<<

Progress in Retinal and Eye Research xxx (2010) 1e13

Contents lists available at ScienceDirect

Progress in Retinal and Eye Research

journal homepage: www.elsevier.com/locate/prer

Molecular and cellular aspects of lens

Jonathan J. Henry a,*, Panagiotis A. Tsonis b,* a Department of and Developmental , University of Illinois, Urbana, IL 61801, USA b Department of Biology and Center for Tissue Regeneration and Engineering, University of Dayton, Dayton, OH 45469-2320, USA

abstract

Lens regeneration among is basically restricted to some . The most notable cases are the ones that occur in premetamorphic and in adult . Frogs and newts regenerate their lens in very different ways. In frogs the lens is regenerated by transdifferentiation of the cornea and is limited only to a time before . On the other hand, regeneration in newts is mediated by transdifferentiation of the pigment epithelial cells of the dorsal iris and is possible in adult as well. Thus, the study of both systems could provide important information about the process. Molecular tools have been developed in frogs and recently also in newts. Thus, the process has been studied at the molecular and cellular levels. A synthesis describing both systems was long due. In this review we describe the process in both Xenopus and the . The known molecular mechanisms are described and compared. Ó 2010 Published by Elsevier Ltd.

Contents

1. Background ...... 00 2. Lens regeneration in Xenopus ...... 00 2.1. Morphological events ...... 00 2.2. Molecular aspects of lens regeneration in Xenopus ...... 00 2.3. Signaling factors involved in Xenopus lens regeneration ...... 00 3. Studies of lens regeneration and development in Xenopus ...... 00 4. Lens regeneration in newts ...... 00 4.1. Morphological events ...... 00 4.2. regulation ...... 00 4.3. Signaling pathways in newt lens regeneration ...... 00 4.4. ...... 00 4.5. Knock-down technology and in newts ...... 00 4.6. Transdifferentiation in newts: a model for differentiation? ...... 00 5. Xenopus and newt lens regeneration: some additional questions ...... 00 Acknowledgements ...... 00 References...... 00

1. Background remarkable because the lens is regenerated by transdifferentiation of terminally differentiated somatic cells. Regeneration of the lens in Among vertebrates the ability of regenerating the eye lens is adult newts was observed first by Collucci (1891) and independently mainly restricted to frogs and newts. Such regeneration is also by Wolff (1895), after whom the process is often called Wolffian regeneration. The process of lens regeneration in frogs was charac- terized more recently by Freeman (1963). There are, however, some * Corresponding authors. E-mail addresses: [email protected] (J.J. Henry), panagiotis.tsonis@notes. basic differences between regeneration in frogs and newts. Frogs can udayton.edu (P.A. Tsonis). normally regenerate the lens during a certain period before

1350-9462/$ e see front matter Ó 2010 Published by Elsevier Ltd. doi:10.1016/j.preteyeres.2010.07.002

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 2 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 metamorphosis and the source of the regenerated lens is the cornea a new lens will occur in the presence of the original lens provided an . On the other hand, lens regeneration in newts occurs opening is created that allows for retinal factors to reach the corneal throughout their adult life and is mediated via transdifferentiation of epithelium (Reeve and Wild, 1978; Filoni et al., 1978, 1980; Bosco et al., the dorsal iris pigment epithelial cells (PECs). A few other 1979, 1980). The process of lens regeneration can be recapitulated in can apparently replace the lens via Wolffian lens regeneration, culture (Bosco et al., 1993, 1994, 1997a,b). For example, trans- including cobitid fish (Sato, 1961; Mitashov, 1966). In this review we differentiation will occur in co-cultures of cornea epithelial and neural will outline and compare these processes mainly in both amphibians tissues (Fig. 2D). In fact, the efficiency is nearly as good, or even with a focus on the molecular and cellular aspects. In recent years, better than that which occurs in vivo. This represents a very convenient advances in molecular technology and manipulation in these animals system in which to study this process under a more controlled set of has led to important understandingof the process andits relevance to conditions. Direct contact between the cornea epithelium and the regenerative biology in general, and positions these models as neural retina does not appear to be required for lens regeneration to excellent systems in which to undertake further studies. take place, as lens cell differentiation will occur in cultures of corneal epithelium grown in retina-conditioned media (Bosco et al., 1997a). 2. Lens regeneration in Xenopus Together, these findings indicate that the retinal tissue provides diffusible factors sufficient to support lens regeneration. 2.1. Morphological events Through a variety of tissue transplantation experiments, it has been shown that only the larval cornea and the pericorneal Though not as extensive as the forms of regeneration displayed by epidermis, which includes a combined area twice the diameter of certain urodele amphibians (e.g., newts and ), some the eyecup, has the potential to form lens cells in X. laevis (Freeman, of anurans (frogs) exhibit the capacity to regenerate certain 1963). The competence of this larval tissue to respond to these structures such as the intestine, larval tail (including the spinal cord, signals is established by developmental history, which includes the and muscles), limb structures, and even parts of the eye (including the initial series of early and late embryonic lens inductive interactions retina, and lens; reviewed by Henry, 2003; Slack et al., 2004, 2008; and the presence of the eyecup (Bosco and Filoni, 1992; Cannata Callery, 2006; Tseng and Levin, 2008; Henry et al., 2008; Beck et al., et al., 2003; Arresta et al., 2005). Once competence is established, 2009; Filoni, 2009). The most widely studied species are African the continued presence of the eyecup does not appear to be required frogs of the Xenopus.Onespeciesinparticular,Xenopus laevis, to maintain this condition (Cannata et al., 2003), since the eye can be has been used for over 60 years as an experimental model system in removed late during embryogenesis or from the older larvae and the studies of development and regeneration (Kay and Peng, 1991; overlying tissues will still retain the ability to respond if transplanted Tinsley and Kobel, 1996; Gurdon and Hopwood, 2000; Beck and into the vitreous chamber. Slack, 2001; Henry, 2003; Callery, 2006; Henry et al., 2008). More The success and extent of lens regeneration gradually decreases as recently, many researchers have adopted a related species, Xenopus the larvae approach metamorphosis (Freeman, 1963; Filoni et al., tropicalis. The latter has certain advantages that include a shorter 1997). This appears to be due to the increasing rapidity with which generation time and X. tropicalis is a true diploid, in contrast to the the inner corneal endothelium heals back to cover the pupillary allotetraploid condition found in X. laevis. These features and the opening, and mayalso be related to the extent of differentiation of the smaller size favored the selection of X. tropicalis for genome cornea epithelium. Experimentally, one can demonstrate that the sequencing, which was been completed by the U.S. Department of capacity to differentiate lens cells is still present in the corneal Energy’s Joint Genome Institute (http://genome.jgi-psf.org/Xentr4/ epithelium throughout this time interval and apparently even after Xentra4.home.html). Given the recent advances in high-throughput metamorphosis, if one exposes this tissue to the key retinal factors via sequencing technology, an effort is being mounted to sequence the transplantation into the vitreous chamber (Freeman and Overton, genome of X. laevis (http://www.xenbase.org/common/), which will 1962; Filoni et al., 1997). Likewise, the factors provided by the serve as an additional resource for molecular level studies, like those neural retina are still present in adult eyes (Bosco and Willems,1992). described below. Researchers have demonstrated that corneal epithelium of post- Species in the genus Xenopus are the only frogs known to regen- metamorphic frogs can undergo transdifferentiation, if implanted erate lenses (reviewed by Henry, 2003; Filoni, 2009). Lens regeneration into the vitreous chamber of either pre- or post-metamorphic frogs in X. laevis was first described by Freeman (1963) and differs signifi- (Freeman and Overton, 1961; Bosco and Willems, 1992; Bosco and cantly from that of Wolffian lens regeneration, which has been well Filoni, 1992; Filoni et al., 1997). More recently, Yoshii et al. (2007) described for newts and salamanders (discussed below). Following observed examples of lens regeneration in adult Xenopus and sug- removal of the original lens in Xenopus , the inner layer of the gested that these may arise from small numbers of lens epithelial outer corneal epithelium undergoes a series of transformations to cells that remain in the eye following incomplete removal of the reform a new lens. Freeman sub-divided this process into 5 stages, original lens (though no attempt to trace these cells was made in that which are illustrated in Fig.1. This process has been described as one of study to prove this claim). transdifferentiation (metaplasia) of the corneal epithelium (though see X. tropicalis is also capable of regenerating lenses though the further discussion, below), and shares many similarities to that of success rate is much lower than that found in X. laevis (Fig. 2AeC; embryonic lens formation, but there are also some interesting differ- Henry and Elkins, 2001). In the former species the inner corneal ences (Henry, 2003; discussed below). Lens formation typically occurs endothelium heals more rapidlycompared to that in X. laevis, and this in the cornea epithelium that lies directly over the pupillary opening tends to cut off the critical signaling factors required to support lens and requires secreted factors provided by the neural retina (Freeman, regeneration. Another species, Xenopus borealis does not normally 1963; Filoni et al., 1981, 1982, 1983). Under normal circumstances, regenerate a lens following lentectomy, due to the rapidity with the physical presence of the inner cornea endothelium (derived from which the inner cornea heals back to cover the pupillary opening ) and the lens prevent these factors from reaching the (Filoni et al., 2006). On the other hand, the corneal epithelium of X. cornea epithelium (Freeman,1963; Filoni et al.,1997; Henry and Elkins, borealis is capable of forming a lens if implanted directly into the 2001). Removal of either barrier alone will not initiate lens regenera- vitreous chamber where it can be continuously exposed to the key tion (Bosco et al., 1980; Filoni et al., 1980; Cioni et al., 1982). Further- inducing factors. In contrast, the corneal epithelium of other more, experiments reveal that the lens and inner cornea do not species, (e.g., members of the genera Rana, Bufo, Hyla and produce inhibitors of lens regeneration. For instance, regeneration of Disscoglossus) do not appear to have the competence to respond to

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 3

Fig. 1. Cornea-lens transdifferentiation in X. laevis. Sections show different stages of lens regeneration following removal of the lens (stages follow the convention of Freeman, 1963). A. Stage 1. During this stage cells of the inner corneal epithelium assume a cuboidal shape within 24 h following lens removal. B. Stage 2. During this stage, cells begin to assume a thickened placodal arrangement. Nuclei of these cells are found to contain one nucleolus (characteristic of lens epithelial cells), rather than two (characteristic of cornea epithelial cells). C. Early stage 3. D. Middle stage 3. E. Late stage 3. During stage 3, a loosely organized aggregate of cells begins to separate from the corneal epithelium. These cells begin to organize with distinct apical-basal polarity to form a lens vesicle. F. Early stage 4. G. Middle stage 4. H. Late stage 4. During stage 4 a definitive lens vesicle is present. Elongated primary lens fiber cells have formed on the side closest to the vitreous chamber. Typically the lens vesicle is separated from the overlying corneal epithelium at this stage. I. Early stage 5. During this stage, secondary lens fiber cells are being added from the lens epithelium at the equatorial zone. Nuclei of the primary fiber cells begin to disappear. This stage is followed by further addition of secondary fiber cells and growth of the lens. ce, corneal epithelium; cn, corneal endothelium; ic, inner layer of the corneal epithelium; ir, iris; oc, outer layer of the corneal epithelium; le, lens epithelium; lf, lens fibers; rl, regenerating lens vesicle. Refer to text for further details. Figure from Henry (2003), after Freeman (1963). Scale bar equals 25 mm. these inductive signals; however, it appears that the eyes of all signals that trigger lens regeneration are either short ranged and/or frog species examined do produce the factors required to support the response is concentration dependant. The initial observations regeneration, as established in xenoplastic (cross-species) trans- made by Freeman (1963) indicate that lens-forming transformations plantation experiments (reviewed by Henry and Elkins, 2001; Henry, initially occur at multiple foci within this region (each reaching 2003 and Filoni et al., 2006). stages 1e2, refer to Fig. 1 for description of these stages), however, As mentioned above, the new lens normallyarises from the tissue only one of these foci eventually forms a lens. The other foci appear that lies directly over the pupillary opening. This suggests that the to regress. Rarely does one see the formation of multiple lenses. This

Fig. 2. In vivo and in vitro lens regeneration in living transgenic X. tropicalis and X. laevis larval tissues carrying a transgene encoding the jellyfish green fluorescent (GFP) coupled to a g-crystallin enhancer/promoter. Combined light and epifluorescence micrographs show expression of GFP (green) in the regenerating lenses of these examples. A. Eye of transgenic X. tropicalis following removal of the lens. Note absence of differentiated lens cells and GFP in the eye at one week following lens removal. B. Eye at 10 days following lens removal. Note the presence of a small regenerating lens containing GFP expressing cells. C. Eye at 21 days following lens removal. Note the growth of a larger regenerating lens containing GFP expressing cells. D. In vitro transdifferentiation in co-cultures of transgenic X. laevis corneal epithelium and non-transgenic larval eyecup. Note GFP expressing lens cells derived from the transgenic corneal epithelium. pl, pupil; tl, transdifferentiating lens cells. AeC from Henry et al. (2008), after Henry and Elkins (2001). D, from L. Fukui and J. Henry (unpublished data, University of Illinois, Champaign-Urbana). Scale bar equals 200 mm.

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 4 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 suggests that some mechanism may operate to ensure that only one (Pannese et al., 1995; Kablar et al., 1996; Cvekl and Piatigorsky, 1996; lens is ultimately formed, possibly involving some form of lateral Zygar et al.,1998; Schaefer et al.,1999; Cui et al., 2004). As is the case in inhibition (see further discussion below). other organisms, ectopic expression of Pax6 leads to the formation of ectopic eyes and lenses in Xenopus (Altmann et al., 1997; Chow et al., 2.2. Molecular aspects of lens regeneration in Xenopus 1999). Sox3 is a member of the SOX family of SRY testis determining factor family of HMG box transcription factors known to play Patterns of crystallin expression have been characterized during important roles in eye development (Penzel et al., 1997; Furuta and lens regeneration in X. laevis and some interesting differences have Hogan, 1998; Zygar et al., 1998; Ashery-Padan et al., 2000; Koster been found compared to the situation encountered during embry- et al., 2000). Sox play roles in regulating crystallin expres- onic lens formation. The use of antibodies reveals that a- and sion (Uwanogho et al., 1995; Kamachi et al., 1995, 1998, 2001; b-crystallinproteins are detected in both fibercells and lens epithelial Nishiguchi et al., 1998; Chow and Lang, 2001; Ishibashi and Yasuda, cells beginning at Freeman stage 4 of lens regeneration, while 2001). Pitx3 is a paired-like (bicoid) homeodomain containing tran- g-crystallin proteins are apparently restricted only to lens fiber cells scription factor expressed early in presumptive and placodal lens (Campbell,1965, Brahma and McDevitt,1974; Campbell and Truman, (Chang et al., 2001; Pommereit et al., 2001). Pitx3 and 1977; Reeve and Wild, 1978; Brahma, 1980; Henry and Mittleman, a related gene Pitx1 are required for normal lens and retinal devel- 1995). During embryonic lens development translation of various opment (Semina et al., 2000; Khosrowshahian et al., 2005; Shi et al., crystallins, however, is restricted to lens fiber cells, beginning around 2005; Medina-Martinez et al., 2009). Another gene, Prox1 encodes stage 29/30 of development (stages of Nieuwkoop and Faber, 1956). a prospero-like , which is initially expressed in the The transcription of certain crystallin has also been examined lens placode (Oliver et al., 1993; Tomarev et al., 1996, 1998; Schaefer during lens regeneration (Schaefer et al., 1999; Mizuno et al., 1999a; et al., 1999; Chow and Lang, 2001; Reza et al., 2002). Prox1 is impor- Malloch et al., 2009). Again, some interesting differences have been tant for lens fiber cell differentiation (Wigle et al., 1999) and serves to observed compared to the patterns of expression detected during activate crystallin expression (Cvekl and Piatigorsky,1996; Ring et al., embryonic stages. For instance, aA-, and bB1-crystallin transcripts 2000). are first detected in presumptive lens fiber cells of the regenerated Significantly, Otx2, Pax6, Sox3, and Prox1 are all expressed during lens vesicle (middle to late Freeman stage 3), and subsequently only lens regeneration in Xenopus (Schaefer et al., 1999; Mizuno et al., in differentiated lens fiber cells at later stages. g-crystallin transcripts 1999b; Cannata et al., 2003). Furthermore, Henry et al. (2002) are not detected until early Freeman stage 4 of regeneration and only showed that Pax6 is expressed in larval corneal epithelium prior to in lens fiber cells. However, during embryogenesis, aA- bB1- and g- removal of the lens. Following lens removal, Pax6 expression appears crystallin transcripts are detected simultaneously in the lens placode, to be up-regulated in the cornea and regenerating lens (Schaefer and only in differentiated lens fiber cells at later stages of develop- et al., 1999; Mizuno et al., 1999b; Cannata et al., 2003). Gargioli ment. These findings indicate that there are some differences in the et al. (2008) examined the expression of Otx2, Pax6, Sox3, Pitx3, regulation of crystallin expression during regeneration vs. develop- Prox1 and bB1-cry prior to lens removal in Xenopus. These investi- ment of the lens. In contrast, Mizuno et al. (2005) demonstrated that gators noted that only Pax6 is expressed in the larval ectoderm, and the expression of bB1-crystallin during lens regeneration requires further, its expression is restricted to the lentogenic area that includes the same promoter elements as those required during embryonic both the corneal and pericorneal ectoderm. They further demon- lens development, suggesting that elements of a shared regulatory strated that injections of exogenous Pax6 in early embryonic cells (at network appear to be operating in both of these lens-forming the 2-cell stage) trigger ectopic, endogenous Pax6 expression. processes. Presumably this occurred through auto-regulation of Pax6 expres- A large number of transcription factors play key roles in eye and sion, though they did not actually demonstrate that the injected, lens development (Beebe, 1994; Wride, 1996; Cvekl and Piatigorsky, exogenous message was translated. This treatment, however, was 1996; Graw, 1996, 2003; Fini et al., 1997; Oliver and Gruss, 1997; apparently sufficient to impart lentogenic capacity in flank Tomarev, 1997; Filoni et al., 1997; Jean et al., 1998; Kondoh, 1999; epidermis, as assayed by transplanting this tissue inside the vitreous Lupo et al., 2000, 2006; Ogino and Yasuda, 2000; Wawersik and chamber. These findings suggest that expression is linked with Maas, 2000; Hanson, 2001; Chow and Lang, 2001; Zhang et al., the acquisition of lens regeneration competence in larval ectoderm. 2002; Graw and Loster, 2003; Zuber et al., 2003; Zaghloul et al., Other researchers have described patterns of Pax6 expression as 2005; Adler and Canto-Soler, 2007; Cvekl and Duncan, 2007), being linked with embryonic lens-forming competence in other including Otx2, Pax6, Sox3, Pitx3,andProx1 as well as others. For systems (e.g., chicken and rat, Li et al.,1994; Fujiwara et al., instance, Otx2 encodes an orthodenticle-related transcription factor 1994). that contains a bicoid class DNA-binding homeodomain. Otx2 acts as In order to further characterize the molecular level events that a “head-field selector” and is required for the development of the underlie the process of lens regeneration in X. laevis, a subtracted forebrain and midbrain (Chow and Lang, 2001; Ogino et al., 2008). cDNA library enriched for genes expressed during the first four days Otx2 is expressed early in embryonic head ectoderm, including the of this process was prepared (Freeman stages 1e3; Henry et al., 2002; presumptive lens ectoderm (Pannese et al., 1995; Kablar et al., 1996; Malloch et al., 2009). Representative clones were sequenced and the Zygar et al., 1998), and is required for the formation of the lens and information was submitted for BLAST analysis to characterize gene retinal pigmented epithelium in mice (Martinez-Morales et al., 2001). expression during the early stages of lens regeneration. A searchable Otx2 has recently been shown to play an important role in regulating database of information pertaining to each of these sequences is Notch induced FoxE3 expression (a forkhead transcription factor available on-line at www.life.illinois.edu/henry/EST_databases.html. related to Xenopus lens1), which is essential for lens formation A total of 734 unique genes were identified. The assemblage of genes (Kenyon et al., 1999; Ogino et al., 2008). Pax6 represents a central is typical of those associated with metazoan organismal develop- transcriptional regulator of eye development, containing two sepa- ment and includes a high concentration of genes involved in rate DNA binding domains that include a homeodomain and a paired regulating transcription and . Groups with more box (Ashery-Padan and Gruss, 2001). Pax6 is expressed early during prevalent representation include transcription factors, proteins development in a relatively broad area of anterior embryonic ecto- involved in RNA synthesis and processing, integral membrane and derm and plays specific roles in eye development and lens cell transmembrane proteins, components of a number of conserved differentiation, including the regulation of crystallin gene expression signaling pathways, proteins involved in protein processing,

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 5 transport and degradation, proteins involved in matrix remodeling (Furuta and Hogan, 1998; Belecky-Adams et al., 2002; Chow and including a number of matrix metalloproteases (MMPs), regulators of Lang, 2001; Yang, 2004; Esteve and Bovolenta, 2006; Cvekl and the cell cycle, metabolism, , inflammation and immune Duncan, 2007; Wordinger and Clark, 2007; Adler and Canto-Soler, responses, proteins involved in chromatin remodeling in addition to 2007). known lens proteins (e.g., crystallins). Using cultures of isolated larval corneal epithelium, Bosco et al. Of the transcription factors identified in this set, a number have (1994, 1997b); demonstrated that addition of FGF1 appears to be not been previously implicated in the process of lens formation; sufficient to trigger lens cell differentiation. Furthermore, Arresta though some, including eyes-absent homolog 2 and Prox1, are clearly et al. (2005) found that a commercial polyclonal antibody that important for eye/lens formation (Malloch et al., 2009). On the other recognizes the FGFR2 (bek variant) labels stage 53 larval hand, elements of several signal transduction cascades are also cornea epithelium but not dorsal head or flank epidermis (stages of present, which have previously been shown to play important roles Nieuwkoop and Faber,1956). In addition, this antibody was found to in lens development, regeneration and differentiation, including the label dorsal head epidermis if this was previously subjected to eyes Bone Morphogenetic Protein (BMP), Delta-Notch, Wnt, FGF, implanted at stage 46 of larval development. These observations link Hedgehog, TGF-b, Rho-Ras and retinoic acid signaling pathways FGFR2 expression with the acquisition of lentogenic capacity, and (Manns and Fritzsch, 1991; Hyuga et al., 1993; Kastner et al., 1994; provide further evidence that FGF signaling may be involved in lens Bosco et al., 1994, 1997b; Kodama and Eguchi, 1995; Graw, 1996; regeneration in Xenopus. The Henry lab (University of Illinois, Cvekl and Piatigorsky, 1996; Li et al., 1997; McDevitt et al., 1997; Del Urbana-Champaign) also has several lines of evidence that FGF Rio-Tsonis et al., 1997, 1998, 2000; Gopal-Srivastava et al., 1998; signaling is functionally required in vivo for lens regeneration in X. Enwright and Grainger, 2000; Wagner et al., 2000; Tsonis et al., laevis (Fukui and Henry, unpublished data). 2000, 2002, 2004; de Iongh et al., 2001; Onuma et al., 2002; In another set of studies, Dr. Caroline Beck’s lab (University of Kawamorita et al., 2002; Stump et al., 2003; Beebe et al., 2004; Ang Otago, New Zealand) has shown that over-expression of Noggin in et al., 2004; Lang, 2004; Lupo et al., 2005; Lovicu and McAvoy, transgenic Xenopus larvae inhibits lens regeneration, suggesting that 2005; Grogg et al., 2005, 2006; Chen et al., 2008). elements of the TGF-b signaling pathways, specifically those Expression patterns for most (703) of the genes identified by involving the Bone Morphogenetic Proteins (BMPs) play an impor- Malloch et al. (2009) were examined during embryonic develop- tant role in this process (Caroline W. Beck, personal communication). ment, 634 of these exhibited some embryonic expression. Signifi- As mentioned above, the early observations of Freeman (1963) cantly, 57.7% of the latter are expressed in developing eye tissues and suggest that multiple, small populations of cells scattered over the nearly half (46.8%) included the developing lens. These findings pupillary opening begin to undergo transdifferentiation (reaching indicate that there are significant similarities, as well as some Freeman stages 1e2) but only one of these foci appears to form the differences, between the processes of lens development and lens news lens. An element of the Delta- known regeneration. Some of the differences may be associated with the to play roles in lateral inhibition and boundaries-inductive mecha- processes of , as well as cellular dedifferentiation that nisms (Artavanis-Tsakonas et al., 1999; Schweisguth, 2004; Gazave may be associated with lens regeneration (Carinato et al., 2000; et al., 2009) is expressed during lens regeneration (including Mind Henry et al., 2002; Malloch et al., 2009). It will be important to bomb homolog 1, which encodes a E3 ubiquitin protein ligase known decipher the roles of those genes that only appear to be expressed in to regulate Delta-mediated Notch signaling, Malloch et al., 2009). regenerating lenses. Perhaps Delta-Notch signaling plays a role in regulating the devel- Comparison of the genes expressed during lens regeneration in opment of these foci. As argued above, some mechanism may Xenopus with those expressed in other regenerating tissues/organ- operate to ensure that only a single lens is ultimately regenerated. A isms, did not reveal a substantial conserved core of genes that appear similar mechanism could also function in the context of embryonic to underlie different regenerative phenomena (Malloch et al., 2009). lens development given that a larger field of competent tissue is There are, however, some commonalities. For instance, MMPs established early during development than actually generates the appear to be expressed in a number of different regenerating tissues, developing lens. As mentioned above, direct contact between the including regenerating lenses (Yang and Bryant, 1994; Miyazaki cornea epithelium and the retina is not required for lens regenera- et al., 1996; Yang et al., 1999; Kherif et al., 1999; Carinato et al., tion, and this might preclude the involvement of Delta-Notch 2000; Kato et al., 2003; Vinarsky et al., 2005; Odelberg, 2005; signaling in that particular interaction. Malloch et al., 2009). In addition, elements of the stress, inflamma- tion and immune response pathways also appear to be expressed in 3. Studies of lens regeneration and development in Xenopus many regenerating systems, including Xenopus lens regeneration (Lu and Richardson, 1991; Meyer-Franke et al., 1998; Leon et al., As a model vertebrate system, Xenopus offers significant advan- 2000; Patruno et al., 2001; Imokawa and Brockes, 2003; Harty tages for the study of regeneration, which have been extolled in et al., 2003; Imokawa et al., 2004; Mescher and Neff, 2005; a number of recent reviews (Slack et al., 2004, 2008; Callery, 2006; Makino et al., 2005; Kanao and Miyachi, 2006; Brockes and Kumar, Tseng and Levin, 2008; Henry et al., 2008; Beck et al., 2009; Filoni, 2008; Filbin, 2006; Godwin and Brockes, 2006; Mescher et al., 2009). Tremendous molecular and genomic resources exist for this 2007; Pearl et al., 2008; Malloch et al., 2009). These researchers system (Klein et al., 2002, 2006; Carruthers and Stemple, 2006). propose conflicting hypotheses as to whether stress, inflammation Likewise, Xenopus has been used widely in the study of eye devel- and immune response pathways stimulate or inhibit regeneration. opment (Henry et al., 2008). Maintenance of a colony of adult frogs is rather simple and inexpensive, and the adults provide an abundant 2.3. Signaling factors involved in Xenopus lens regeneration supply of embryonic material, which is available year-round. Furthermore, the large eggs and develop externally and The inductive signals that trigger lens regeneration have not yet rapidly. In addition, a vast array of molecular tools are available to been identified. There is some evidence, however, that they may be facilitate studies of gene function in these animals. As previously similar to those that are responsible for embryonic lens induction mentioned, these include the availability of a sequenced genome (Henryand Mittleman,1995; Cannata et al., 2003, 2008; Arresta et al., (http://genome.jgi-psf.org/Xentr4/Xentra4.home.html). The X. tro- 2005). Studies indicate that members of the FGF family and specific picalis genome has a high number of regions with long stretches of BMPs (BMP4, and BMP7) play key roles in eye and lens development synteny with mammalian (each typically a hundred or

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 6 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 more genes). Hence, it is evolutionarily well positioned to identify tropicalis biology and (e.g, “,” www.xenbase.org conserved elements in vertebrate genomes (e.g., Ogino et al., 2008). and see also “Xine,” http://blumberg.bio.uci.edu/xine/index.htm). A It is a relatively simple matter to carry out both gain- and loss-of- long-standing course taught at Cold Spring Harbor covers topics function analyses to examine the necessity and sufficiency of specific related to the cell and of Xenopus, and the genes. For example may be used to knockdown trans- and Courses taught at the Marine Biological lation of specificmessages(Ekker, 2000; Corey and Abrams, 2001; Nutt Laboratory also cover Xenopus biology, training future generations of et al., 2001; Heasman, 2002; Gene Tools, LLC, Philomath, OR; www. scientists in the use of this system. These resources continue to gene-tools.com). Affymetrix produces micro-arrays to study gene position Xenopus as a key system for the study of regeneration and expression in both X. laevis and X. tropicalis (www.affymetrix.com). A development for many years to come. reverse /TILLING resource (Stemple, 2004)attheSanger Centre (Cambridge, UK) has recently led to the identification of the first 4. Lens regeneration in newts null mutation in the Rx gene (a key regulator of eye formation, S. Carruthers, D. Stemple and R. Grainger. University of 4.1. Morphological events Virgina, Unpublished, see also Mathers et al., 1997) and other muta- tions are in the initial stages of characterization. In the case of the newt, lens regeneration involves trans- The relative ease with which one can prepare transgenic frogs is differentiation of the pigmented epithelial cells of the iris (PECs). a tremendous benefit and facilitates molecular functional studies at Following removal of the lens, the PECs re-enter the cell cycle, more advanced stages of larval development, which is essential for dedifferentiate, and lose their characteristic pigmentation. In vivo, studying regenerative phenomenon (Kroll and Amaya,1996; Amaya regeneration occurs from the dorsal iris population of PECs. Despite and Kroll, 1999; Amaya et al., 1998; Huang et al., 1999; Marsh- the apparent similarity between the dorsal and ventral PECs, the Armstrong et al., 1999; Offield et al., 2000; Sparrow et al., 2000; ability to regenerate through transdifferentiation under normal Hirsch et al., 2002; Ogino et al., 2006; Pan et al., 2006; Slack et al., conditions, belongs exclusively to the dorsal PECs. Interestingly, 2008: Beck et al., 2009). For instance, transgenic frogs carrying however, culturing of both dorsal and ventral PECs results in lentoid various genes under the control of the heat-shock (hsp70) promoter body formation (Del Rio-Tsonis and Tsonis, 2003; Tsonis and Del (see Wheeler et al., 2000) have been used to study tail and limb Rio-Tsonis, 2004). This means that the ventral PECs have the regeneration (Beck and Slack, 2001; Beck et al., 2003, 2006; Slack potential for transdifferentiation, but this is not permitted in vivo. et al., 2004; Lin and Slack, 2008). Beck et al. (2003, 2006), see also Early events of dedifferentiation occur through day 8 of the Slack et al. (2004) investigated the activity of the BMP and Notch regeneration process. is initiated by day 4 post- signaling pathways during spinal cord and muscle regeneration in lentectomy. Visible dedifferentiation and loss of pigment from the the tail and limb, akin to the unpublished study by Beck’s lab that PECs begins by day 8. By day 10 of the process, a lens vesicle has examined lens regeneration (as described above). Knocking down formed, which represents the precursor to what will become a fully either BMP or Notch inhibited regeneration while activation of differentiated lens. After the lens vesicle has formed, the posterior either promoted regeneration. These experiments showed further cells start to elongate, express crystallins and differentiate to lens that BMP acts upstream of Notch in spinal cord regeneration, but fibers. The anterior cells become lens epithelium (Fig. 3)(Eguchi, these two pathways appeared to act independently in the case of 1963, 1964). muscle regeneration. Lin and Slack (2008) also used transgenic frogs Once lens differentiation has started the process is remarkably to show that FGF and Wnt signaling are important in Xenopus tail similar to lens development (as far as the sequential appearance of regeneration by expressing the Wnt inhibitor dkk1 and a dominant the different crystallins is concerned). Indeed, studies using anti- negative (truncated) form of an FGF receptor under the control of the bodies to crystallins or cDNA probes have conclusively shown that hsp70 heat-shock promoter. there is a parallel of their synthesis during development and regen- Tissues isolated from transgenic frogs carrying a GFP reporter eration. For instance, aA, bB1- and g-crystallins appeared all at the driven by the cytomegalovirus (CMV) promoter have been used in same time at the ventral-posterior portion of the lens vesicle (Sato transplantation experiments to follow the fates of cells that stage IVeV; nearly 10e12 days post-lentectomy (Yamada, 1977)), contribute to regenerated tissues in the tail (Gargioli and Slack, with g-crystallin (in contrast with the other two) being specific for 2004). This study demonstrated that notochord and spinal cord the lens fibers and not the lens epithelial cells (Mizuno et al., 2002). both regenerated from prexisting notochord and spinal cord stump tissues, respectively. However, muscles arose from a population of satellite “stem” cells, rather than from pre-existing myofibers. Chen 4.2. Gene regulation et al. (2006) showed further that this population of satellite cells specifically express Pax7. Using a similar approach, Lin et al. (2007) Focusing on the possible mechanisms of genetic regulation, it showed that melanophores do not arise from a pre-existing pop- would only make sense to hypothesize that different genes are at play ulation of unpigmented, melanophores precursors during tail in the dorsal and ventral iris creating two very different populations regeneration. The cre/lox system has also been employed to of cells, one that regenerates and one that does not, respectively. generate transgenic frogs that express yellow fluorescent protein as There are of course, many genes and proteins to consider in the a muscle specific lineage marker (under control of the cardiac regeneration process. As previously mentioned, a master regulator promoter) to show that differentiated muscle cells do not undergo gene in eye development, Pax6 is found in both the dorsal and ventral transdifferentiation to form other cell types during tail regeneration iris during Wolffian lens regeneration (Del Rio-Tsonis et al., 1995; (Ryffel et al., 2003). Transgenic X. tropicalis lines carrying a g1- Madhavan et al., 2006). The same is true for Six3, a partner of Pax6 crystallin promoter driving lens GFP expression have proven to be and by itself sufficient to induce lens when ectopically expressed in very useful for the study of lens development and regeneration frogs and fish (Oliver et al., 1996; Altmann et al., 1997). Prox1 is (Offield et al., 2000; Henry and Elkins, 2001, see Fig. 2AeC). Labs, a transcriptional regulator of crystallin synthesis and is also including our own, are now applying these transgenic approaches to expressed just before cells of the vesicle elongate and express crys- study lens regeneration. tallins (Del Rio-Tsonis et al., 1999). Interestingly these genes are also Finally, tremendous NIH-funded community web resources are expressed in the ventral iris tissues. This finding was unexpected and available that provide valuable information on X. laevis and X. it means that the ventral iris does initially undergo similar events as

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 7

Fig. 3. The process of lens regeneration in newts. Lens formation from the dorsal iris during regeneration shown through scanning electron microscopy. (A) Day 10 after lentectomy, a lens vesicle (grey) forms at the dorsal margin of the iris. (B) Day 14, elongation and further lens development as differentiation of lens fibers occurs at the posterior part (arrow) of the vesicle. (C) Day 20 of lens reformation. The anterior portion has become the lens epithelium (colored yellow). In contrast, lens fiber cells are forming in the posterior region. (D) Day 30, regeneration has concluded. the dorsal iris. In fact there may be a general inhibitory effect that collaborate with the Wnt pathway. When explants of dorsal iris were prevents further regeneration from taking place. treated with FGF2 in the presence of Wnt inhibitors, the action of To gain more insights about gene expression patterns, a micro- FGF2 was inhibited. Combined addition of FGF2 and Wnt3a was in array analysis with newt cDNA was utilized and it revealed that gene fact able to induce lens transdifferentiation of ventral explants as signatures in the dorsal and ventral iris are in fact very similar well (Hayashi et al., 2006). (Makarev et al., 2007). Even genes responsible for tissue remodeling, Interestingly, research on another signaling pathway, the Bone such as collagenase and cathepsin are present and up-regulated in Morphogenetic Protein pathway (BMP), revealed that its inhibition both dorsal and ventral irises during regeneration. Surprisingly allows the ventral iris to transdifferentiate into lens. Importantly, expression levels for some of these genes are even higher in the such induction was also observed when ventral PECs were trans- ventral iris. It is not unreasonable to suggest, given these unexpected fected with Six-3 and treated with retinoic acid (Grogg et al., 2005). results on gene expression, that we might encounter novel regula- These manipulations of signaling pathways and regulatory genes tory events during newt regenerative processes (Makarev et al., clearly indicate that induction of the ventral iris is possible and such 2007). EST analysis from irises 8 days after lentectomy, when observations might open newavenues in experimenting with higher dedifferentiation of PECs is ongoing, has also revealed very impor- animals that are unable to regenerate eye tissues. tant information. An initial set of 10,449 cDNA sequence reads were obtained. The total length of these sequences was 9.85 Mbp that was reduced to 4.82 Mbp by assembling overlapping reads using CAP3 4.4. MicroRNAs software (Huang and Madan,1999). This assembly was composed of 4725 unique sequences (1368 contigs and 3357 singlets). Of 1219 MicroRNAs, or miRNAs are short RNAs, about 22 nucleotides long, contigs and 3357 singlets, 780 contigs and 1666 singlets were which can bind to complementary sequences of RNA and subse- annotated by Blast2GO (Conesa et al., 2005). The 15 most frequently quently prevent mRNA translation. miRNAs may have multiple counted biological process terms, cellular component terms, and binding sequences due to their short size and can block translation molecular functions were reported (Maki et al., 2010). It is inter- on a wide scale. As such they might be involved in the transition from esting to note here that when compared with a same analysis one cell type to another as we see during regeneration. Cloning of performed during Xenopus lens regeneration the most frequent GO newt miRNA from the eye has pinpointed differential regulation in annotations were very similar (Table 1). GO () is used both dorsal and ventral iris. Some of the targets of the cloned miR- to describe the function of a gene as it pertains to processes and NAs were predicted to be FGFR2, and Sox9 (for miR-124a), Pax3, place in the cell. There are 3 independent sets of ontologies, the chordin, and TGFbR1 for let7b (Makarev et al., 2006). Based on this, molecular function (or molecular component, e.g. DNA binding), the further study of the role of miRNA regulation in regeneration biological process (e.g. transcription) and the cellular component (or compartment) where the gene product can be found (e.g. nucleus). Table 1 The 5 most frequent GO identified in Xenopus and Newt.

4.3. Signaling pathways in newt lens regeneration GO Xenopus Newt Biological Process Regulation of transcription, Regulation of transcription, A number of cell signaling pathways have been implicated in the DNA dependent DNA dependent process of lens regeneration in the newt. Members of the hedgehog Transcription Signal transduction signaling pathway are also expressed during the regeneration Transport Ribosome biogenesis process; (Shh) and Indian hedgehog (Ihh)onlybeing Signal transduction Translation Multicellular organismal Electron transport expressed in the regenerating and developing lens, and no longer development expressed once the lens is fully reformed. When one interferes with Cellular Nucleus Cytoplasm this signaling pathway, the overall regeneration process is inhibited. Compartment Membrane Nucleus Also, cell proliferation rates are decreased and differentiation in the Cytoplasm Integral to membrane regenerating lens suffers (Tsonis et al., 2004). Retinoic acid receptors Integral to membrane Transcription factor complex are also expressed in the lens during regeneration and their inhibition Intracellular Extracellular space might account for aberrant lens formation (Tsonis et al., 2000, 2002). Molecular Protein binding Protein binding Exogenous FGFs have been shown to elicit regeneration of Component Metal ion binding ATP binding a second lens from the dorsal iris (Del Rio-Tsonis et al.,1997; Hayashi DNA binding Zinc ion binding Nucleotide binding RNA binding et al., 2004). It has been suggested that this action of FGFs is medi- Zinc ion binding DNA binding ated via an induction of cell proliferation. The FGF pathway seems to

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 8 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 showed that members of the let7 family were found to be down- differentiate into the cells needed to create a new lens. Such regulated in the regenerating dorsal iris. Examination of the ventral a hypothesis was first proposed in 2000, using the example of iris revealed that miR-148 is up-regulated in both intact and mesenchymal stem cell similarity to limb cells (Tsonis, regenerating ventral iris when compared with dorsal counterparts 2000, 2004). Similar processes may also occur during lens regen- (Tsonis et al., 2007). Thus, miRNAs might be very useful regulators of eration in the frog (Henry, 2003). Research in this area is being regenerative processes. pursued, and some initial studies in the newt support this hypoth- Our previous study on the expression of miRNAs during lens and esis. For example, the stem cell nuclear protein nucleostemin (also regeneration in newts was the first to indicate a possible a cancer cell marker) is found highly expressed in the nucleus of involvement and regulation. The study suggested that, indeed, undifferentiated cells, like pluripotent embryonic stem cells, as well miRNAs can have a role in regeneration. Other recent reports as multipotent stem cells of the nervous system and primitive cells of support this as well. During zebrafish fin regeneration it has been the bone marrow (Maki et al., 2007). As the onset of differentiation of shown that miR-203 regulates the tran- these cells occurs, accumulation of nucleostemin within the nucleus scription factor Lef1. Down-regulation of Lef1 by miR-203 blocks is shown to decrease. When dedifferentiated PECs were studied regeneration, while loss of miR-203 results in up-regulation of Lef1 during regeneration, as compared to non-regenerating PECs, this and fin overgrowth (Thatcher et al., 2008). In another study it was stem cell nuclear protein was found to be highly expressed in both found that depletion of miR-133 promotes fin regeneration and that dorsal and ventral regenerating PECs. The amount of protein present this is FGF-dependent (Yin et al., 2008). In a different study it was decreased faster in the ventral PECs, and after lens differentiation shown that miR-196 is involved in tail regeneration. Inhibi- the amount of nucleostemin also decreased in dorsal PECs (Maki tion of miR-196 blocks regeneration by acting upstream BMP4 and et al., 2007). Pax-7 (Sehm et al., 2009). Further studies were undertaken to characterize gene expression during lens regeneration in the newt. A list of the lens-regenerating 4.5. Knock-down technology and gene expression in newts iris ESTs, possible candidate genes for participating in nuclear regulation during newt dedifferentiation have been identified (Maki Transgenesis and knockout technologies are not as developed in et al., 2010). This is similar to the types of genes expressed during the newt when compared to Xenopus or even the axolotl (another lens regeneration in the frog (Malloch et al., 2009). Epigenetic that can regenerate body parts such as tail and limb, regulation, a range of heritable chromatin modifications including but is unable to regenerate the lens). Nevertheless knockdown histone modifications, DNA methylation and chromatin remodeling, technology has been developed in the newt and it promises to shed play a pivotal role in the control of differentiation and maintenance more light to the mechanisms of regeneration. Application of such of cellular identity. So it is expected that epigenetic regulation plays technology in the adult newt has been quite informative on the role an important role during newt PEC dedifferentiation. Indeed, many of Pax6 in this process. When Pax6 expression is decreased in the factors involved in reprogramming were identified in the ESTs and eye, lens regeneration suffers dramatically due to the decreases in interestingly were also found in newt ovarian tissues as well. proliferation of the pigmented epithelial cells. When Pax6 is The expression patterns of these genes may be significant in the knocked down during later regeneration events, crystallins are not field of cellular reprogramming, stem cells and regeneration in made and lens fibers production is decreased (Fig. 4)(Madhavan general. While the newt does not reprogram its adult differentiated et al., 2006). cells to form pluripotent progenitors, it does reprogram them to specific progenitor stem cell state according to their origin and to 4.6. Transdifferentiation in newts: a model for stem cell their contribution, as evidenced by their ability to regenerate with differentiation? precision only those parts lost to injury (Kragl et al., 2009). Indeed, aggregates from dorsal iris PECs will transdifferentiate only to lens One could argue that during the process of lens regeneration the even if transplanted to regenerating limb (Ito et al., 1999; Tsonis, dedifferentiated cells become stem cell-like, going from a terminally 2000). These data might suggest an underlying mechanism for this differentiated state to an undifferentiated state and can then further property in the newt.

Fig. 4. Pax6 treatment inhibits crystallin expression. Morpholinos were injected at day 10 after lentectomy, and animals were collected for assay at day 13 after lentectomy. (A and F) Lens vesicles from untreated control eyes express a- and b-crystallin (a-cry or b-cry). (B and G) Eyes treated with control morpholino (C-Mo) also express a and b crystallin. (D and I) Lens vesicles from eyes treated with Pax6 morpholino (Pax6-Mo) do not show any a-orb-crystallin expression. (C, E, H, and J) a-orb-crystallin staining only from B and D (C and E) and b-crystallin staining only from G and I (H and J).

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 9

Recently, it has been shown that differentiated mammalian cells, 1999; Mizuno et al., 1999a, 2005; Henry et al., 2002; Henry, 2003; including human, can also be reprogrammed to become induced Cannata et al., 2003; Malloch et al., 2009; Filoni, 2009). Answers to pluripotent stem cells (iPSCs) that can subsequently differentiate to these questions will require careful molecular characterization and different tissues. This reprogramming is mediated by inducing the comparison of the genes expressed in these tissues and the changes expression of four factors, Oct4, , c- and . Another factor, that occur during the processes of lens regeneration and nanog, seems to be also important (Takahashi and Yamanaka, 2006; development. Takahashi et al., 2007; Okita et al., 2007; Wernig et al., 2007). All It is unclear whether the larval cornea or the adult iris contains these are transcriptional factors expressed in embryonic stem cells. populations of specialized somatic stem cells. For instance, it is The critical question that arises from these studies is: Does reprog- known in other vertebrates that there exists a population of limbal ramming that mediates the formation iPSCs share any similarities to stem cells that supports repair of the cornea epithelium (Buck, reprogramming during regeneration in newts? A recent study 1985; Cotsarelis et al., 1989; Huang and Tseng, 1991; Auran et al., examined expression of these stem cell pluripotency-maintaining 1995; Lavker et al., 1998; Pellegrini et al., 1999; Tseng and Sun, factors during newt lens regeneration (Maki et al., 2009). Interest- 1999). We have looked for a population of slowly dividing stem ingly, there was significant regulation of three of the factors that we cells (BrdU-retaining-cells) in Xenopus and have not found any examined, Sox2, Klf4 and c-myc. Oct4 and nanog were not detected in evidence that these exist in the limbal region. Rather, these these tissues beyond the levels of negative control (-RT), however, preliminary investigations revealed that dividing cells are seen they were expressed in control ovarian tissues (meaning that the scattered throughout the cornea epithelium (see also Freeman, newt does have these genes). 1963). Hence, it is not clear if there is a specialized subpopulation During lens regeneration, Sox2 and Klf4 were up-regulated of stem cells within the cornea that support regeneration. Likewise during the very early stages of regeneration (day 2), while c-myc it is not believed at the present time that the newt iris contains showed a peak of expression at day 8. Day 2 marks an early response a stem cell population. Rather, all the evidence testifies for a clear to lens removal and is expected to be characterized by events that case of transdifferentiation. However, more studies on cell pop- may prepare pre-existing tissues for reprogramming and cell cycle ulations using advanced techniques will eventually clear this issue. re-entry. In fact, cell proliferation is not detected until day 4. Those Though the regenerated lenses appear to be normal on the basis rapid responses to lens removal prior to cell cycle re-entry are of morphological and histological criteria, no attempt has been similar to those observed for nucleostemin (Maki et al., 2007). c-myc made to examine lens function or visual acuity following regener- showed quite opposite patterns to Sox2 and Klf4. It was highly ation in either Xenopus or newts. In addition, it is unclear whether expressed at day 8, which correlated with the establishment of the other associated structures are also fully regenerated, such as the lens vesicle, but without major differences between dorsal and muscles that reposition the lens, etc. ventral iris. It should be noted here that the regeneration-incom- Finally, we do not understand what factors may prevent lens petent ventral iris does show activity such as cell cycle re-entry and regeneration from occurring in other vertebrates, including humans. gene expression, which must be suppressed later (Grogg et al., 2005; Do all vertebrates possess a similar conserved suite of genes that Madhavan et al., 2006). Therefore, expression differences between could be active in these processes? Some evidence suggests that dorsal and ventral iris might be correlated with these events. there might be species specific differences that underlie some The specific stage-related regulation of Sox2, Klf4 and c-myc,and regenerative phenomena (e.g., limb regeneration in newts, Kumar the absence of Oct4 and nanogexpression might indicatewhy the newt et al., 2007; Brockes and Kumar, 2008; Garza-Garcia et al., in cells do not become pluripotent. Manipulating these factors in the press). In the case of lens regeneration, barriers may be present to newt will open new avenues in the field and should be of enormous ensure that abnormal, supernumerary lenses do not form within the importance to compare or contrast the biology of regeneration in eye that would prevent regeneration from occurring. This could classical models and in stem cells. involve the production of specific inhibitors of lens regeneration, or it is possible that key inductive signals are either not present or the 5. Xenopus and newt lens regeneration: some additional receptors or downstream elements of the signal cascade are missing questions in eye tissues. Once we understand the factors required to support lens regeneration, and the reasons why some vertebrates are unable While there have been many advances in our understanding of to regenerate these structures, it may be possible to “restore” this the cellular and molecular mechanisms that control lens regenera- capacity, even in humans, as a therapeutic approach to treat injured tion in Xenopus, and newts, a number of key questions still need to be or diseased lenses. addressed in these systems, in addition to those already raised above. The process of “transdifferentiation” is described as one that takes a population of differentiated cells though an initial process of Acknowledgements cellular dedifferentiation, followed by re-differentiation along an altered trajectory (e.g., Eguchi and Kodama, 1993; Burke and Tosh, The authors’ research is supported by NIH/NEI grant EY09844 2005). While it is clear this does take place during Wolffian lens (JJH) and EY10540 (PAT). regeneration in the newt, investigators have questioned whether the process of lens regeneration in Xenopus actually involves dedifferentiation. In fact, the cornea is not fully differentiated until References after the process of metamorphosis is completed (Yamada, 1982; Adler, R., Canto-Soler, M.V., 2007.Molecular mechanisms of optic vesicle development: McDevitt and Brahma, 1982; Bosco, 1988; Henry, 2003). The larval complexities, ambiguities and controversies. Developmental Biology 305, 1e13. cornea and lens are originally derived from the same embryonic Altmann, C.R., Chow, R.L., Lang, R.A., HemmatiBrivanlou, A., 1997. Lens induction by e tissue, and the phenomenon of lens regeneration in Xenopus may Pax-6 in Xenopus laevis. Developmental Biology 185, 119 123. Amaya, E., Offield, M.F., Grainger, R.M., 1998. Frog genetics: Xenopus tropicalis jumps simply represent a condition in which there is persistent compe- into the future. Trends in Genetics 14, 253e255. tence of embryonic and larval ectoderm to respond to key lens Amaya, E., Kroll, K.L., 1999. A method for generating transgenic frog embryos. inducing signals. Likewise, the precise relationships between Molecular Embryology 97, 393e414. Ang, S.J., Stump, R.J.W., Lovicu, F.J., McAvoy, J.W., 2004. Spatial and temporal embryonic lens development and lens regeneration, though expression of Wnt and Dickkopf genes during murine lens development. Gene apparently similar processes, is not entirely clear (see Schaefer et al., Expression Patterns 4, 289e295.

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 10 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13

Arresta, E., Bernardini, S., Gargioli, C., Filoni, S., Cannata, S.M., 2005. Lens-forming Carruthers, S., Stemple, D.L., 2006. Genetic and genomic prospects for Xenopus competence in the epidermis of Xenopus laevis during development. Journal of tropicalis research. Seminars in Cell & Developmental Biology 17, 146e153. Experimental Part A-Comparative Experimental Biology 303A, 1e12. Chang, W.Y., KhosrowShahian, F., Chang, R., Crawford, M.J., 2001. xPitx1 plays a role Artavanis-Tsakonas, S., Rand, M.D., Lake, R.J., 1999. Notch signaling: cell fate control in specifying cement gland and head during early Xenopus development. and signal integration in development. Science 284, 770e776. Genesis 29, 78e90. Ashery-Padan, R., Marquardt, T., Zhou, X.L., Gruss, P., 2000. Pax6 activity in the lens Chen, Y., Lin, G.F., Slack, J.M.W., 2006. Control of muscle regeneration in the Xenopus is required for lens formation and for correct placement of a single tail by Pax7. Development 133, 2303e2313. retina in the eye. Genes & Development 14, 2701e2711. Chen, Y.J., Stump, R.J.W., Lovicu, F.J., Shimono, A., McAvoy, J.W., 2008. Wnt signaling Ashery-Padan, R., Gruss, P., 2001. Pax6 lights-up the way for eye development. is required for organization of the lens fiber cell cytoskeleton and development Current Opinion in Cell Biology 13, 706e714. of lens three-dimensional architecture. Developmental Biology 324, 161e176. Auran, J.D., Koester, C.J., Kleiman, N.J., Rapaport, R., Bomann, J.S., Wirotsko, B.M., Chow, R.L., Altmann, C.R., Lang, R.A., Hemmati-Brivanlou, A., 1999. Pax6 induces Florakis, G.J., Koniarek, J.P., 1995. Scanning slit confocal microscopic observation ectopic eyes in a vertebrate. Development 126, 4213e4222. of cell morphology and movement within the normal human anterior cornea. Chow, R.L., Lang, R.A., 2001. Early eye development in vertebrates. Annual Review of Ophthalmology 102, 33e41. Cell and Developmental Biology 17, 255e296. Beck, C.W., Christen, B., Slack, J.M.W., 2003. Molecular pathways needed for regen- Cioni, C., Filoni, S., Bosco, L., 1982. Inhibition of lens regeneration in larval Xenopus eration of spinal cord and muscle in a vertebrate. Developmental Cell 5, 429e439. laevis. Journal of Experimental Zoology 220, 103e108. Beck, C.W., Christen, B., Barker, D., Slack, J.M.W., 2006. Temporal requirement for Collucci, V., 1891. Sulla rigenerazione parziale deell’occhio nei tritoni: Isogenesi esvil- bone morphogenetic proteins in regeneration of the tail and limb of Xenopus luppo-Studio seprimentale. Mem. Accad. Sci. Istt. Bologna, Ser 5 (1), 593e629. tadpoles. Mechanisms of Development 123, 674e688. Conesa, A., Gotz, S., Garcia-Gomez, J.M., Terol, J., Talon, M., Robles, M., 2005. Beck, C.W., Belmonte, J.C.I., Christen, B., 2009. Beyond early development: Xenopus Blast2GO: a universal tool for annotation, visualization and analysis in as an emerging model for the study of regenerative mechanisms. Develop- functional genomics research. 21, 3674e3676. mental Dynamics 238, 1226e1248. Corey, D.R., Abrams, J.M., 2001. Morpholino antisense : tools for Beck, C.W., Slack, J.M., 2001. An amphibian with ambition: a new role for Xenopus in investigating vertebrate development. Genome Biology 2 REVIEWS 1015. the 21st century. Genome Biology 2 REVIEWS 1029. Cotsarelis, G., Cheng, S.Z., Dong, G., Sun, T.T., Lavker, R.M., 1989. Existence of Beebe, D.C., 1994. Homeobox genes and vertebrate eye development. Investigative slow-cycling limbal epithelial basal cells that can be preferentially stimulated to Ophthalmology & Visual Science 35, 2897e2900. proliferate e implications on epithelial stem-cells. Cell 57, 201e209. Beebe, D., Garcia, C., Wang, X.H., Rajagopal, R., Feldmeier, M., Kim, J.Y., Chytil, A., Cui, W.W., Tomarev, S.I., Piatigorsky, J., Chepelinsky, A.B., Duncan, M.K., 2004. Mafs, Moses, H., Ashery-Padan, R., Rauchman, M., 2004. Contributions by members of Prox1, and Pax6 can regulate chicken beta B1-crystallin gene expression. Journal the TGF-b superfamily to lens development. International Journal of Develop- of Biological Chemistry 279, 11088e11095. mental Biology 48, 845e856. Cvekl, A., Duncan, M.K., 2007. Genetic and epigenetic mechanisms of gene regula- Belecky-Adams, T.L., Adler, R., Beebe, D.C., 2002. Bone morphogenetic protein signaling tion during lens development. Progress in Retinal and Eye Research 26, and the initiation of lens fiber cell differentiation. Development 129, 3795e3802. 555e597. Bosco, L., 1988. Transdifferentiation of ocular-tissues in larval Xenopus laevis. Cvekl, A., Piatigorsky, J., 1996. Lens development and crystallin gene expression: Differentiation 39, 4e15. many roles for Pax-6. Bioessays 18, 621e630. Bosco, L., Filoni, S., Cannata, S., 1979. Relationships between eye factors and lens- Del Rio-Tsonis, K., Trombley, M.T., McMahon, G., Tsonis, P.A., 1998. Regulation of lens forming transformations in the cornea and peri-corneal epidermis of larval regeneration by fibroblast growth factor receptor 1. Developmental Dynamics Xenopus laevis. Journal of Experimental Zoology 209, 261. 213, 140e146. Bosco, L., Filoni, S., Cioni, C., 1980. Lens formation from cornea in the presence of the Del Rio-Tsonis, K., Tsonis, P.A., 2003. Eye regeneration at the molecular age. old lens in larval Xenopus laevis. Journal of Experimental Zoology 213, 9e14. Developmental Dynamics 226, 211e224. Bosco, L., Valle, C., Willems, D., 1993. In-vivo and in-vitro experimental-analysis of Del Rio-Tsonis, K., Washabaugh, C.H., Tsonis, P.A., 1995. Expression of Pax-6 during lens regeneration in Larval Xenopus laevis. Development, Growth and Differ- Urodele eye development and lens regeneration. Proceedings of the National entiation 35, 257e270. Academy of Sciences of the United States of America 92, 5092e5096. Bosco, L., Venturini, G., Willems, D., 1994. First evidence of lens-transdifferentiation Del Rio-Tsonis, K., Jung, J.C., Chiu, I.M., Tsonis, P.A., 1997. Conservation of fibroblast of larval Xenopus laevis induced by brain-derived acidic FGF. Rendiconti Lincei 5, growth factor function in lens regeneration. Proceedings of the National 261e268. Academy of Sciences of the United States of America 94, 13701e13706. Bosco, L., Testa, O., Venturini, G., Willems, D., 1997a. Lens fibre transdifferentiation Del Rio-Tsonis, K., Tomarev, S.I., Tsonis, P.A., 1999. Regulation of Prox 1 during lens in cultured larval Xenopus laevis outer cornea under the influence of neural regeneration. Investigative Ophthalmology Visual Science 40, 2039e2045. retina-conditioned medium. Cellular and Molecular Life Sciences 53, 921e928. Eguchi, G., 1963. Electron microscopic studies on lens regeneration I. Mechanism of Bosco, L., Venturini, G., Willems, D., 1997b. In vitro lens transdifferentiation of depigmentation of the iris. Embryologia 8, 45e62. Xenopus laevis outer cornea induced by Fibroblast Growth Factor (FGF). Eguchi, G., 1964. Electron microscopic studies on lens regeneration II. Formation Development 124, 421e428. and growth of lens vesicle and differentiation of lens fibers. Embryologia 8, Bosco, L., Filoni, S., 1992. Relationships between presence of the eye cup and 247e287. maintenance of lens-forming capacity in larval Xenopus laevis. Development, Eguchi, G., Kodama, R., 1993. Transdifferentiation. Current Opinion in Cell Biology 5, Growth and Differentiation 34, 619e625. 1023e1028. Bosco, L., Willems, D., 1992. Persistence of the lens-inducing capacity of the neural Ekker, S.C., 2000. Morphants: a new systematic vertebrate functional genomics retina in adult Anura. Rendiconti Lincei 3, 345e351. approach. Yeast 17, 302e306. Brahma, S.K., 1980. Isofocusing and immunoelectrophoretic studies of soluble eye Enwright, J.F., Grainger, R.M., 2000. Altered retinoid signaling in the heads of small lens proteins from regenerated and normally developed Xenopus laevis. eye mouse embryos. Developmental Biology 221, 10e22. Experimental Eye Research 30, 269e275. Esteve, P., Bovolenta, P., 2006. Secreted inducers in vertebrate eye development: more Brahma, S.K., McDevitt, D.S., 1974. and localization of lens crystallins in functions for old morphogens. Current Opinion in Neurobiology 16, 13e19. Xenopus laevis lens regeneration. Journal of Embryology and Experimental Filbin, M.T., 2006. How inflammation promotes regeneration. Nature Neuroscience Morphology 32, 783e794. 9, 715e717. Brockes, J.R., Kumar, A., 2008. Comparative aspects of regeneration. Annual Filoni, S., 2009. Retina and lens regeneration in anuran amphibians. Seminars in Cell Review of Cell and Developmental Biology 24, 525e549. & Developmental Biology 20, 528e534. Buck, R.C., 1985. Measurement of centripetal migration of normal corneal epithelial- Filoni, S., Bosco, L., Cannata, S., 1978. Primi dati sperimentali sui fattori necessari per cells in the mouse. Investigative Ophthalmology & Visual Science 26,1296e1299. la transformazione lentogena della cornea esterna di larve di Xenopus laevis. Burke, Z.D., Tosh, D., 2005. Therapeutic potential of transdifferentiated cells. Clinical Acta Embryologiae et Morphologiae Experimentalis 3, 344. Science 108, 309e321. Filoni, S., Bosco, L., Paglioni, N., Cioni, C., 1980. Lens Formation from pericorneal Callery, E.M., 2006. There’s more than one frog in the pond: a survey of the epidermis in the presence of the old lens in larval Xenopus laevis. Journal of Amphibia and their contributions to developmental biology. Seminars in Cell & Experimental Zoology 211, 303e309. Developmental Biology 17, 80e92. Filoni, S., Bosco, L., Cioni, C., 1981. Experimental analysis of lens regeneration in Campbell, J.C., 1965. An Immuno-fluorescent study of lens regeneration in larval larval Xenopus laevis, the role of the retina and lens. Acta Embryologiae et Xenopus laevis. Journal of Embryology and Experimental Morphology 13, 171. Morphologiae Experimentalis 2, XVIeXXVII. Campbell, J.C., Truman, D.E.S., 1977. Variations in differentiation in regenerating lens Filoni, S., Bosco, L., Cioni, C., 1982. The role of neural retina in lens regeneration from of Xenopus laevis. Experimental Eye Research 25, 99e100. cornea in larval Xenopus laevis. Acta Embryologiae et Morphologiae Exper- Cannata, S.M., Arresta, E., Bernardini, S., Gargioli, C., Filoni, S., 2003. Tissue inter- imentalis 3, 15e28. actions and lens-forming competence in the outer cornea of larval Xenopus Filoni, S., Bosco, L., Cioni, C., Venturini, G., 1983. Lens forming transformations in laevis. Journal of Experimental Zoology Part A-Comparative Experimental larval Xenopus laevis induced by denatured eye-cup or its whole protein Biology 299A, 161e171. complement. Experientia 39, 315e317. Cannata, S.M., Bernardini, S., Filoni, S., Gargioli, C., 2008. The optic vesicle promotes Filoni, S., Bernardini, S., Cannata, S.M., Dalessio, A., 1997. Lens regeneration in larval cornea to lens transdifferentiation in larval Xenopus laevis. Journal of Anatomy Xenopus laevis: experimental analysis of the decline in the regenerative capacity 212, 621e626. during development. Developmental Biology 187, 13e24. Carinato, M.E., Walter, B.E., Henry, J.J., 2000. Xenopus laevis gelatinase B (Xmmp-9): Filoni, S., Bernardini, S., Cannata, S.M., 2006. Experimental analysis of lens-forming development, regeneration, and wound healing. Developmental Dynamics 217, capacity in Xenopus borealis larvae. Journal of Experimental Zoology Part 377e387. A-Comparative Experimental Biology 305A, 538e550.

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 11

Fini, M.E., Strissel, K.J., WestMays, J.A., 1997. Perspectives on eye development. Hyuga, M., Kodama, R., Eguchi, G., 1993. Basic fibroblast growth-factor as one of the Developmental Genetics 20, 175e185. essential factors regulating lens transdifferentiation of pigmented epithelial- Freeman, G., 1963. Lens regeneration from cornea in Xenopus laevis. Journal of cells. International Journal of Developmental Biology 37, 319e326. Experimental Zoology 154, 39. Imokawa, Y., Brockes, J.P., 2003. Selective activation of thrombin is a critical Freeman, G., Overton, J., 1961. Lens regeneration from the cornea in Xenopus laevis. determinant for vertebrate lens regeneration. Current Biology 13, 877e881. American Zoologist 1, 448e449. Imokawa, Y., Simon, A., Brockes, J.P., 2004. A critical role for thrombin in vertebrate Freeman, G., Overton, J., 1962. The effects of thyroxin on the competence of lens lens regeneration. Philosophical Transactions of the Royal Society of London regeneration in Xenopus. Anatomical Record 142, 305. Series B-Biological Sciences 359, 765e776. Fujiwara, M., Uchida, T., Osumiyamashita, N., Eto, K., 1994. Uchida Rat (Rsey) e de Iongh, R.U., Lovicu, F.J., Overbeek, P.A., Schneider, M.D., Joya, J., Hardeman, E.D., a new mutant rat with craniofacial abnormalities resembling those of the McAvoy, J.W., 2001. Requirement for TGF-b receptor signaling during terminal mouse Sey Mutant. Differentiation 57, 31e38. lens fiber differentiation. Development (Cambridge) 128, 3995e4010. Furuta, Y., Hogan, B.L.M., 1998. BMP4 is essential for lens induction in the mouse Ishibashi, S., Yasuda, K., 2001. Distinct roles of genes during Xenopus lens . Genes & Development 12, 3764e3775. development. Mechanisms of Development 101, 155e166. Gargioli, C., Giambra, V., Santoni, S., Bernardini, S., Frezza, D., Filoni, S., Ito, M., Hayashi, T., Kuroiwa, A., Okamoto, M., 1999. Lens formation by pigmented Cannata, S.M., 2008. The lens-regenerating competence in the outer cornea and epithelial cell reaggregate from dorsal iris implanted into limb blastema in the epidermis of larval Xenopus laevis is related to pax6 expression. Journal of adult newt. Development, Growth and Differentiation 41, 429e440. Anatomy 212, 612e620. Jean, D., Ewan, K., Gruss, P., 1998. Molecular regulators involved in vertebrate eye Gargioli, C., Slack, J.M.W., 2004. Cell lineage tracing during Xenopus tail regenera- development. Mechanisms of Development 76, 3e18. tion. Development 131, 2669e2679. Kablar, B., Vignali, R., Menotti, L., Pannese, M., Andreazzoli, M., Polo, C., Garza-Garcia, A.A., Driscoll, P.C., and Brockes, J.P. Evidence for the local of Giribaldi, M.G., Boncinelli, E., Barsacchi, G., 1996. Xotx genes in the developing mechanisms underlying limb regeneration in salamanders. Integrative and brain of Xenopus laevis. Mechanisms of Development 55, 145e158. Comparative Biology, In press. Kamachi, Y., Sockanathan, S., Liu, Q.R., Breitman, M., Lovellbadge, R., Kondoh, H., Gazave, E., Lapebie, P., Richards, G.S., Brunet, F., Ereskovsky, A.V., Degnan, B.M., 1995. Involvement of Sox proteins in lens-specific activation of crystallin genes. Borchiellini, C., Vervoort, M., Renard, E., 2009. Origin and evolution of the Notch Embo Journal 14, 3510e3519. signalling pathway: an overview from eukaryotic genomes. BMC Evolutionary Kamachi, Y., Uchikawa, M., Collignon, J., Lovell-Badge, R., Kondoh, H., 1998. Biology 9 Article No. 249. Involvement of Sox1, 2 and 3 in the early and subsequent molecular events of Godwin, J.W., Brockes, J.P., 2006. Regeneration, tissue injury and the immune lens induction. Development 125, 2521e2532. response. Journal of Anatomy 209, 423e432. Kamachi, Y., Uchikawa, M., Tanouchi, A., Sekido, R., Kondoh, H., 2001. Pax6 and SOX2 Gopal-Srivastava, R., Cvekl, A., Piatigorsky, J., 1998. Involvement of retinoic acid form a co-DNA-binding partner complex that regulates initiation of lens retinoid receptors in the regulation of murine alpha B-crystallin small heat development. Genes & Development 15, 1272e1286. shock protein gene expression in the lens. Journal of Biological Chemistry 273, Kanao, T., Miyachi, Y., 2006. Lymphangiogenesis promotes lens destruction and 17954e17961. subsequent lens regeneration in the newt eyeball, and both processes can be Graw, J., 1996. Genetic aspects of embryonic eye development in vertebrates. accelerated by transplantation of dendritic cells. Developmental Biology 290, Developmental Genetics 18, 181e197. 118e124. Graw, J., 2003. The genetic and molecular basis of congenital eye defects. Nature Kastner, P., Grondona, J.M., Mark, M., Gansmuller, A., Lemeur, M., Decimo, D., Reviews Genetics 4, 876e888. Vonesch, J.L., Dolle, P., Chambon, P., 1994. Genetic-analysis of Rxr-Alpha, Graw, J., Loster, J., 2003. Developmental genetics in ophthalmology. Ophthalmic developmental function e convergence of Rxr and Rar signaling pathways in Genet 24, 1e33. heart and eye . Cell 78, 987e1003. Grogg, M.W., Call, M.K., Okamoto, M., Vergara, M.N., Del Rio-Tsonis, K., Tsonis, P.A., Kato, T., Miyazaki, K., Shimizu-Nishikawa, K., Koshiba, K., Obara, M., Mishima, H.K., 2005. BMP inhibition-driven regulation of six-3 underlies induction of newt Yoshizato, K., 2003. Unique expression patterns of matrix metalloproteinases in lens regeneration. Nature 438, 858e862. regenerating newt limbs. Developmental Dynamics 226, 366e376. Grogg, M.W., Call, M.K., Tsonis, P.A., 2006. Signaling during lens regeneration. Kawamorita, M., Suzuki, C., Saito, G., Sato, T., Sato, K., 2002. In vitro differentiation of Seminars in Cell & Developmental Biology 17, 753e758. mouse embryonic stem cells after activation by retinoic acid. Hum Cell 15, Gurdon, J.B., Hopwood, N., 2000. The introduction of Xenopus laevis into develop- 178e182. mental biology: of empire, pregnancy testing and ribosomal genes. Interna- Kay, B.K., Peng, H.B., 1991. Xenopus laevis e practical uses in cell and molecular- tional Journal of Developmental Biology 44, 43e50. biology e preface. Methods in Cell Biology 36, R19eR22. Hanson, I.M., 2001. Mammalian homologues of the Drosophila eye specification Kenyon, K.L., Moody, S.A., Jamrich, M., 1999. A novel fork head gene mediates early genes. Seminars in Cell & Developmental Biology 12, 475e484. steps during Xenopus lens formation. Development 126, 5107e5116. Harty, M., Neff, A.W., King, M.W., Mescher, A.L., 2003. Regeneration or scarring: an Kherif, S., Lafuma, C., Dehaupas, M., Lachkar, S., Fournier, J.G., Verdiere-Sahuque, M., immunologic perspective. Developmental Dynamics 226, 268e279. Fardeau, M., Alameddine, H.S., 1999. Expression of matrix metalloproteinases 2 Hayashi, T., Mizuno, N., Ueda, Y., Okamoto, M., Kondoh, H., 2004. FGF2 triggers and 9 in regenerating skeletal muscle: a study in experimentally injured and iris-derived lens regeneration in newt eye. Mechanisms of Development 121, mdx muscles. Developmental Biology 205, 158e170. 519e526. Khosrowshahian, F., Wolanski, M., Chang, W.Y., Fujiki, K., Jacobs, L., Crawford, M.J., Hayashi, T., Mizuno, N., Takada, R., Takada, S., Kondoh, H., 2006. Determinative role 2005. Lens and retina formation require expression of Pitx3 in Xenopus pre-lens of Wnt signals in dorsal iris-derived lens regeneration in newt eye. Mechanisms ectoderm. Developmental Dynamics 234, 577e589. of Development 123, 793e800. Klein, S.L., Strausberg, R.L., Wagner, L., Pontius, J., Clifton, S.W., Richardson, P., 2002. Heasman, J., 2002. Morpholino oligos: making sense of antisense? Developmental Genetic and genomic tools for Xenopus research: the NIH Xenopus initiative. Biology 243, 209e214. Developmental Dynamics 225, 384e391. Henry, J.J., 2003. The cellular and molecular bases of vertebrate lens regenera- Klein, S.L., Gerhard, D.S., Wagner, L., Richardson, P., Schriml, L.M., Sater, A.K., tion. International Review of Cytology e a Survey of Cell Biology 228, Warren, W.C., McPherson, J.D., 2006. Resources for genetic and genomic studies 195e265. of Xenopus. Methods in Molecular Biology, 1e16. Henry, J.J., Elkins, M.B., 2001. Cornea-lens transdifferentiation in the anuran, Kodama, R., Eguchi, G., 1995. From lens regeneration in the newt to in-vitro Xenopus tropicalis. Development Genes and Evolution 211, 377e387. transdifferentiation of vertebrate pigmented epithelial-cells. Seminars in Cell Henry, J.J., Carinato, M.E., Schaefer, J.J., Wolfe, A.D., Walter, B.E., Perry, K.J., Elbl, T.N., Biology 6, 143e149. 2002. Characterizing gene expression during lens formation in Xenopus laevis: Kondoh, H., 1999. Transcription factors for lens development assessed in vivo. evaluating the model for embryonic lens induction. Developmental Dynamics Current Opinion in Genetics & Development 9, 301e308. 224, 168e185. Koster, R.W., Kuhnlein, R.P., Wittbrodt, J., 2000. Ectopic Sox3 activity elicits sensory Henry, J.J., Wever, J.A., Veragara, M.N., Fukui, L., 2008. An ideal vertebrate system for placode formation. Mechanisms of Development 95, 175e187. studies of eye development and regeneration. In: Tsonis, P.A. (Ed.), Animal Kragl, M., Knapp, D., Nacu, E., Khattak, S., Maden, M., Epperlein, H.H., Tanaka, E.M., Models for Eye Research. Academic Press, City. San Diego, pp. 57e92. 2009. Cells keep a memory of their tissue origin during axolotl limb regener- Henry, J.J., Mittleman, J.M., 1995. The matured eye of Xenopus laevis tadpoles ation. Nature 460, 60eU69. produces factors that elicit a lens-forming response in embryonic ectoderm. Kroll, K.L., Amaya, E., 1996. Transgenic Xenopus embryos from sperm nuclear Developmental Biology 171, 39e50. transplantations reveal FGF signaling requirements during . Hirsch, N., Zimmerman, L.B., Gray, J., Chae, J., Curran, K.L., Fisher, M., Ogino, H., Development 122, 3173e3183. Grainger, R.M., 2002. Xenopus tropicalis transgenic lines and their use in the Kumar, A., Godwin, J.W., Gates, P.B., Garza-Garcia, A.A., Brockes, J.P., 2007. Molecular study of embryonic induction. Developmental Dynamics 225, 522e535. basis for the nerve dependence of limb regeneration in an adult vertebrate. Huang, H.C., Marsh-Armstrong, N., Brown, D.D., 1999. Metamorphosis is inhibited in Science 318, 772e777. transgenic Xenopus laevis tadpoles that overexpress type III deiodinase. Lang, R.A., 2004. Pathways regulating lens induction in the mouse. International Proceedings of the National Academy of Sciences of the United States of Journal of Developmental Biology 48, 783e791. America 96, 962e967. Lavker, R.M., Wei, Z.G., Sun, T.T., 1998. Phorbol ester preferentially stimulates mouse Huang, X.Q., Madan, A., 1999. CAP3: a DNA sequence assembly program. Genome fornical conjunctival and limbal epithelial cells to proliferate in vivo. Investi- Research 9, 868e877. gative Ophthalmology & Visual Science 39, 301e307. Huang, A.J.W., Tseng, S.C.G., 1991. Corneal epithelial wound-healing in the absence Leon, S., Yin, Y.Q., Nguyen, J., Irwin, N., Benowitz, L.I., 2000. Lens injury stimulates of limbal epithelium. Investigative Ophthalmology & Visual Science 32, axon regeneration in the mature rat optic nerve. Journal of Neuroscience 20, 96e105. 4615e4626.

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 12 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13

Li, H.S., Yang, J.M., Jacobson, R.D., Pasko, D., Sundin, O., 1994. Pax-6 Is first expressed Mizuno, N., Mochii, M., Yamamoto, T.S., Takahashi, T.C., Eguchi, G., Okada, T.S., in a region of ectoderm anterior to the early neural plate e implications for 1999b. Pax-6 and Prox 1 expression during lens regeneration from Cynops iris stepwise determination of the lens. Developmental Biology 162, 181e194. and Xenopus cornea: evidence for a genetic program common to embryonic Li, X., Cvekl, A., Bassnett, S., Piatigorsky, J., 1997. Lens-preferred activity of chicken lens development. Differentiation 65, 141e149. delta 1- and delta 2-crystallin enhancers in transgenic mice and evidence for Mizuno, N., Agata, K., Sawada, K., Mochii, M., Eguchi, G., 2002. Expression of retinoic acid-responsive regulation of the delta 1-crystallin gene. Develop- crystallin genes in embryonic and regenerating newt lenses. Development, mental Genetics 20, 258e266. Growth and Differentiation 44, 251e256. Lin, G.F., Chen, Y., Slack, J.M.W., 2007. Regeneration of neural crest derivatives in the Mizuno, N., Ueda, Y., Kondoh, H., 2005. Requirement for beta B1-crystallin promoter Xenopus tadpole tail. Bmc Developmental Biology 7. of Xenopus laevis in embryonic lens development and lens regeneration. Lin, G., Slack, J.M.W., 2008. Requirement for Wnt and FGF signaling in Xenopus Development, Growth and Differentiation 47, 131e140. tadpole tail regeneration. Developmental Biology 316, 323e335. Nieuwkoop, P.D., Faber, J., 1956. Normal Table of Xenopus laevis Daudin. North Lovicu, F.J., McAvoy, J.W., 2005. Growth factor regulation of lens development. Holland Publishing, Amsterdam. Developmental Biology 280, 1e14. Nishiguchi, S., Wood, H., Kondoh, H., Lovell-Badge, R., Episkopou, V., 1998. Sox1 Lu, X., Richardson, P.M., 1991. Inflammation near the nerve-cell body enhances directly regulates the gamma-crystallin genes and is essential for lens axonal regeneration. Journal of Neuroscience 11, 972e978. development in mice. Genes & Development 12, 776e781. Lupo, G., Andreazzoli, M., Gestri, G., Liu, Y., He, R.Q., Barsacchi, G., 2000. Homeobox Nutt, S.L., Bronchain, O.J., Hartley, K.O., Amaya, E., 2001. Comparison of morpholino genes in the genetic control of eye development. International Journal of based translational inhibition during the development of Xenopus laevis and Developmental Biology 44, 627e636. Xenopus tropicalis. Genesis 30, 110e113. Lupo, G., Liu, Y., Qiu, R., Chandraratna, R.A.S., Barsacchi, G., He, R.Q., Harris, W.A., Odelberg, S.J., 2005. Cellular plasticity in vertebrate regeneration. Anatomical 2005. Dorsoventral patterning of the Xenopus eye: a collaboration of Retinoid, Record Part B: The New Anatomist 287 ISSN 1552e4906(print)j1552e4914 Hedgehog and FGF receptor signaling. Development 132, 1737e1748. (electronic). Lupo, G., Harris, W.A., Lewis, K.E., 2006. Mechanisms of ventral patterning in the Offield, M.F., Hirsch, N., Grainger, R.M., 2000. The development of Xenopus tropicalis vertebrate nervous system. Nature Reviews Neuroscience 7, 103e114. transgenic lines and their use in studying lens developmental timing in living Madhavan, M., Haynes, T.L., Frisch, N.C., Call, M.K., Minich, C.M., Tsonis, P.A., Del embryos. Development 127, 1789e1797. Rio-Tsonis, K., 2006. The role of Pax-6 in lens regeneration. Proceedings of the Ogino,H., McConnell, W.B.,Grainger,R.M.,2006. Highlyefficient transgenesis in Xenopus National Academy of Sciences of the United States of America 103,14848e14853. tropicalis using I-SceI meganuclease. Mechanisms of Development 123, 103e113. Makarev, E., Spence, J., Del Rio-Tsonis, K., Tsonis, P., 2006. Identification of micro- Ogino, H., Fisher, M., Grainger, R.M., 2008. Convergence of a head-field selector Otx2 and RNAs and other small RNAs from the adult newt eye. Molecular Vision 12, Notch signaling: a mechanism for lens specification. Development 135, 249e258. 1386e1391. Ogino, H., Yasuda, K., 2000. Sequential activation of transcription factors in lens Makarev, E., Call, M.K., Grogg, M.W., Atkinson, D.L., Milash, B., Odelberg, S.J., induction. Development, Growth and Differentiation 42, 437e448. Tsonis, P.A., 2007. Gene expression signatures in the newt irises during lens Okita, K., Ichisaka, T., Yamanaka, S., 2007. Generation of -competent regeneration. Febs Letters 581, 1865e1870. induced pluripotent stem cells. Nature 448, 313e321. Maki, N., Martinson, J., Nishimura, O., Tarui, H., Meller, J., Tsonis, P.A., Agata, K., 2010. Oliver, G., Sosapineda, B., Geisendorf, S., Spana, E.P., Doe, C.Q., Gruss, P., 1993. Prox-1, Expression profiles during dedifferentiation in newt lens regeneration revealed a prospero-related homeobox gene expressed during mouse development. by expressed sequence tags. Molecular Vision 16, 72e78. Mechanisms of Development 44, 3e16. Maki, N., Takechi, K., Sano, S., Tarui, H., Sasai, Y., Agata, K., 2007. Rapid accumulation Oliver, G., Loosli, F., Koster, R., Wittbrodt, J., Gruss, P., 1996. Ectopic lens induction in of nucleostemin in nucleolus during newt regeneration. Developmental fish in response to the murine homeobox gene Six3. Mechanisms of Develop- Dynamics 236, 941e950. ment 60, 233e239. Maki, N., Suetsugu-Maki, R., Tarui, H., Agata, K., Del Rio-Tsonis, K., Tsonis, P.A., 2009. Oliver, G., Gruss, P., 1997. Current views on eye development. Trends in Neurosci- Expression of stem cell pluripotency factors during regeneration in newts. ences 20, 415e421. Developmental Dynamics 238, 1613e1616. Onuma, Y., Takahashi, S., Asashima, M., Kurata, S., Gehring, W.J., 2002. Conservation Makino, S., Whitehead, G.G., Lien, C.L., Kim, S., Jhawar, P., Kono, A., Kawata, Y., of Pax 6 function and upstream activation by Notch signaling in eye Keating, M.T., 2005. Heat-shock protein 60 is required for blastema formation development of frogs and flies. Proceedings of the National Academy of and maintenance during regeneration. Proceedings of the National Academy of Sciences of the United States of America 99, 2020e2025. Sciences of the United States of America 102, 14599e14604. Pan, F.C., Chen, Y.L., Loeber, J., Henningfeld, K., Pieler, T., 2006. I-SceI meganuclease- Malloch, E.L., Perry, K.J., Fukui, L., Johnson, V.R., Wever, J., Beck, C.W., King, M.W., mediated transgenesis in Xenopus. Developmental Dynamics 235, 247e252. Henry, J.J., 2009. Gene expression profiles of lens regeneration and develop- Pannese, M., Polo, C., Andreazzoli, M., Vignali, R., Kablar, B., Barsacchi, G., ment in Xenopus laevis. Developmental Dynamics 238, 2340e2356. Boncinelli, E., 1995. The Xenopus homolog of Otx2 is a maternal homeobox gene Manns, M., Fritzsch, B., 1991. The eye in the brain e retinoic acid effects morpho- that demarcates and specifies anterior body regions. Development 121, genesis of the eye and pathway selection of axons but not the differentiation of 707e720. the retina in Xenopus laevis. Neuroscience Letters 127, 150e154. Patruno, M., Thorndyke, M.C., Carnevali, M.D.C., Bonasoro, F., Beesley, P.W., 2001. Marsh-Armstrong, N., Huang, H.C., Berry, D.L., Brown, D.D., 1999. Germ-line Growth factors, heat-shock proteins and regeneration in . Journal transmission of transgenes in Xenopus laevis. Proceedings of the National of Experimental Biology 204, 843e848. Academy of Sciences of the United States of America 96, 14389e14393. Pearl, E.J., Barker, D., Day, R.C., Beck, C.W., 2008. Identification of genes associated Martinez-Morales, J.R., Signore, M., Acampora, D., Simeone, A., Bovolenta, P., 2001. with regenerative success of Xenopus laevis hindlimbs. Bmc Developmental Otx genes are required for tissue specification in the developing eye. Biology 8. Development 128, 2019e2030. Pellegrini, G., Golisano, O., Paterna, P., Lambiase, A., Bonini, S., Rama, P., De Luca, M., Mathers, P.H., Grinberg, A., Mahon, K.A., Jamrich, M., 1997. The Rx homeobox gene is 1999. Location and clonal analysis of stem cells and their differentiated progeny essential for vertebrate eye development. Nature 387, 603e607. in the human ocular surface. Journal of Cell Biology 145, 769e782. McDevitt, D.S., Brahma, S.K., 1982. Ontogeny and localization of the crystallins in Penzel, R., Oschwald, R., Chen, Y.L., Tacke, L., Grunz, H., 1997. Characterization and eye development and regeneration. In: McDevitt, D.S. (Ed.), Cell Biology of the early embryonic expression of a neural specific transcription factor xSOX3 in Eye. Academic Press, New York, pp. 143e191. Xenopus laevis. International Journal of Developmental Biology 41, 667e677. McDevitt, D.S., Brahma, S.K., Courtois, Y., Jeanny, J.C., 1997. Fibroblast growth factor Pommereit, D., Pieler, T., Hollemann, T., 2001. Xpitx3: a member of the Rieg/Pitx receptors and regenerationof the eyelens. Developmental Dynamics208, 220e226. gene family expressed during pituitary and lens formation in Xenopus laevis. Medina-Martinez, O., Shah, R., Jamrich, M., 2009. Pitx3 controls multiple aspects of Mechanisms of Development 102, 255e257. lens development. Developmental Dynamics 238, 2193e2201. Reeve, J.G., Wild, A.E., 1978. Lens regeneration from cornea of larval Xenopus laevis Mescher, A.L., Wolf, W.L., Moseman, A., Hartman, B., Harrison, C., Nguyen, E., in the presence of the lens. Journal of Embryology and Experimental Neff, A.W., 2007. Cells of cutaneous immunity in Xenopus: studies during larval Morphology 48, 205e214. development and limb regeneration. Developmental and Comparative Immu- Reza, H.M., Ogino, H., Yasuda, K., 2002. L-Maf, a downstream target of Pax6,is nology 31, 383e393. essential for chick lens development. Mechanisms of Development 116, 61e73. Mescher, A.L., Neff, A.W., 2005. Regenerative capacity and the developing immune Ring, B.Z., Cordes, S.P., Overbeek, P.A., Barsh, G.S., 2000. Regulation of mouse lens fiber system. I: theories. Models and Methods 93, 39e66. cell development and differentiation by the Maf gene. Development 127, 307e317. Meyer-Franke, A., Wilkinson, G.A., Kruttgen, A., Hu, M., Munro, E., Hanson, M.G., Ryffel, G.U., Werdien, D., Turan, G., Gerhards, A., Goosses, S., Senkel, S., 2003. Reichardt, L.F., Barres, B.A., 1998. Depolarization and cAMP elevation rapidly Tagging muscle cell lineages in development and tail regeneration using Cre recruit TrkB to the plasma membrane of CNS neurons. Neuron 21, 681e693. recombinase in transgenic Xenopus. Nucleic Acids Research 31, e44. Mitashov, V.I., 1966. Comparitive studies of lens regeneration in Cobitid fish Sato, T., 1961. Über bei der Cobitiden-Fischen. I. Misgurnus anguillicandatus (Cantor). (Misgurnus fossilis, Nemachilus barbatulus, Nemachhilus dorsalis). Doklady Embryologia 6, 251e290. Akademii Nauk SSSR 170, 1439e1442. Schaefer, J.J., Oliver, G., Henry, J.J., 1999. Conservation of gene expression during Miyazaki, K., Uchiyama, K., Imokawa, Y., Yoshizato, K., 1996. Cloning and charac- embryonic lens formation and cornea-lens transdifferentiation in Xenopus terization of cDNAs for matrix metalloproteinases of regenerating newt limbs. laevis. Developmental Dynamics 215, 308e318. Proceedings of the National Academy of Sciences of the United States of Schweisguth, F., 2004. Regulation of notch signaling activity. Current Biology 14, America 93, 6819e6824. R129eR138. Mizuno, N., Mochii, M., Takahashi, T.C., Eguchi, G., Okada, T.S., 1999a. Lens regen- Sehm, T., Sachse, C., Frenzel, C., Echeverri, K., 2009. miR-196 is an essential early- eration in Xenopus is not a mere repeat of lens development, with respect to stage regulator of tail regeneration, upstream of key spinal cord patterning crystallin gene expression. Differentiation 64, 143e149. events. Developmental Biology 334, 468e480.

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002 J.J. Henry, P.A. Tsonis / Progress in Retinal and Eye Research xxx (2010) 1e13 13

Semina, E.V., Murray, J.C., Reiter, R., Hrstka, R.F., Graw, J., 2000. Deletion in the regeneration: Let-7 members as potential regulators of dedifferentiation in lens promoter region and altered expression of Pitx3 homeobox gene in aphakia and inner ear hair cell regeneration of the adult newt. Biochemical and mice. Human Molecular Genetics 9, 1575e1585. Biophysical Research Communications 362, 940e945. Shi, X.H., Bosenko, D.V., Zinkevich, N.S., Foley, S., Hyde, D.R., Semina, E.V., Uwanogho, D., Rex, M., Cartwright, E.J., Pearl, G., Healy, C., Scotting, P.J., Sharpe, P.T., Vihtelic, T.S., 2005. Zebrafish is necessary for normal lens and retinal 1995. Embryonic expression of the chicken Sox2, Sox3 and Sox11 genes development. Mechanisms of Development 122, 513e527. suggests an interactive role in neuronal development. Mechanisms of Devel- Slack, J.M.W., Beck, C.W., Gargioli, C., Christen, B., 2004. Cellular and molecular opment 49, 23e36. mechanisms of regeneration in Xenopus. Philosophical Transactions of the Royal Vinarsky, V., Atkinson, D.L., Stevenson, T.J., Keating, M.T., Odelberg, S.J., 2005. Society of London Series B-Biological Sciences 359, 745e751. Normal newt limb regeneration requires matrix metalloproteinase function. Slack, J.M.W., Lin, G., Chen, Y., 2008. The Xenopus tadpole: a new model for Developmental Biology 279, 86e98. regeneration research. Cellular and Molecular Life Sciences 65, 54e63. Wagner, E., McCaffery, P., Drager, U.C., 2000. Retinoic acid in the formation of the Sparrow, D.B., Latinkic, B., Mohun, T.J., 2000. A simplified method of generating dorsoventral retina and its central projections. Developmental Biology 222, transgenic Xenopus. Nucleic Acids Res 28, E12. 460e470. Stemple, D.L., 2004. TILLING e a high-throughput harvest for functional genomics. Wawersik, S., Maas, R.L., 2000. Vertebrate eye development as modeled in Nature Reviews Genetics 5, 145e149. Drosophila. Human Molecular Genetics 9, 917e925. Stump, R.J.W., Ang, S., Chen, Y.J., von Bahr, T., Lovicu, F.J., Pinson, K., de Iongh, R.U., Wernig, M., Meissner, A., Foreman, R., Brambrink, T., Ku, M.C., Hochedlinger, K., Yamaguchi, T.P., Sassoon, D.A., McAvoy, J.W., 2003. A role for Wnt/beta-catenin Bernstein, B.E., Jaenisch, R., 2007. In vitro reprogramming of fibroblasts into signaling in lens epithelial differentiation. Developmental Biology 259, 48e61. a pluripotent ES-cell-like state. Nature 448, 318e322. Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., Wheeler, G.N., Hamilton, F.S., Hoppler, S., 2000. Inducible gene expression in Yamanaka, S., 2007. Induction of pluripotent stem cells from adult human transgenic Xenopus embryos. Current Biology 10, 849e852. fibroblasts by defined factors. Cell 131, 861e872. Wigle, J.T., Chowdhury, K., Gruss, P., Oliver, G., 1999. Prox1 function is crucial for Takahashi, K., Yamanaka, S., 2006. Induction of pluripotent stem cells from mouse mouse lens-fibre elongation. Nature Genetics 21, 318e322. embryonic and adult fibroblast cultures by defined factors. Cell 126, 663e676. Wolff, G., 1895. Entwicklungsphyiologische Studien. I. Die Regeneration der Thatcher, E.J., Paydar, I., Anderson, K.K., Patton, J.G., 2008. Regulation of zebrafish fin Urodelenlinse. Wilhelm Roux’ Arch. Entwickl. Mech. Org. 1, 380e390. regeneration by microRNAs. Proceedings of the National Academy of Sciences of Wordinger, R.J., Clark, A.F., 2007. Bone morphogenetic proteins and their receptors the United States of America 105, 18384e18389. in the eye. Experimental Biology and Medicine 232, 979e992. Tinsley, R.C., Kobel, H.R., 1996. The Biology of Xenopus. Clarendon Press, Oxford, Wride, M.A., 1996. Cellular and molecular features of lens differentiation: a review Great Briton. of recent advances. Differentiation 61, 77e93. Tomarev, S.I., 1997. Pax-6, Eyes absent, and Prox 1 in eye development. International Yamada, T., 1977. Control mechanisms in cell-type conversion in newt lens Journal of Developmental Biology 41, 835e842. regeneration. Monographs in Developmental Biology 13, 1e126. Tomarev, S.I., Sundin, O., BanerjeeBasu, S., Duncan, M.K., Yang, J.M., Piatigorsky, J., Yamada, T., 1982. Transdifferentation of lens cells and its regulation. In: 1996. Chicken homeobox gene Prox 1 related to Drosophila prospero is expressed McDevitt, D.S. (Ed.), Cell Biology of the Eye. Academic Press, New York, pp. in the developing lens and retina. Developmental Dynamics 206, 354e367. 193e242. Tomarev, S.I., Zinovieva, R.D., Chang, B., Hawes, N.L., 1998. Characterization of the Yang, X.H., 2004. Roles of cell-extrinsic growth factors in vertebrate eye pattern mouse Prox1 gene. Biochemical and Biophysical Research Communications 248, formation and retinogenesis. Seminars in Cell & Developmental Biology 15, 684e689. 91e103. Tseng, A.S., Levin, M., 2008. Tail regeneration in Xenopus laevis as a model for Yang, E.V., Bryant, S.V., 1994. Developmental regulation of a matrix metal- understanding tissue repair. Journal of Dental Research 87, 806e816. loproteinase during regeneration of axolotl appendages. Developmental Biology Tseng, S., Sun, T.T., 1999. Stem cells: ocular surface maintenance. In: Brigthbill, F. 166, 696e703. (Ed.), Corneal Surgery: Theory, Technique Tissue. Mosby, St Louis, pp. 9e18. Yang, E.V., Gardiner, D.M., Carlson, M.R.J., Nugas, C.A., Bryant, S.V., 1999. Expression Tsonis, P.A., 2000. Regeneration in vertebrates. Developmental Biology 221, of Mmp-9 and related matrix metalloproteinase genes during axolotl limb 273e284. regeneration. Developmental Dynamics 216, 2e9. Tsonis, P.A., 2004. Stem cells from differentiated cells. Molecular Interventions 4, 81. Yin, V.P., Thomson, J.M., Thummel, R., Hyde, D.R., Hammond, S.M., Poss, K.D., 2008. Tsonis, P.A., Del Rio-Tsonis, K., 2004. Lens and retina regeneration: trans- Fgf-dependent depletion of microRNA-133 promotes appendage regeneration differentiation, stem cells and clinical applications. Experimental Eye Research in zebrafish. Genes & Development 22, 728e733. 78, 161e172. Yoshii, C., Ueda, Y., Okamoto, M., Araki, M., 2007. Neural retinal regeneration in the Tsonis, P.A., Trombley, M.T., Rowland, T., Chandraratna, R.A.S., Del Rio-Tsonis, K., anuran amphibian Xenopus laevis post-metamorphosis: transdifferentiation of 2000. Role of retinoic acid in lens regeneration. Developmental Dynamics 219, retinal pigmented epithelium regenerates the neural retina. Developmental 588e593. Biology 303, 45e56. Tsonis, P.A., Tsavaris, M., Call, M.K., Chandraratna, R.A.S., Del Rio-Tsonis, K., 2002. Zaghloul, N.A., Yan, B., Moody, S.A., 2005. Step-wise specification of retinal stem Expression and role of alpha in lens regeneration. cells during normal embryogenesis. Biology of the Cell 97, 321e337. Development, Growth and Differentiation 44, 391e394. Zhang, S.S.M., Fu, X.Y., Barnstable, C.J., 2002. Molecular aspects of vertebrate retinal Tsonis, P.A., Vergara, M.N., Spence, J.R., Madhavan, M., Kramer, E.L., Call, M.K., development. Molecular Neurobiology 26, 137e152. Santiago, W.G., Vallance, J.E., Robbins, D.J., Del Rio-Tsonis, K., 2004. A novel role Zuber, M.E., Gestri, G., Viczian, A.S., Barsacchi, G., Harris, W.A., 2003. Specification of of the hedgehog pathway in lens regeneration. Developmental Biology 267, the vertebrate eye by a network of eye field transcription factors. Development 450e461. 130, 5155e5167. Tsonis, P.A., Call, M.K., Grogg, M.W., Sartor, M.A., Taylor, R.R., Forge, A., Fyffe, R., Zygar, C.A., Cook, T.L., Grainger, R.M., 1998. Gene activation during early stages of Goldenberg, R., Cowper-Sallari, R., Tomlinson, C.R., 2007. MicroRNAs and lens induction in Xenopus. Development.

Please cite this article in press as: Henry, J.J., Tsonis, P.A., Molecular and cellular aspects of amphibian lens regeneration, Progress in Retinal and Eye Research (2010), doi:10.1016/j.preteyeres.2010.07.002