid salamander
research and informational resources. BMC
Genomics 6:181
3. Voss SR, Kump DK, Putta S, Pauly N, Reynolds
A, Henry RJ, Smith JJ et al (2011) Origin of
amphibian and avian chromosomes by fission,
fusion, and retention of ancestral chromosomes. Genome Res 21:13061312
4. Smith JJ, Putta S, Zhu W, Pao GM, Verma IM,
Hunter T, Bryant SV, Gardiner DM, Harkins
TT, Voss SR (2009) Genic regions of a large
salamander genome contain long introns and
novel genes. BMC Genomics 10:19
5. Voss SR, Prudic KL, Oliver JC, Shaffer HB
(2003) Candidate gene analysis of metamorphic timing in ambystomatid salamanders. Mol
Ecol 12:12171223
6. Dixon JE, Allegrucci C, Redwood C, Kump
DC, Bian Y, Chatfield J, Chen Y-S, Sottile V,
Voss SR, Alberio R, Johnson AD (2010)
Axolotl Nanog activity in mouse embryonic
stem cells demonstrates that ground state
pluripotency is conserved from urodele
amphibians to mammals. Development 137:
29732980
7. Tapia N, Reinhardt P, Duemmler A, Wu G,
Arazo-Bravo MJ, Esch D, Greber B, Cojocaru
V, Rascon CA, Tazaki A, Kump K, Voss R,
8.
9.
10.
11.
12.
13.
14.
Tanaka
EM,
Schler
HR
(2012)
Reprogramming to pluripotency is an ancient
trait of vertebrate Oct4 and Pou2 proteins. Nat
Commun 3:1279
Voss SR, Shaffer HB (1997) Adaptive evolution via a major gene effect: paedomorphosis in
the Mexican axolotl. Proc Natl Acad Sci USA
94:1418514189
Voss SR, Smith JJ (2005) Evolution of salamander life cycles: a major-effect quantitative
trait locus contributes to discrete and continuous variation for metamorphic timing. Genetics
170:275281
Johnson CK, Voss SR (2013) Salamander paedomorphosis: linking thyroid hormone to life
history and life cycle evolution. Curr Top Dev
Biol 103:229258
Page RB, Boley MA, Kump DK, Voss SR
(2013) Genomics of a metamorphic timing
QTL: met1 maps to a unique genomic position
and regulates morph and species-specific patterns of brain transcription. Genome Biol Evol
5:17161730
Altschul SF, Gish W, Miller W, Myers EW,
Lipman DJ (1990) Basic local alignment search
tool. J Mol Biol 215:403410
Huggins P, Johnson CK, Schoergendorfer A,
Putta S, Bathke AC, Stromberg AJ, Voss SR
(2012) Identification of differentially expressed
thyroid hormone responsive genes from the
brain of the Mexican Axolotl (Ambystoma mexicanum). Comp Biochem Physiol C Toxicol
Pharmacol 155:128135
Irizarry RA, Hobbs B, Collin F, Beazer Barclay
YD, Antonellis KJ, Scherf U, Speed TP (2003)
Exploration, normalization, and summaries of
high-density oligonucleotide array probe level
data. Biostatistics 4:249264
Chapter 26
Data Mining in Newt-Omics, the Repository
for Omics Data from the Newt
Mario Looso and Thomas Braun
Abstract
Salamanders are an excellent model organism to study regenerative processes due to their unique ability to
regenerate lost appendages or organs. Straightforward bioinformatics tools to analyze and take advantage
of the growing number of omics studies performed in salamanders were lacking so far. To overcome this
limitation, we have generated a comprehensive data repository for the red-spotted newt Notophthalmus
viridescens, named newt-omics, merging omics style datasets on the transcriptome and proteome level
including expression values and annotations. The resource is freely available via a user-friendly Web-based
graphical user interface (http://newt-omics.mpi-bn.mpg.de) that allows access and queries to the database
without prior bioinformatical expertise. The repository is updated regularly, incorporating new published
datasets from omics technologies.
Key words Newt, Salamander, Database, Transcriptomics, Proteomics
Introduction
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0_26, Springer Science+Business Media New York 2015
337
338
Data Mining in Newt-Omics, the Repository for Omics Data from the Newt
339
Fig. 1 Flow chart of information processing in the newt data repository. The newt repository is generated by
automated pipelines for multiple raw data formats that perform an extracttransformload (ETL) process to fill
the database. Each part of the database is updated independently of other parts facilitating the update process
to include new datasets
340
1.3 Assignment
of Annotations
to Sequence Data
Data Mining in Newt-Omics, the Repository for Omics Data from the Newt
341
Subsequent to transcript annotation, we assigned transcripts to different functional groups. Since Gene Ontology (GO) annotations
for amphibians are limited [17], we used the UniProt BLAST
results from mouse, zebra fish, cow, human, and chicken. GO
annotations for each transcript were stored in a taxon-dependent
manner. We stored the best hit for each taxon having at least one
GO annotation. Further, we used functional domains, interaction
partners, protein families, and pathways from the UniProt entries.
Materials
To provide access to the freely accessible newtomics resource, we
implemented a PhP Web-based graphical user interface (GUI).
This can be used by any user by pointing any browser (GUI optimized for Firefox) to newtomics.mpi-bn.mpg.de.
Methods
3.1 General
Considerations
342
Fig. 2 Design of the database. The database is divided into five main parts, namely, the sequence part, the
annotation part, the functional annotation part, the expression part, and the proteome part. The central element
is the transcript that interlinks all other sections. Queries on multiple parts of the database utilize the central
table. The scheme can get upgraded easily by adding new tables with transcript IDs as foreign key
Data Mining in Newt-Omics, the Repository for Omics Data from the Newt
343
Newt-omics
BLAST
Server
Transcripts
Peptides
Expression
Search Module
- GO terms / Interpro
- Uniprot / GeneSymbol
- Pathway / keywords
- BLAST / eValues
- eValue
- different Blast dbs
- wordsize
Transcript result
Result / Alignment
- modifications
- length of seq
- mass
- mascot score
Result
- time-points
- pValue
- Fold Change
- Time-course
Result heatmap
Fig. 3 Modes of database assessment. The database can be accessed by the graphical user interface (GUI) via
the main search forms, focusing on the transcript itself, the sequence similarity (BLAST), the proteome
(Peptides), or the expression values. Each search form offers specific search parameters in the corresponding
search module. Each search module generates a specific result table or visualization. The single entities of the
result structure are linked to the single transcript view, presenting all available information for a single database entry
344
Fig. 4 Transcript view of the Web-based GUI of the newt-omics repository. Section (a) gives detailed information about the sequence, sequence download options, and information about the subsequences used in an
alignment step. Section (b) gives an overview on detected sequence similarities. The view can be filtered for
single taxa, and a mouse-sensitive graph gives positional information on single alignments. Section (c) represents identified functional annotations such as Gene Ontology terms or known pathways. Section (d) illustrates
all quantitative measurements that were performed for the chosen transcript. The expression values are visualized by heat maps and line plots. Section (e) visualizes the coding potential of transcripts under investigation.
The complete sequence as well as detected open reading frames are visualized; detected peptides are marked
on the corresponding position
Data Mining in Newt-Omics, the Repository for Omics Data from the Newt
345
346
Protocols
Data Mining in Newt-Omics, the Repository for Omics Data from the Newt
347
Fig. 5 Web-based GUI of the newt-omics repository. The GUI presents the main entrance panels available for
queries of the repository. Screenshot (a) presents the search form based on single transcripts, screenshot (b)
presents the BLAST interface for homology searches, screenshot (c) presents the expression based search
form, and screenshot (d) illustrates the peptide search form. All search results will link to single transcripts
matching the search query, visualized by the central transcript view (Fig. 4)
348
Data Mining in Newt-Omics, the Repository for Omics Data from the Newt
349
Notes
1. The transcriptome assembly presented in the database is based
on an EST sequencing approach. Each assembled transcript is
derived from one or more EST sequences. The OVERVIEW
OF ESTS table gives information about the number of ESTs
that contributed to a transcript as wells as the number of spots/
replicates that are represented in the microarray study, since all
ESTs were used as spots on the custom-made arrays. You might
use the number of spots representing an individual sequence
but were not included in the array analysis (panel expression)
to estimate the quality/expression level of these spots. If a
large proportion of sequences is missing, low-level expression
of the EST can be assumed.
2. The Show annotation overview view is useful to analyze similarity hits provided for a transcript of interest. First, you should
check the taxa distribution for widely explored mammalians
such as human and mouse. Next, you compare this percentage
to the percentile for close relatives of the newt such as
Xenopus and axolotl. If you see a large proportion of hits for
mammalians, you will obtain sequences with high similarity
and robust annotation. In case you receive a high percentage
of close relatives, you will also see closely related sequences,
but the quality of annotations will be low.
3. Multiple BLAST hits for multiple taxa are stored in the
database. These hits are presented in the annotation panel of
350
Acknowledgment
This work was supported by the Max-Planck-Society, the Excellence
Cluster Cardiopulmonary System (ECCPS), the University of
Giessen-Marburg Lung Center (UGMLC), the Cell and Gene
Therapy Center (CGT) of the University of Frankfurt, the DFG
(SFB TRR81), and the German Center for Cardiovascular Research
(DZHK). The authors would like to thank Marc Bruckskotten for
his invaluable input for creating newt-omics and for preparation of
the figures.
Data Mining in Newt-Omics, the Repository for Omics Data from the Newt
351
References
1. Looso M (2014) Opening the genetic toolbox
of niche model organisms with high throughput techniques: novel proteins in regeneration
as a case study. Bioessays 36(4):407418
2. Sousounis K, Michel CS, Bruckskotten M,
Maki N, Borchardt T, Braun T, Looso M,
Tsonis PA (2013) A microarray analysis of gene
expression patterns during early phases of newt
lens regeneration. Mol Vis 19:135145
3. Mercer SE, Cheng CH, Atkinson DL, Krcmery
J, Guzman CE, Kent DT, Zukor K, Marx KA,
Odelberg SJ, Simon HG (2012) Multi-tissue
microarray analysis identifies a molecular signature of regeneration. PLoS One 7(12):e52375.
doi:10.1371/journal.pone.0052375
4. Wu CH, Tsai MH, Ho CC, Chen CY, Lee HS
(2013) De novo transcriptome sequencing of
axolotl blastema for identification of differentially expressed genes during limb regeneration. BMC Genomics 14:434. doi:10.1186/
1471-2164-14-434
5. Rao N, Jhamb D, Milner DJ, Li B, Song F,
Wang M, Voss SR, Palakal M, King MW,
Saranjami B, Nye HL, Cameron JA, Stocum
DL (2009) Proteomic analysis of blastema formation in regenerating axolotl limbs. BMC
Biol 7:83. doi:10.1186/1741-7007-7-83
6. Stewart R, Rascon CA, Tian S, Nie J, Barry C,
Chu LF, Ardalani H, Wagner RJ, Probasco
MD, Bolin JM, Leng N, Sengupta S, Volkmer
M, Habermann B, Tanaka EM, Thomson JA,
Dewey CN (2013) Comparative RNA-seq analysis in the unsequenced axolotl: the oncogene
burst highlights early gene expression in the
blastema. PLoS Comput Biol 9(3):e1002936.
doi:10.1371/journal.pcbi.1002936
7. Boser A, Drexler HC, Reuter H, Schmitz H,
Wu G, Scholer HR, Gentile L, Bartscherer K
(2013) SILAC proteomics of planarians
identifies Ncoa5 as a conserved component
of pluripotent stem cells. Cell Rep 5(4):
11421155
8. Sandmann T, Vogg MC, Owlarn S, Boutros
M, Bartscherer K (2011) The head-regeneration transcriptome of the planarian Schmidtea
9.
10.
11.
12.
13.
14.
15.
16.
17.
mediterranea.
Genome
Biol
12:R76.
doi:10.1186/gb-2011-12-8-r76
Chapman JA et al (2010) The dynamic genome
of Hydra. Nature 464:592596
Borchardt T, Looso M, Bruckskotten M, Weis P,
Kruse J, Braun T (2010) Analysis of newly established EST databases reveals similarities between
heart regeneration in newt and fish. BMC
Genomics 11:4. doi:10.1186/1471-2164-11-4
Looso M, Borchardt T, Kruger M, Braun T
(2010) Advanced identification of proteins in
uncharacterized proteomes by pulsed in vivo
stable isotope labeling-based mass spectrometry. Mol Cell Proteomics 9:11571166
Looso M, Michel CS, Konzer A, Bruckskotten
M, Borchardt T, Kruger M, Braun T (2012)
Spiked-in pulsed in vivo labeling identifies a new
member of the CCN family in regenerating
newt hearts. J Proteome Res 11:46934704
Looso M et al (2013) A de novo assembly of
the newt transcriptome combined with proteomic validation identifies new protein families
expressed during tissue regeneration. Genome
Biol 14:R16. doi:10.1186/gb-2013-14-2-r16
Sousounis K, Looso M, Maki N, Ivester CJ,
Braun T, Tsonis PA (2013) Transcriptome
analysis of newt lens regeneration reveals distinct gradients in gene expression patterns.
PLoS One 8(4):e61445. doi:10.1371/journal.
pone.0061445
Elias JE, Gygi SP (2007) Target-decoy search
strategy for increased confidence in large-scale
protein identifications by mass spectrometry.
Nat Methods 4:207214
Bruckskotten M, Looso M, Reinhardt R,
Braun T, Borchardt T (2012) Newt-omics: a
comprehensive repository for omics data from
the newt Notophthalmus viridescens. Nucleic
Acids Res 40:D895900
Barrell D, Dimmer E, Huntley RP, Binns D,
ODonovan C, Apweiler R (2009) The GOA
database in 2009an integrated Gene Ontology
Annotation resource. Nucleic Acids Res 37:
D396403
INDEX
A
Accessory limb model (ALM) ..................................101113
A1 cells .....................................................................173, 182
Adaptive immune cells .....................................................205
AEC. See Apical epithelial cap (AEC)
Aeromonas ............................................................................ 40
Affymetrix Expression Console ................................312, 314
Affymetrix GeneChip .............................. 310, 314, 315, 331
Affymetrix microarray ..............................................310311
AGP. See Anterior gradient protein (AGP)
AGSC. See Ambystoma Genetic Stock Center (AGSC)
AL-1 cells ................................................. 187, 188, 190195
ALM. See Accessory limb model (ALM)
Alvegesic................................................... 175, 243, 247, 249
Ambystoma
A. maculatum ....................................... 149, 151, 152, 154
A. mexicanum .............................................. 2743, 49, 50,
116, 141145, 206, 209, 269277, 310, 316, 321,
322, 324, 325, 327, 328, 333, 334
A. tigrinum..........................................28, 30, 33, 141, 324
Ambystoma Genetic Stock Center (AGSC) ................. 28, 42,
103, 270, 310, 322, 333335
Ambystomatidae...........................................................27, 71
AMEM. See Amphibian minimal essential medium
(AMEM)
Amikacin ......................................................................38, 40
3-Aminobenzoic acid ethyl ester (MS-222) ............. 254, 257
Aminoglycosides.......................................................254, 255
Ammonium chloride ................................................230, 235
Amniotes ............................................................ 7, 9, 12, 148
Amphibian minimal essential medium
(AMEM) ......................................176, 177, 179181,
189191, 193, 254, 255
Amphibian papilla ....................................................253, 264
Amphiumids.........................................................................9
Amplexus.......................................................... 18, 24, 48, 64
Ampullae .......................................................... 253, 259, 260
Amquel ............................................................... 41, 142, 281
Aneurogenic .............................................................147155
Animal pole ......................... 64, 275, 277, 288, 300, 301, 303
Anterior gradient protein (AGP) ............................. 148, 149
Aortic arches.......................................................................31
Apateon .......................................................................... 10, 11
Apical epithelial cap (AEC) ..................................... 102, 103
Apomorphy ....................................................................8, 11
Artemia ............................. 38, 39, 41, 51, 52, 56, 60, 66, 270
Ascorbic acid ...................................................... 95, 230, 235
Autopodial elements .........................................................148
Autotomy ............................................................... 71, 73, 76
Axolomics .........................................................................291
Axolotl genome ................. 159, 280, 291, 293, 322, 324, 327
Azide .................................................230, 231, 235, 237, 239
B
Bacterial Artificial Chromosome (BAC) .......... 269, 323, 324
Basale commune ............................................................ 7, 9, 12
Basic Local Alignment Tool (BLAST) ................... 317318,
324, 325, 327332, 340, 341, 343, 347350
Benzocaine ........................................117, 119, 272, 276, 277
B1H1 cells ........................................................................182
Bicistronic.........................................................................160
Bioanalyzer ...............................................................310, 313
Biofilters .............................................................................52
Biotin-16-dUTP ..........................................................85, 87
Blackworms .................................22, 24, 38, 39, 42, 142145
BLAST. See Basic Local Alignment Tool (BLAST)
Blastema cells ........................................... 101, 102, 171, 213
Bolitoglossa ............................................................... 71, 72, 76
Brachial ...................................................... 36, 102, 106, 148
Brachial nerves .........................................................102, 148
Brine shrimps .......................... 38, 39, 41, 154, 270, 299, 302
C
Cacodylate buffer ..................................... 255, 256, 258, 261
Caecilians ...........................................................................10
Calretinin .................................................................257, 263
Capillary electrophoresis...................................................280
Cardiac troponins .............................................................228
Cardiomyocytes ........... 19, 227239, 242, 243, 245, 248249
Cas9 RNA-guided nuclease .....................................279294
Catecholaminergic neurons ................................................92
cDNA sequencing ............................................................280
Centricon .................................................................130, 132
Chromatophores .................................................................33
Chytridiomycosis ................................................................74
Ciprofloxacin ............................................................244, 254
Clodronate liposome delivery ...........................................206
Clonal density...................................................................172
Clustered Regularly Interspersed Short Palindromic Repeats
(CRISPR) ..................................................... 280, 291
Anoop Kumar and Andrs Simon (eds.), Salamanders in Regeneration Research: Methods and Protocols, Methods in Molecular
Biology, vol. 1290, DOI 10.1007/978-1-4939-2495-0, Springer Science+Business Media New York 2015
353
D
Daphnia ........................................................................22, 23
DAPI. See 4,6-diamino-2-phenylindole
(DAPI) ...........................................93, 200, 202, 207,
213, 219, 220, 231, 243, 245, 246, 248249, 257, 263
de Bruijn graph.................................................................316
Demembranation...............................256, 259, 261, 263, 264
Dextran-rhodamine ...........................207, 210, 211, 213, 220
4,6-Diamino-2-phenylindole (DAPI).......93, 200, 202, 207,
213, 219, 220, 231, 243, 245, 246, 248249, 257, 263
Dimethyl sulfoxide (DMSO) ................... 177, 181, 190, 192
Diploid genome ..................................................................31
Direct development .................................................... 5, 8, 71
Dissorophoidea ...................................................................10
DMSO. See Dimethyl sulfoxide (DMSO) .............. 177, 181,
190, 192
Dopaminergic neurons .................................................92, 93
E
EasySep magnet .................................... 208, 209, 218220
ECM. See Extracellular matrix (ECM)
Ectopic ..............................................102, 105, 116, 148, 149
transplantation ............................................................149
edgeR package ..................................................................318
EDTA. See Ethylene diaminetetraacetic acid (EDTA)
EdU. See 5-Ethynyl-2-deoxyuridine (EdU)
Electrode ..................................................... 95, 96, 118121,
123, 150, 151, 154, 161, 162, 165
Electron microscope .................................................260, 262
Electroporation....... 92, 9498, 115124, 127, 128, 148, 149,
160, 162, 165, 178, 182, 184, 188, 190, 192, 194196
Entity relationship model (ERM) ....................................339
Ependymoglial cells ................................................9294, 97
Epimorphic ................................................................19, 187
EST ........................... 291, 324328, 340, 341, 346, 349, 350
Ethylene diaminetetraacetic acid (EDTA) .............. 177, 179,
189, 206208, 211, 212, 217219, 225, 256, 264,
272, 281
F
False discovery rate (FDR) ....................................... 315, 340
Fast Green ................................................117, 119, 121, 123,
130, 134, 138, 139, 161, 162, 165
Fibrosis .......................................................................34, 206
Ficoll PM 400 ..................................................................282
FIMO clay................................................................150, 154
Flow cytometer ................................. 207, 208, 213, 220222
Fossils ......................................................................... 3, 913
G
Gelatin..................................................... 103, 104, 108112,
154, 173, 176, 177, 179181, 183, 184
bead ............................................................ 103, 109112
Gene Ontology (GO)........................315, 341, 344, 345, 350
Genome .................................... 313, 31, 47, 83, 92, 94, 116,
128, 159, 172, 269, 270, 279, 280, 291, 293, 297, 302,
304, 309311, 316, 317, 321334, 337339, 341, 342
resources ..................................................... 310, 321334
Genotyping ....................... 282, 289, 291293, 298, 302, 304
Gentamicin sulfate ....................149, 151, 175, 255, 298, 302
GentleMacs apparatus ..................................................212
Germplasm .........................................................................37
Gerobatrachus ..............................................................10, 12
GFAP. See Glial fibrillary acidic protein (GFAP)
Giemsa Stain .................................................... 208, 214, 215
Gigantism .............................................................................6
Glial fibrillary acidic protein (GFAP) ......................... 93, 94,
199, 200, 202
Glutaraldehyde .......... 105, 108, 109, 112, 255, 256, 258, 261
GPI-anchored protein ......................................................148
Green fluorescent protein (GFP) .......................... 32, 33, 93,
94, 116, 160165, 174, 192, 287
Grindal worm ................................................... 52, 56, 60, 66
H
Hanks balanced salt solution (HBSS) ..................... 206208,
211, 212, 217219, 225
Heart cultures ...................................................................247
Hematopoiesis ....................................................................31
Hemocytometer........................................ 191, 237, 282, 288
HiDI formamide ..............................................................290
Histoacryl ......................................................... 243, 247, 249
Holtfreters solution ..................................... 38, 53, 104, 117,
142, 144, 207, 209, 211, 218, 272, 281, 287, 288, 298
Human chorionic gonadotropin ..................... 37, 53, 59, 298
Hyaluronic acid ........................................................230, 239
I
Iberian ribbed newts .................................................297304
Illumina paired-end sequencing ...............................311, 316
Image J .....................................................................161, 163
Immuno-FISH .............................................................8189
Immunoglobulin .........................................................32, 222
Immunogold labelling ...................................... 256, 260, 264
Inflammatory response .....................................................205
Inflammatory signals ................................................206, 222
Ingenio .....................................................................178, 190
Inner ear ...................................................................253265
Insulin ...................................................................... 176, 189
Insulin-transferrin-selenium (ITS)........................... 189, 194
Intraventricular injection ..............................................9397
I-SceI meganuclease ......................................... 270, 272, 273
I-SceI transgenesis............................................................275
Isogroup............................................................................325
Isolectin B4 ............................... 207, 212, 213, 219, 220, 222
Isopentane ................................................................230, 234
J
JMP Genomics ................................................. 312, 314, 315
K
Keratins ............................................................................173
L
Lagena ..............................................................................253
Lateral line system ........................................................31, 51
Lentectomy ..................................................................8189
Leucistic larvae ...................................................................33
Leydig cells ...................................................................34, 35
L-15 Leibovitz medium ...........................................244, 249
Lobectomy..........................................................................36
Lymphocytes ..............................................................33, 205
Lysine ........... 85, 129, 136, 198, 200, 206, 214, 230, 235, 257
M
Macrophages ............... 33, 205, 206, 209, 215, 220223, 241
Macropinocytosis .............................................................220
Marcs modified ringers ....................................................282
Mass spectrometry....................................................338, 340
May-Grunwald stain ................................................208, 214
Membranous labyrinth .....................................................260
Mesencephalon ...................................................................34
Messenger RNA (mRNA) synthesis ................ 281, 286, 304
Microarray ....................................................... 310315, 317,
318, 323, 324, 331, 332, 334, 337, 340, 341, 348, 349
Microcapillary needle ...............................................130, 134
Microdissection ..................................................................94
Microinjection ................................................. 116124, 130,
160, 165, 282, 292, 300, 301, 303
Microloader tips ............................................ 94, 95, 97, 117,
119, 123, 130, 161, 162, 282, 287
Micromanipulator ........................................... 118120, 123,
124, 161163, 165, 271, 275, 288, 299, 301, 302
Micropipette puller .................. 94, 95, 97, 119, 271, 274, 282
MicroRNA ...............................................................159165
Microtome ..................................................................85, 248
Moloney murine leukemia virus (MMLV), 128
Monoclonal antibody ...............................................173, 219
Morpholinos .............................................................116, 280
Mutagenized animals ...............................................280, 290
Myeloid cells .................................... 206, 212215, 220223
Myofibrils .........................................................................242
Myotubes .......................................................... 172, 173, 229
MySQL database..............................................................312
N
Nanodrop spectrophotometer ...........................................313
Neotenic ............................................................. 5, 10, 27, 34
Nerve dependence ....................................................147, 148
Neural plasticity..................................................................35
Neural stem cells ................................................................91
Neurogenesis ................................................ 91, 92, 115, 197
Neuromast ..........................................................................31
Neurospheres ............................................................197203
Neurotransmission............................................................147
Neutrophils..........................................33, 205, 215, 217, 221
Newt-Omics .............................................................337350
Notophthalmus viridescens ................................. 1724, 29, 49,
50, 91, 93, 171, 253, 254, 338, 340
Novaqua ............................................................. 41, 142, 281
Novel splice isoforms ........................................................316
Nucleofection ............................178, 181184, 190, 193196
O
Olfactory epithelium ..........................................................31
Oligonucleotides .............................................. 281, 283, 324
Ontogenic transition ..........................................................71
Open reading frames ......... 129, 326, 328, 338, 340, 344, 346
Orthotopic transplantation ...............................................149
Osmium tetroxide............................................. 255, 256, 263
Osmolarity.................................173, 175, 176, 189, 198, 202
Otoconia ...................................................................258, 260
Ototoxic ...................................................................253, 254
Ovomucoid .......................................................................199
P
Pacific giant salamander .......................................................8
Paedomorphosis .........................................................10, 141
Paleozoic...................................................................4, 1013
Parabiosis ..................................................................147155
R
Red efts .................................................................. 18, 19, 23
Red fluorescent protein ............................................160, 161
Red spotted newt.......................................... 17, 19, 338, 340
Retinoid............................................................................173
Retroelements.......................................................................6
Retrotransposition ................................................................6
Retroviral .............................................. 6, 128131, 133, 136
infections ....................................................................128
Retrovirus ..................................................... 6, 127139, 188
Rhombencephalon..............................................................34
Robust Multiarray Average (RMA) algorithm .........314, 315
RNA-guided nucleases (RGNs) ........280, 283, 290, 291, 294
RNeasy Mini Kit .............................................. 310, 313314
RSEM software ................................................................317
S
Saccule ...................................................... 253, 259261, 264
Salamander embryos.................................................147155
Salamander-specific novelties .............................................12
Salamandrella keyserlingii....................................................... 8
T
TALEN. See Transcription activator-like effector nucleases
(TALEN)
293T cells ................................................. 128133, 136, 138
TCH. See Thiocarbohydrazide (TCH)
Telencephalon...............................................................34, 35
Temnospondyls...................................................................10
Tetrapods ...................................................... 712, 30, 31, 36
Tetrodotoxin .......................................................................23
TH4B cells .......................................................................173
Thiocarbohydrazide (TCH) ............................. 255, 260, 261
Thymidine ................................................................230, 235
Thyroid hormone ............................................... 29, 141, 310
Thyroid stimulating hormone (TSH) ..............................141
Thyroxine .................................................................141145
Tissue primordia ................................................................32
T7 mMessage mMachine kit ...................................281, 286
Tol2 transposase ..................................94, 270, 272, 273, 276
Tol2 transposon ..........................................................94, 116
Trabeculation ....................................................................242
Transcript .....................................................6, 135, 279, 316,
317, 324326, 331, 339, 341350
Transcription activator-like effector nucleases
(TALEN)...............................280, 299, 301, 302, 304
Transcriptome ............................................. 6, 280, 309, 316,
317, 323326, 337, 338, 340, 348350
Transdifferentiation ..............................................8189, 254
V
Vimentin .......................................................... 243, 245248
Viral vectors...................................................... 116, 128, 188
W
Winged infusion set ..........................208, 209, 217, 218, 225