Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genesis ; 62(1): e23580, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37974491

RESUMO

Bop1 can promote cell proliferation and is a component of the Pes1-Bop1-WDR12 (PeBoW) complex that regulates ribosomal RNA processing and biogenesis. In embryos, however, bop1 mRNA is highly enriched in the neural plate, cranial neural crest and placodes, and potentially may interact with Six1, which also is expressed in these tissues. Recent work demonstrated that during development, Bop1 is required for establishing the size of the tadpole brain, retina and cranial cartilages, as well as controlling neural tissue gene expression levels. Herein, we extend this work by assessing the effects of Bop1 knockdown at neural plate and larval stages. Loss of Bop1 expanded neural plate gene expression domains (sox2, sox11, irx1) and reduced neural crest (foxd3, sox9), placode (six1, sox11, irx1, sox9) and epidermal (dlx5) expression domains. At larval stages, Bop1 knockdown reduced the expression of several otic vesicle genes (six1, pax2, irx1, sox9, dlx5, otx2, tbx1) and branchial arch genes that are required for chondrogenesis (sox9, tbx1, dlx5). The latter was not the result of impaired neural crest migration. Together these observations indicate that Bop1 is a multifunctional protein that in addition to its well-known role in ribosomal biogenesis functions during early development to establish the craniofacial precursor domains.


Assuntos
Crista Neural , Fatores de Transcrição , Crista Neural/metabolismo , Fatores de Transcrição/metabolismo , Cabeça , Crânio/metabolismo , Ribossomos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
2.
Dev Dyn ; 252(12): 1407-1427, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37597164

RESUMO

BACKGROUND: Members of the sulfotransferase superfamily (SULT) influence the activity of a wide range of hormones, neurotransmitters, metabolites and xenobiotics. However, their roles in developmental processes are not well characterized even though they are expressed during embryogenesis. We previously found in a microarray screen that Six1 up-regulates LOC100037047, which encodes XB5850668.L, an uncharacterized sulfotransferase. RESULTS: Since Six1 is required for patterning the embryonic ectoderm into its neural plate, neural crest, preplacodal and epidermal domains, we used loss- and gain-of function assays to characterize the role of XB5850668.L during this process. Knockdown of endogenous XB5850668.L resulted in the reduction of epidermal, neural crest, cranial placode and otic vesicle gene expression domains, concomitant with neural plate expansion. Increased levels had minimal effects, but infrequently expanded neural plate and neural crest gene domains, and infrequently reduced cranial placode and otic vesicle gene domains. Mutation of two key amino acids in the sulfotransferase catalytic domain required for PAPS binding and enzymatic activity tended to reduce the effects of overexpressing the wild-type protein. CONCLUSIONS: Our analyses indicates that XB5850668.L is a member of the SULT2 family that plays important roles in patterning the embryonic ectoderm. Some aspects of its influence likely depend on sulfotransferase activity.


Assuntos
Ectoderma , Crista Neural , Crista Neural/metabolismo , Crânio/metabolismo , Desenvolvimento Embrionário/genética , Sulfotransferases/genética , Sulfotransferases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
3.
Front Cell Dev Biol ; 10: 786052, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35198557

RESUMO

The embryonic ectoderm is composed of four domains: neural plate, neural crest, pre-placodal region (PPR) and epidermis. Their formation is initiated during early gastrulation by dorsal-ventral and anterior-posterior gradients of signaling factors that first divide the embryonic ectoderm into neural and non-neural domains. Next, the neural crest and PPR domains arise, either via differential competence of the neural and non-neural ectoderm (binary competence model) or via interactions between the neural and non-neural ectoderm tissues to produce an intermediate neural border zone (NB) (border state model) that subsequently separates into neural crest and PPR. Many previous gain- and loss-of-function experiments demonstrate that numerous TFs are expressed in initially overlapping zones that gradually resolve into patterns that by late neurula stages are characteristic of each of the four domains. Several of these studies suggested that this is accomplished by a combination of repressive TF interactions and competence to respond to local signals. In this study, we ectopically expressed TFs that at neural plate stages are characteristic of one domain in a different domain to test whether they act cell autonomously as repressors. We found that almost all tested TFs caused reduced expression of the other TFs. At gastrulation these effects were strictly within the lineage-labeled cells, indicating that the effects were cell autonomous, i.e., due to TF interactions within individual cells. Analysis of previously published single cell RNAseq datasets showed that at the end of gastrulation, and continuing to neural tube closure stages, many ectodermal cells express TFs characteristic of more than one neural plate stage domain, indicating that different TFs have the opportunity to interact within the same cell. At neurula stages repression was observed both in the lineage-labeled cells and in adjacent cells not bearing detectable lineage label, suggesting that cell-to-cell signaling has begun to contribute to the separation of the domains. Together, these observations directly demonstrate previous suggestions in the literature that the segregation of embryonic ectodermal domains initially involves cell autonomous, repressive TF interactions within an individual cell followed by the subsequent advent of non-cell autonomous signaling to neighbors.

4.
Dev Biol ; 462(2): 165-179, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32259520

RESUMO

Xenopus laevis frogs from laboratory stocks normally lay eggs exhibiting extensive size variability. We find that these initial size differences subsequently affect the size of the embryos prior to the onset of growth, and the size of tadpoles during the growth period. Even though these tadpoles differ in size, their tissues, organs, and structures always seem to be properly proportioned, i.e. they display static allometry. Initial axial patterning events in Xenopus occur in a spherical embryo, allowing easy documentation of their size-dependent features. We examined the size distribution of early Xenopus laevis embryos and measured diameters that differed by about 38% with a median of about 1.43 â€‹mm. This range of embryo sizes corresponds to about a 1.9-fold difference in surface area and a 2.6-fold difference in volume. We examined the relationship between embryo size and gene expression and observed a significant correlation between diameter and RNA content during gastrula stages. In addition, we investigated the expression levels of genes that pattern the mesoderm, induce the nervous system and mediate the progression of ectodermal cells to neural precursors in large and small embryos. We found that most of these factors were expressed at levels that scaled with the different embryo sizes and total embryo RNA content. In agreement with the changes in transcript levels, the expression domains in larger embryos increased proportionally with the increase in surface area, maintaining their relative expression domain size in relation to the total size of the embryo. Thus, our study identified a mechanism for adapting gene expression domains to embryo size by adjusting the transcript levels of the genes regulating mesoderm induction and patterning. In the neural plate, besides the scaling of the expression domains, we observed similar cell sizes and cell densities in small and large embryos suggesting that additional cell divisions took place in large embryos to compensate for the increased size. Our results show in detail the size variability among Xenopus laevis embryos and the transcriptional adaptation to scale gene expression with size. The observations further support the involvement of BMP/ADMP signaling in the scaling process.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Morfogênese/fisiologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Tamanho Celular , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/fisiologia , Gástrula/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mesoderma/metabolismo , Morfogênese/genética , Transdução de Sinais/fisiologia , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo
5.
Curr Top Dev Biol ; 117: 93-112, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26969974

RESUMO

Initial interest in understanding how the fertilized egg becomes a multicellular animal suggested two possible answers: either the embryo came from preformed components or it arose through epigenetic processes. Extensive research during the past few decades has identified aspects of development that depend on preformed elements, such as cytoplasmic components and a cell's lineage; it also has identified aspects that depend on epigenetic processes, such as cell interactions and morphogen gradients. These advances have depended on understanding embryonic cell lineage and cell fate. This essay explains how lineage analysis and fate mapping have contributed to our current understanding of embryonic development.


Assuntos
Diferenciação Celular , Linhagem da Célula/genética , Embrião de Mamíferos/citologia , Animais , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento
6.
Genesis ; 53(5): 308-20, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25892704

RESUMO

BMP signaling distinguishes between neural and non-neural fates by activating epidermis-specific transcription and repressing neural-specific transcription. The neural ectoderm forms after the Organizer secrets antagonists that prevent these BMP-mediated activities. However, it is not known whether neural genes also are transcriptionally activated. Therefore, we tested the ability of nine Organizer transcription factors to ectopically induce the expression of four neural ectodermal genes in epidermal precursors. We found evidence for two pathways: Foxd4 and Sox11 were only induced by Sia and Twn, whereas Gmnn and Zic2 were induced by Sia, Twn, as well as seven other Organizer transcription factors. The induction of Foxd4, Gmnn and Zic2 by Sia/Twn was both non-cell autonomous (requiring an intermediate protein) and cell autonomous (direct), whereas the induction of Sox11 required Foxd4 activity. Because direct induction by Sia/Twn could occur endogenously in the dorsal-equatorial blastula cells that give rise to both the Organizer mesoderm and the neural ectoderm, we knocked down Sia/Twn in those cells. This prevented the blastula expression of Foxd4 and Sox11, demonstrating that Sia/Twn directly activate some neural genes before the separation of the Organizer mesoderm and neural ectoderm lineages.


Assuntos
Blástula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Placa Neural/embriologia , Placa Neural/metabolismo , Ativação Transcricional , Animais , Anuros , Fatores de Transcrição/metabolismo
7.
Am J Stem Cells ; 2(2): 74-94, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23862097

RESUMO

THE EARLIEST STEPS OF EMBRYONIC NEURAL DEVELOPMENT ARE ORCHESTRATED BY SETS OF TRANSCRIPTION FACTORS THAT CONTROL AT LEAST THREE PROCESSES: the maintenance of proliferative, pluripotent precursors that expand the neural ectoderm; their transition to neurally committed stem cells comprising the neural plate; and the onset of differentiation of neural progenitors. The transition from one step to the next requires the sequential activation of each gene set and then its down-regulation at the correct developmental times. Herein, we review how these gene sets interact in a transcriptional network to regulate these early steps in neural development. A key gene in this regulatory network is FoxD4L1, a member of the forkhead box (Fox) family of transcription factors. Knock-down experiments in Xenopus embryos show that FoxD4L1 is required for the expression of the other neural transcription factors, whereas increased FoxD4L1 levels have three different effects on these genes: up-regulation of neural ectoderm precursor genes; transient down-regulation of neural plate stem cell genes; and down-regulation of neural progenitor differentiation genes. These different effects indicate that FoxD4L1 maintains neural ectodermal precursors in an immature, proliferative state, and counteracts premature neural stem cell and neural progenitor differentiation. Because it both up-regulates and down-regulates genes, we characterized the regions of the FoxD4L1 protein that are specifically involved in these transcriptional functions. We identified a transcriptional activation domain in the N-terminus and at least two domains in the C-terminus that are required for transcriptional repression. These functional domains are highly conserved in the mouse and human homologues. Preliminary studies of the related FoxD4 gene in cultured mouse embryonic stem cells indicate that it has a similar role in promoting immature neural ectodermal precursors and delaying neural progenitor differentiation. These studies in Xenopus embryos and mouse embryonic stem cells indicate that FoxD4L1/FoxD4 has the important function of regulating the balance between the genes that expand neural ectodermal precursors and those that promote neural stem/progenitor differentiation. Thus, regulating the level of expression of FoxD4 may be important in stem cell protocols designed to create immature neural cells for therapeutic uses.

8.
PLoS One ; 8(4): e61845, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23610594

RESUMO

FoxD4L1 is a forkhead transcription factor that expands the neural ectoderm by down-regulating genes that promote the onset of neural differentiation and up-regulating genes that maintain proliferative neural precursors in an immature state. We previously demonstrated that binding of Grg4 to an Eh-1 motif enhances the ability of FoxD4L1 to down-regulate target neural genes but does not account for all of its repressive activity. Herein we analyzed the protein sequence for additional interaction motifs and secondary structure. Eight conserved motifs were identified in the C-terminal region of fish and frog proteins. Extending the analysis to mammals identified a high scoring motif downstream of the Eh-1 domain that contains a tryptophan residue implicated in protein-protein interactions. In addition, secondary structure prediction programs predicted an α-helical structure overlapping with amphibian-specific Motif 6 in Xenopus, and similarly located α-helical structures in other vertebrate FoxD proteins. We tested functionality of this site by inducing a glutamine-to-proline substitution expected to break the predicted α-helical structure; this significantly reduced FoxD4L1's ability to repress zic3 and irx1. Because this mutation does not interfere with Grg4 binding, these results demonstrate that at least two regions, the Eh-1 motif and a more C-terminal predicted α-helical/Motif 6 site, additively contribute to repression. In the N-terminal region we previously identified a 14 amino acid motif that is required for the up-regulation of target genes. Secondary structure prediction programs predicted a short ß-strand separating two acidic domains. Mutant constructs show that the ß-strand itself is not required for transcriptional activation. Instead, activation depends upon a glycine residue that is predicted to provide sufficient flexibility to bring the two acidic domains into close proximity. These results identify conserved predicted motifs with secondary structures that enable FoxD4L1 to carry out its essential functions as both a transcriptional repressor and activator of neural genes.


Assuntos
Biologia Computacional , Sequência Conservada , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/metabolismo , Inativação Gênica , Ativação Transcricional , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Concentração de Íons de Hidrogênio , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo
9.
Dev Biol ; 365(2): 363-75, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22425621

RESUMO

FoxD4/5, a forkhead transcription factor, plays a critical role in establishing and maintaining the embryonic neural ectoderm. It both up-regulates genes that maintain a proliferative, immature neural ectoderm and down-regulates genes that promote the transition to a differentiating neural plate. We constructed deletion and mutant versions of FoxD4/5 to determine which domains are functionally responsible for these opposite activities, which regulate the critical developmental transition of neural precursors to neural progenitors to differentiating neural plate cells. Our results show that up-regulation of genes that maintain immature neural precursors (gem, zic2) requires the Acidic blob (AB) region in the N-terminal portion of the protein, indicating that the AB is the transactivating domain. Additionally, down-regulation of those genes that promote the transition to neural progenitors (sox) and those that lead to neural differentiation (zic, irx) involves: 1) an interaction with the Groucho co-repressor at the Eh-1 motif in the C-terminus; and 2) sequence downstream of this motif. Finally, the ability of FoxD4/5 to induce the ectopic expression of neural precursor genes in the ventral ectoderm also involves both the AB region and the Eh-1 motif; FoxD4/5 accomplishes ectopic neural induction by both activating neural precursor genes and repressing BMP signaling and epidermal genes. This study identifies the specific, conserved domains of the FoxD4/5 protein that allow this single transcription factor to regulate a network of genes that controls the transition of a proliferative neural ectodermal population to a committed neural plate population poised to begin differentiation.


Assuntos
Ectoderma/embriologia , Fatores de Transcrição Forkhead/química , Regulação da Expressão Gênica no Desenvolvimento , Placa Neural/embriologia , Ativação Transcricional , Proteínas de Xenopus/química , Sequência de Aminoácidos , Animais , Diferenciação Celular , Ectoderma/citologia , Ectoderma/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Placa Neural/citologia , Placa Neural/metabolismo , Estrutura Terciária de Proteína , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
10.
Methods Mol Biol ; 322: 1-16, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16739712

RESUMO

The National Institutes of Health Xenopus Initiative is a concerted effort to interact with the Xenopus research community to identify the community's needs; to devise strategies to meet those needs; and to support, oversee, and coordinate the resulting projects. This chapter provides a brief description of several genetic and genomic resources generated by this initiative and explains how to access them. The resources described in this chapter are (1) complementary deoxyribonucleic acid (cDNA) libraries and expressed sequence tag (EST) sequences; (2) UniGene clusters; (3) full-insert cDNA sequences; (4) a genetic map; (5) genomic libraries; (6) a physical map; (7) genome sequence; (8) microarrays; (9) mutagenesis and phenotyping; and (10) bioinformatics. The descriptions presented here were based on data that were available at the time of manuscript submission. Because these are ongoing projects, they are constantly generating new data and analyses. The Web sites cited in each subheading present current data and analyses.


Assuntos
Biologia Computacional , Biblioteca Gênica , Genômica , Internet , Xenopus/embriologia , Xenopus/genética , Animais , Biologia Computacional/métodos , Biologia Computacional/tendências , Biologia do Desenvolvimento/métodos , Biologia do Desenvolvimento/tendências , Humanos , National Institutes of Health (U.S.) , National Library of Medicine (U.S.) , Estados Unidos , Xenopus/crescimento & desenvolvimento
11.
Genome Res ; 16(6): 796-803, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16672307

RESUMO

Sequencing of full-insert clones from full-length cDNA libraries from both Xenopus laevis and Xenopus tropicalis has been ongoing as part of the Xenopus Gene Collection Initiative. Here we present 10,967 full ORF verified cDNA clones (8049 from X. laevis and 2918 from X. tropicalis) as a community resource. Because the genome of X. laevis, but not X. tropicalis, has undergone allotetraploidization, comparison of coding sequences from these two clawed (pipid) frogs provides a unique angle for exploring the molecular evolution of duplicate genes. Within our clone set, we have identified 445 gene trios, each comprised of an allotetraploidization-derived X. laevis gene pair and their shared X. tropicalis ortholog. Pairwise dN/dS, comparisons within trios show strong evidence for purifying selection acting on all three members. However, dN/dS ratios between X. laevis gene pairs are elevated relative to their X. tropicalis ortholog. This difference is highly significant and indicates an overall relaxation of selective pressures on duplicated gene pairs. We have found that the paralogs that have been lost since the tetraploidization event are enriched for several molecular functions, but have found no such enrichment in the extant paralogs. Approximately 14% of the paralogous pairs analyzed here also show differential expression indicative of subfunctionalization.


Assuntos
Sequência de Bases , Biblioteca Gênica , Poliploidia , Xenopus laevis/genética , Xenopus/genética , Animais , Evolução Molecular , Expressão Gênica , Genes Duplicados , Genoma , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Filogenia , Homologia de Sequência do Ácido Nucleico
12.
Genome Res ; 14(10B): 2121-7, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15489334

RESUMO

The National Institutes of Health's Mammalian Gene Collection (MGC) project was designed to generate and sequence a publicly accessible cDNA resource containing a complete open reading frame (ORF) for every human and mouse gene. The project initially used a random strategy to select clones from a large number of cDNA libraries from diverse tissues. Candidate clones were chosen based on 5'-EST sequences, and then fully sequenced to high accuracy and analyzed by algorithms developed for this project. Currently, more than 11,000 human and 10,000 mouse genes are represented in MGC by at least one clone with a full ORF. The random selection approach is now reaching a saturation point, and a transition to protocols targeted at the missing transcripts is now required to complete the mouse and human collections. Comparison of the sequence of the MGC clones to reference genome sequences reveals that most cDNA clones are of very high sequence quality, although it is likely that some cDNAs may carry missense variants as a consequence of experimental artifact, such as PCR, cloning, or reverse transcriptase errors. Recently, a rat cDNA component was added to the project, and ongoing frog (Xenopus) and zebrafish (Danio) cDNA projects were expanded to take advantage of the high-throughput MGC pipeline.


Assuntos
Clonagem Molecular/métodos , DNA Complementar , Biblioteca Gênica , Fases de Leitura Aberta/fisiologia , Animais , Biologia Computacional , Primers do DNA , DNA Complementar/genética , DNA Complementar/metabolismo , Humanos , Camundongos , National Institutes of Health (U.S.) , Ratos , Estados Unidos , Xenopus laevis/genética , Peixe-Zebra/genética
13.
Dev Dyn ; 225(4): 384-91, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12454917

RESUMO

The NIH Xenopus Initiative is establishing many of the genetic and genomic resources that have been recommended by the Xenopus research community. These resources include cDNA libraries, expressed sequence tags, full-length cDNA sequences, genomic libraries, pilot projects to mutagenize and phenotype X. tropicalis, and sequencing the X. tropicalis genome. This review describes the status of these projects and explains how to access their data and resources. Current information about these activities is available on the NIH Xenopus Web site (http://www.nih.gov/science/models/xenopus/).


Assuntos
Xenopus/embriologia , Xenopus/genética , Animais , DNA Complementar/metabolismo , Bases de Dados como Assunto , Biologia do Desenvolvimento/métodos , Etiquetas de Sequências Expressas , Biblioteca Gênica , Mutação , National Institutes of Health (U.S.) , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Software , Estados Unidos
14.
Rouxs Arch Dev Biol ; 199(7): 427-436, 1991 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28305619

RESUMO

Frog embryos that are treated with lithium during cleavage stages produce increased amounts of anterodorsal structures and decreased amounts of posterior and ventral structures. This alteration in pattern formation involves at least two changes in cell fate: the blastomeres that normally produce ventral structures populate anterodorsal structures, and the blastomeres that normally produce posterodorsal structures populate anterodorsal structures. The dorsalization of the ventral mesoderm has been shown to occur because Li alters the response to the ventral component of mesoderm induction. The posterior to anterior changes can be attributed to a reversal of embryonic polarity caused by a marked reduction in the dorsal involution of chordamesoderm. However, it is not known whether the reduction of chordamesodermal involution is produced secondarily by the abnormal migratory properties of the dorsalized ventral mesoderm, or whether it is caused by Li directly. In order to distinguish between these possibilities, chimeras of normal and Li-treated embryos were produced at the beginning of gastrulation. Chimeras with Li-treated ventral halves and normal dorsal halves developed into embryos with two heads and a single normal trunk, confirming the conclusion that the dorsalization of ventral mesoderm is produced directly by the Li. Chimeras with normal ventral halves and Li-treated dorsal halves developed into embryos that lacked postcephalic dorsal structures. These results indicate that the Li-produced lack of postcephalic dorsal structures does not depend on the altered ventral morphogenetic movements. The fate changes on the dorsal side of the embryo suggest that Li also alters the dorsal component of mesoderm induction.

15.
Rouxs Arch Dev Biol ; 199(4): 237-245, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28306109

RESUMO

The consistency of the frog blastula's fate map is produced, in part, because the progeny of blastomeres located in dfferent regions do not intermix with one another. We examined the cause for this restriction of intermixing in two types of cultures. In one type of culture, two groups of cells were excised from blastulae and stuck together; the movement of cells between the groups was monitored. Cells migrated more extensively between groups derived from the same region than between groups derived from different regions. In the other type of culture, a single cell was implanted into a group of cells that was excised from the blastula. The rate of division and the extent of migration of the implanted cell's clone were monitored. The implanted cell divided more rapidly among cells from its own region than among cells from a different region. Both experiments show that the restriction of intermixing that occurs between regions of the intact embryo also occurs in vitro. These results indicate that the restriction does not result secondarily from normal morphogenetic movements, which are absent from the explants, but probably from cellular interactions that limit the extent of cell migration. This limitation is correlated with a reduction in the rate of cell division.

16.
Rouxs Arch Dev Biol ; 197(5): 275-281, 1988 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28305787

RESUMO

The proteins synthesized before the 512-cell stage by Xenopus blastomeres with different fates were compared by one dimensional PAGE. Blastomeres that contributed more progeny to antero-dorsal axial structures produced proportionately more of two proteins of 225000 and 245000 daltons. Additionally, these proteins were reversibly increased in ventralized embryos and were decreased in dorsalized embryos. These observations indicate that some proteins that are synthesized during cleavage stages are expressed to different degrees in different regions of the embryo, that their expression can be correlated to cell fate in the normal embryo, and that their expression is altered quantitatively in dorsalized and ventralized embryos. The inverse relationship between the production of these proteins and the potential to produce dorsal structures in the normal and in dorsalized/ventralized embryos is consistent with a model in which cell fate is influenced by a gradient of particular proteins.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...