SCCR | SETE | SNCGE | SPB | UCT | UCSF | SMM | UNC | Main Page

 

MOLECULAR mechanismS OF FROG METAMORPHOSIS

 

Yun-Bo Shi, PhD, Head, Section on Molecular Morphogenesis

Tosikazu Amano, PhD, Visiting Fellow

Liezhen Fu, PhD, Visiting Fellow

Takashi Hasebe, PhD, Visiting Fellow

ShaoChung V. Hsia, PhD, Visiting Fellow

Hiroki Matsuda, PhD, Visiting Fellow

Bindu Diana Paul, PhD, Visiting Fellow

Yukiyasu Sato, MD, PhD, Visiting Fellow

Akihiro Tomita, MD, PhD, Visiting Fellow

Daniel Buchholz, PhD, Adjunct Scientist       

 

 

The laboratory explores molecular mechanisms in amphibian metamorphosis. The control of this developmental process by thyroid hormone (TH) offers a unique paradigm in which to study gene function in postembryonic organ development. During metamorphosis, different organs undergo vastly different changes. Some, like the tail, undergo complete resorption while others, such as the limb, are developed de novo. The majority of the larval organs persist through metamorphosis but are dramatically remodeled to function in a frog. For example, tadpole intestine in Xenopus laevis is a simple tubular structure consisting mostly of a single layer of primary epithelial cells. During metamorphosis, it is transformed, through specific cell death and selective cell proliferation and differentiation, into an organ of a multiply folded adult epithelium surrounded by elaborate connective tissue and muscles. The wealth of knowledge from past research and the ability to manipulate amphibian metamorphosis both in vivo, by using transgenesis or hormone treatment of whole animals, and in vitro in organ cultures offer excellent opportunities to study the developmental function of thyroid hormone receptors (TRs) and the underlying mechanisms in vivo and to identify and functionally characterize genes that are critical for postembryonic organ development in vertebrates.

Function of TR during development

Buchholz, Tomita, Shi

Based on earlier studies, we proposed a dual-function model for TR during frog development: the heterodimers between TR and RXR (9-cis retinoic acid receptor) activate gene expression during metamorphosis when TH is present while, in premetamorphic tadpoles, they repress gene expression to prevent metamorphosis, thus ensuring a proper tadpole growth period. Such a model argues that transcriptional activation by TR is essential for frog metamorphosis. To test such a hypothesis, we previously adapted the sperm-mediated transgenic method to generate transgenic animals expressing a dominant negative TR (dnTR). Phenotypic analysis indicated that dnTR overexpression inhibits TH-induced metamorphosis. Molecular studies revealed that dnTR specifically blocks the expression of TH response genes by competing for binding to TH target promoters with endogenous wild-type TRs, leading to the retention of corepressors N-CoR and SMRT and to continued histone deacetylation even in the presence of TH. These studies thus show that gene activation is necessary for metamorphosis.

Given that TH is also known to have nongenomic effects and that, in the mouse, TH deficiency causes distinct phenotypes compared with TR-knockout animals, we were interested in determining whether gene activation by TR is sufficient for TH-dependent metamorphosis. For this purpose, we developed a dominant positive mutant TR (dpTR). In the frog oocyte transcription system, dpTR bound to a T3-responsive promoter and activated the promoter independently of T3. Transgenic expression of dpTR under the control of a heat shock–inducible promoter in premetamorphic tadpoles led to precocious metamorphic transformations. Molecular analyses showed that dpTR induced metamorphosis by specifically binding to known T3-target genes, leading to increased local histone acetylation and gene activation, similar to T3-bound TR during natural metamorphosis. Our experiments indicated that the metamorphic role of T3 operates predominantly, if not exclusively, through genomic action of the hormone. They further show, for the first time, that TR alone can directly mediate the developmental effects of T3 in individual organs by regulating target gene expression in these organs.

Buchholz DR, Hsia VS-C, Shi Y-B. A dominant negative thyroid hormone receptor blocks amphibian metamorphosis by retaining corepressors at target genes. Mol Cell Biol 2003;23:6750-6758.

Buchholz DR, Tomita A, Fu L, Paul BD, Shi Y-B. Transgenic analysis reveals that thyroid hormone receptor is sufficient to mediate the thyroid hormone signal in frog metamorphosis. Mol Cell Biol 2004;24:9026-9037.

Stewart D, Tomita A, Shi Y-B, Wong J. Chromatin immunoprecipitation for studying transcriptional regulation in Xenopus oocytes and tadpoles. In: Liu J, ed. Xenopus Protocols: Cell Biology and Signal Transduction. Totowa, NJ: Humana Press, in press.

Roles of cofactors in gene regulation by TR

Hsia, Matsuda, Paul, Sato, Tomita, Shi

TR regulates gene transcription by recruiting cofactors to target genes. In the presence of TH, TR can bind to coactivators while the unliganded TR binds to corepressors. Despite the completion of several biochemical and molecular studies on such cofactors, little is known about whether and how the cofactors participate in gene regulation by TR in different biological processes in vivo. Our focus is to investigate how TR uses different cofactors in the context of development in various organs.

To study the role of cofactors in TR function during metamorphosis, we chose several that, based on studies in tissue culture cells and in vitro, are likely to be involved in development. Thus, we have begun to study p300 and SRC, showing that Xenopus p300, SRC2, and SRC3 are expressed during metamorphosis. Using the frog oocyte system that we had established earlier for studying TR function in the context of chromatin, we recently demonstrated, by using a chromatin immunoprecipitation (ChIP) assay, that TR can recruit SRC3 to a target promoter. Furthermore, we generated a dominant negative form of SRC3 (dnSRC3) that can be recruited by TR, thus blocking the recruitment of wild-type SRC and gene activation. Similar studies on p300 suggest that it is also recruited by TR and that a dominant negative p300 can also inhibit gene activation by TH in the frog oocyte. To determine the cofactor’s roles in vivo, we are employing the ChIP assay to analyze the recruitment of both SRC3 and p300 to endogenous TH-response genes during development. Furthermore, by using sperm-mediated transgenesis, we obtained preliminary evidence to show that transgenic tadpoles expressing dnSRC3 are resistant to TH-induced as well as natural metamorphosis. We are conducting molecular studies to determine the underlying mechanisms.

Among the corepressors, we have been studying the role of N-CoR (nuclear receptor corepressor) and SMRT (silencing mediator of retinoid and thyroid receptors) in gene repression by TR in premetamorphic tadpoles. We have shown previously that both are expressed and, more important, bound to TH-response genes in premetamorphic tadpoles. The corepressors’ association with TH-response genes is released upon treatment of the tadpoles with TH, indicating that binding to TH-response gene promoters is attributable to recruitment by unliganded TR. More recently, we have shown that the corepressors are also released during natural metamorphosis when the endogenous TH level rises. Both N-CoR and SMRT are known to exist in histone deacetylase–containing complexes in mammals. Among the other proteins in the complexes is TBLR1 (transducin beta–like protein 1–related protein). We have previously shown that frog TBLR1 is expressed in tadpoles and forms complexes with both N-CoR and SMRT, at least in the frog oocyte. To investigate TBLR1’s involvement in TR function during development, we again made use of the frog oocyte system. We showed that unliganded TR interacts with TBLR1 in vivo. Using the ChIP assay, we demonstrated that unliganded TR recruits TBLR1, together with N-CoR and/or SMRT, to its target promoter, leading to histone deacetylation and gene repression in chromatin. Furthermore, a dominant negative N-CoR that contains only the TBLR1-interacting domain was able to block TBLR1 interaction with TR and derepress the promoter. The results argue that TBLR1 or related factors are required for transcription repression by unliganded TR. Finally, we showed that TBLR1 is expressed throughout frog development and is associated with endogenous TH-target promoters in premetamorphic tadpoles. More important, the association is reduced or eliminated upon treatment of the tadpoles with TH to induce metamorphosis or during natural metamorphosis when TH levels are high, suggesting that the release of TBLR1-containing corepressor complexes is one of the mechanisms by which TH response genes are activated during metamorphosis.

Jones PL, Shi Y-B. N-CoR-HDAC corepressor complexes: roles in transcriptional regulation by nuclear hormone receptors. In: Workman JL, ed. Current Topics in Microbiology and Immunology: Protein Complexes that Modify Chromatin. Berlin: Springer-Verlag, 2003;274:237-268.

Paul BD, Shi Y-B. Distinct expression profiles of transcriptional coactivators for thyroid hormone receptors during Xenopus laevis metamorphosis. Cell Res 2003;13:459-464.

Sachs LM, Jones P, Havis E, Rouse N, Demeneix BA, Shi Y-B. N-CoR recruitment by unliganded thyroid hormone receptor in gene repression during Xenopus laevis development. Mol Cell Biol 2003;22:8527-8538.

Tomita A, Buchholz DR, Shi Y-B. Fusion protein of retinoic acid receptor alpha with promyelocytic leukemia protein or promyelocytic leukemia zinc-finger protein recruits N-CoR-TBLR1 corepressor complex to repress transcription in vivo. J Biol Chem 2003;278:30788-30795.

Tomita A, Buchholz DR, Shi Y-B. Recruitment of N-CoR/SMRT-TBLR1 corepressor complex by unliganded thyroid hormone receptor for gene repression during frog development. Mol Cell Biol 2004;8:3337-3346.

Regulation and function of TH response genes during TH-induced tissue remodeling

Amano, Buchholz, Fu, Hasebe, Shi; in collaboration with Ishizuya-Oka

In an effort to identify genes important for postembryonic development, we isolated many TH response genes during metamorphosis. We have recently characterized one of them as a novel gene (ID14) expressed in tadpoles and adults of Xenopus laevis. ID14 encodes a 315–amino acid protein that has a signal peptide and a nidogen domain. Even though several genes have a nidogen domain, ID14 is not the homolog of any known gene. ID14 is a late thyroid hormone–regulated gene in the tadpole intestine, and its expression in the intestine does not begin until the climax of metamorphosis, correlating with adult intestinal epithelial differentiation. In contrast, ID14 is expressed in tadpole skin and tail throughout postembryonic development and is not regulated by thyroid hormone. In situ hybridization revealed that this putative extracellular matrix (ECM) protein is expressed in the epithelia of the tadpole skin and tail and in the intestinal epithelium after metamorphosis. In the adult, the ID14-encoded protein is found predominantly in the intestine with weak expression in the stomach, lung, and testis. Its exclusive expression in adult intestinal epithelial cells makes it a useful marker for developmental studies and may provide insights into cell-cell interactions in intestinal metamorphosis and adult intestinal stem cell maintenance.

Among the other genes regulated by TH during metamorphosis, those encoding matrix metalloproteinases (MMPs) are of particular interest for functional investigations. MMPs are extracellular enzymes capable of digesting various ECM components. Our earlier studies led us to propose that the MMP stomelysin-3 (ST3) is directly or indirectly involved in ECM remodeling, which in turn influences cell behavior in the remodeling intestine. This notion is supported by intestinal organ culture studies in vitro, in which we showed that ST3 function is important for TH-induced apoptosis of larval intestinal epithelial cells and the invasion of the proliferating adult epithelial cells into the connective tissue. To investigate directly the roles of MMPs in developing animals, we have generated transgenic tadpoles expressing wild-type ST3 or a catalytically inactive mutant under the control of a heat shock–inducible promoter. Heat shock at tadpole stages led to overexpression of wild-type or mutant ST3 in all organs. However, we observed no visible morphological changes in the tadpoles for up to a few weeks, although animals expressing wild-type ST3 transgene appeared to grow more slowly. Importantly, analysis of the intestine showed that overexpression of wild-type but not mutant ST3 caused premature apoptosis in the tadpole epithelium, accompanied by drastic remodeling of the basal lamina or the ECM that separates the connective tissue and epithelium in the intestine. Thus, our results suggest that ST3 directly or indirectly modifies the ECM, which in turn facilitates cell fate changes and tissue morphogenesis during metamorphosis. Current work aims to analyze the transgenic animals in detail, focusing on the intestine.

To determine how ST3 affects tissue remodeling, we have begun to isolate and characterize ST3 substrates. As a first step, we investigated the ability of Xenopus laevis ST3 to cleave an in vitro substrate of mammalian ST3, the alpha 1-proteinase inhibitor (PI). We analyzed the ability of the Xenopus laevis ST3 catalytic domain to cleave frog PI. Surprisingly, we found that ST3 failed to recognize the site in Xenopus PI equivalent to the major site in human PI that is cleaved by mammalian ST3. Sequence and mutagenic analyses revealed that multiple substitutions at P2-P3´ positions between human and Xenopus PI contributed to the failure of Xenopus PI to be cleaved by ST3. Our studies demonstrated that (A)(G/A)(A)(M)(F/A)(L) (P3-P3´) is a preferred cleavage site for ST3. We further demonstrated that mutations in the cleavage sites affected cleavage by ST3 in a manner that differed from several other MMPs. These findings, together with earlier reports on ST3, showed that ST3 has substrate specificities that are distinct from those of other MMPs. Our results further suggest that PI is unlikely to be a physiological substrate for ST3, at least with regard to evolutionarily conserved developmental processes.

Amano T, Liezhen F, Sahu S, Markey M, Shi Y-B. Substrate specificity of Xenopus matrix metalloproteinase stromelysin-3. Int J Mol Med 2004;14:233-239.

Buchholz DR, Fu L, Shi Y-B. Cryopreservation of Xenopus transgenic lines. Mol Reprod Dev 2004;67:65-69.

Buchholz DR, Ishizuya-Oka A, Shi Y-B. Spatial and temporal expression pattern of a novel gene in the frog Xenopus laevis: correlations with adult intestinal epithelial differentiation during metamorphosis. Gene Expr Patterns 2004;4:321-328.

Ishizuya-Oka A, Amano T, Fu L, Shi Y-B. Regulation of apoptosis by extracellular matrix during postembryonic development in Xenopus laevis. In: Lockshin RA, Zakeri Z, eds. When Cells Die II: a Comprehensive Evaluation of Apoptosis and Programmed Cell Death. New York: John Wiley & Sons, 2004;123-141.

COLLABORATOR

Atsuko Ishizuya-Oka, PhD, Department of Biology, Nippon Medical School, Tokyo, Japan


For further information, contact shi@helix.nih.gov