Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Semin Cell Dev Biol ; 127: 37-45, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34840081

RESUMO

The discovery of mesoderm inducing signals helped usher in the era of molecular developmental biology, and today the mechanisms of mesoderm induction and patterning are still intensely studied. Mesoderm induction begins during gastrulation, but recent evidence in vertebrates shows that this process continues after gastrulation in a group of posteriorly localized cells called neuromesodermal progenitors (NMPs). NMPs reside within the post-gastrulation embryonic structure called the tailbud, where they make a lineage decision between ectoderm (spinal cord) and mesoderm. The majority of NMP-derived mesoderm generates somites, but also contributes to lateral mesoderm fates such as endothelium. The discovery of NMPs provides a new paradigm in which to study vertebrate mesoderm induction. This review will discuss mechanisms of mesoderm induction within NMPs, and how they have informed our understanding of mesoderm induction more broadly within vertebrates as well as animal species outside of the vertebrate lineage. Special focus will be given to the signaling networks underlying NMP-derived mesoderm induction and patterning, as well as emerging work on the significance of partial epithelial-mesenchymal states in coordinating cell fate and morphogenesis.


Assuntos
Padronização Corporal , Mesoderma , Animais , Diferenciação Celular , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Somitos
2.
Biochem Biophys Res Commun ; 672: 27-35, 2023 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-37331168

RESUMO

Pluripotent stem cells possess the potential to differentiate into all three germ layers. However, upon removal of the stemness factors, pluripotent stem cells, such as embryonic stem cells (ESCs), exhibit EMT-like cell behavior and lose stemness signatures. This process involves the membrane translocation of the t-SNARE protein syntaxin4 (Stx4) and the expression of the intercellular adhesion molecule P-cadherin. The forced expression of either of these elements induces the emergence of such phenotypes even in the presence of stemness factors. Interestingly, extracellular Stx4, but not P-cadherin, appears to induce a significant upregulation of the gastrulation-related gene brachyury, along with a slight upregulation of the smooth muscle cell-related gene ACTA2 in ESCs. Furthermore, our findings reveal that extracellular Stx4 plays a role in preventing the elimination of CCAAT enhancer binding protein ß (C/EBPß). Notably, the forced overexpression of C/EBPß led to the downregulation of brachyury and a significant upregulation of ACTA2 in ESCs. These observations suggest that extracellular Stx4 contributes to early mesoderm induction while simultaneously activating an element that alters the differentiation state. The fact that a single differentiation cue can elicit multiple differentiation responses may reflect the challenges associated with achieving sensitive and directed differentiation in cultured stem cells.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT , Células-Tronco Pluripotentes , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias , Caderinas/metabolismo , Células Cultivadas
3.
Int J Mol Sci ; 23(18)2022 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-36142249

RESUMO

Progesterone treatment is commonly employed to promote and support pregnancy. While maternal tissues are the main progesterone targets in humans and mice, its receptor (PGR) is expressed in the murine embryo, questioning its function during embryonic development. Progesterone has been previously associated with murine blastocyst development. Whether it contributes to lineage specification is largely unknown. Gastrulation initiates lineage specification and generation of the progenitors contributing to all organs. Cells passing through the primitive streak (PS) will give rise to the mesoderm and endoderm. Cells emerging posteriorly will form the extraembryonic mesodermal tissues supporting embryonic growth. Cells arising anteriorly will contribute to the embryonic heart in two sets of distinct progenitors, first (FHF) and second heart field (SHF). We found that PGR is expressed in a posterior-anterior gradient in the PS of gastrulating embryos. We established in vitro differentiation systems inducing posterior (extraembryonic) and anterior (cardiac) mesoderm to unravel PGR function. We discovered that PGR specifically modulates extraembryonic and cardiac mesoderm. Overexpression experiments revealed that PGR safeguards cardiac differentiation, blocking premature SHF progenitor specification and sustaining the FHF progenitor pool. This role of PGR in heart development indicates that progesterone administration should be closely monitored in potential early-pregnancy patients undergoing infertility treatment.


Assuntos
Gástrula , Gastrulação , Receptores de Progesterona , Animais , Diferenciação Celular , Feminino , Gástrula/fisiologia , Humanos , Mesoderma , Camundongos , Gravidez , Progesterona/metabolismo , Receptores de Progesterona/metabolismo
4.
Int J Mol Sci ; 22(15)2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34360992

RESUMO

Several protocols exist for generating megakaryocytes (MKs) and platelets from human induced pluripotent stem cells (hiPSCs) with limited efficiency. We observed previously that mesoderm induction improved endothelial and stromal differentiation. We, therefore, hypothesized that a protocol modification prior to hemogenic endothelial cell (HEC) differentiation will improve MK progenitor (MKP) production and increase platelet output. We further asked if basic media composition affects MK maturation. In an iterative process, we first compared two HEC induction protocols. We found significantly more HECs using the modified protocol including activin A and CHIR99021, resulting in significantly increased MKs. MKs released comparable platelet amounts irrespective of media conditions. In a final validation phase, we obtained five-fold more platelets per hiPSC with the modified protocol (235 ± 84) compared to standard conditions (51 ± 15; p < 0.0001). The regenerative potency of hiPSC-derived platelets was compared to adult donor-derived platelets by profiling angiogenesis-related protein expression. Nineteen of 24 angiogenesis-related proteins were expressed equally, lower or higher in hiPSC-derived compared to adult platelets. The hiPSC-platelet's coagulation hyporeactivity compared to adult platelets was confirmed by thromboelastometry. Further stepwise improvement of hiPSC-platelet production will, thus, permit better identification of platelet-mediated regenerative mechanisms and facilitate manufacture of sufficient amounts of functional platelets for clinical application.


Assuntos
Plaquetas/citologia , Diferenciação Celular , Técnicas de Reprogramação Celular/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Megacariócitos/citologia , Células Cultivadas , Meios de Cultura/química , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo
5.
Development ; 144(6): 1008-1017, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28279973

RESUMO

Cardiomyocytes and endothelial cells in the heart are in close proximity and in constant dialogue. Endothelium regulates the size of the heart, supplies oxygen to the myocardium and secretes factors that support cardiomyocyte function. Robust and predictive cardiac disease models that faithfully recapitulate native human physiology in vitro would therefore ideally incorporate this cardiomyocyte-endothelium crosstalk. Here, we have generated and characterized human cardiac microtissues in vitro that integrate both cell types in complex 3D structures. We established conditions for simultaneous differentiation of cardiomyocytes and endothelial cells from human pluripotent stem cells following initial cardiac mesoderm induction. The endothelial cells expressed cardiac markers that were also present in primary cardiac microvasculature, suggesting cardiac endothelium identity. These cell populations were further enriched based on surface markers expression, then recombined allowing development of beating 3D structures termed cardiac microtissues. This in vitro model was robustly reproducible in both embryonic and induced pluripotent stem cells. It thus represents an advanced human stem cell-based platform for cardiovascular disease modelling and testing of relevant drugs.


Assuntos
Diferenciação Celular , Células Endoteliais/citologia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Antígenos CD34/metabolismo , Separação Celular , Fenômenos Eletrofisiológicos , Humanos , Mesoderma/citologia , Células-Tronco Pluripotentes/metabolismo , Sarcômeros/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
Development ; 140(24): 4903-13, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24301465

RESUMO

Mesoderm and embryonic axis formation in vertebrates is mediated by maternal and zygotic factors that activate the expression of target genes. Transcriptional derepression plays an important role in the regulation of expression in different contexts; however, its involvement and possible mechanism in mesoderm and embryonic axis formation are largely unknown. Here we demonstrate that XDSCR6, a Xenopus homologue of human Down syndrome critical region protein 6 (DSCR6, or RIPPLY3), regulates mesoderm and embryonic axis formation through derepression of polycomb group (PcG) proteins. Xdscr6 maternal mRNA is enriched in the endoderm of the early gastrula and potently triggers the formation of dorsal mesoderm and neural tissues in ectoderm explants; it also dorsalises ventral mesoderm during gastrulation and induces a secondary embryonic axis. A WRPW motif, which is present in all DSCR6 homologues, is necessary and sufficient for the dorsal mesoderm- and axis-inducing activity. Knockdown of Xdscr6 inhibits dorsal mesoderm gene expression and results in head deficiency. We further show that XDSCR6 physically interacts with PcG proteins through the WRPW motif, preventing the formation of PcG bodies and antagonising their repressor activity in embryonic axis formation. By chromatin immunoprecipitation, we demonstrate that XDSCR6 releases PcG proteins from chromatin and allows dorsal mesoderm gene transcription. Our studies suggest that XDSCR6 might function to sequester PcG proteins and identify a novel derepression mechanism implicated in embryonic induction and axis formation.


Assuntos
Padronização Corporal/fisiologia , Mesoderma/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/embriologia , Animais , Cromatina/metabolismo , Síndrome de Down/genética , Ectoderma/embriologia , Ectoderma/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Indução Embrionária , Gástrula/citologia , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/embriologia , Proteínas do Grupo Polycomb/biossíntese , RNA Mensageiro , Somitos/embriologia , Fatores de Transcrição , Transcrição Gênica , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/genética
7.
Stem Cells ; 32(1): 191-203, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24022884

RESUMO

In the mouse embryo and differentiating embryonic stem cells, the hematopoietic, endothelial, and cardiomyocyte lineages are derived from Flk1+ mesodermal progenitors. Here, we report that surface expression of Podocalyxin (Podxl), a member of the CD34 family of sialomucins, can be used to subdivide the Flk1+ cells in differentiating embryoid bodies at day 4.75 into populations that develop into distinct mesodermal lineages. Definitive hematopoietic potential was restricted to the Flk1+Podxl+ population, while the Flk1-negative Podxl+ population displayed only primitive erythroid potential. The Flk1+Podxl-negative population contained endothelial cells and cardiomyocyte potential. Podxl expression distinguishes Flk1+ mesoderm populations in mouse embryos at days 7.5, 8.5, and 9.5 and is a marker of progenitor stage primitive erythroblasts. These findings identify Podxl as a useful tool for separating distinct mesodermal lineages.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/metabolismo , Mesoderma/metabolismo , Células-Tronco Pluripotentes/metabolismo , Sialoglicoproteínas/biossíntese , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Células-Tronco Pluripotentes/citologia , Sialoglicoproteínas/metabolismo , Análise Serial de Tecidos
8.
Differentiation ; 88(1): 13-15, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25113967

RESUMO

John Gurdon has made major contributions to developmental biology in addition to his Nobel prize winning work on nuclear reprogramming. With the frog, Xenopus, as a vertebrate model, his work on mesoderm induction led him to identify a community effect required for tissue differentiation after progenitor cells have entered a specific mesodermal programme. It is in the context of this biologically important concept, with myogenesis as an example, that we have had most scientific exchanges. Here I trace my contacts with him, from an interest in histone regulation of gene expression and reprogramming, to myogenic determination factors as markers of early mesodermal induction, to the role of the community effect in the spatiotemporal control of skeletal muscle formation. I also recount some personal anecdotes from encounters in Oxford, Paris and Cambridge, to illustrate my appreciation of him as a scientist and a colleague.


Assuntos
Mesoderma/citologia , Animais , Diferenciação Celular , História do Século XX , História do Século XXI , Xenopus
9.
Cells Dev ; 178: 203903, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38295873

RESUMO

The discovery of the Spemann-Mangold organizer strongly influenced subsequent research on embryonic induction, with research aiming to elucidate the molecular characteristics of organizer activity being currently underway. Herein, we review the history of research on embryonic induction, and describe how the mechanisms of induction phenomena and developmental processes have been investigated. Classical experiments investigating the differentiation capacity and inductive activity of various embryonic regions were conducted by many researchers, and important theories of region-specific induction and the concept for chain of induction were proposed. The transition from experimental embryology to developmental biology has enabled us to understand the mechanisms of embryonic induction at the molecular level. Consequently, many inducing substances and molecules such as transcriptional factors and peptide growth factors involved in the organizer formation were identified. One of peptide growth factors, activin, acts as a mesoderm- and endoderm-inducing substance. Activin induces several tissues and organs from the undifferentiated cell mass of amphibian embryos in a concentration-dependent manner. We review the extent to which we can control in vitro organogenesis from undifferentiated cells, and discuss the application to stem cell-based regenerative medicine based on insights gained from animal experiments, such as in amphibians.


Assuntos
Ativinas , Mesoderma , Animais , Mesoderma/metabolismo , Ativinas/metabolismo , Organizadores Embrionários/metabolismo , Indução Embrionária , Humanos , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento
10.
Front Cell Dev Biol ; 10: 887987, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35693948

RESUMO

Nodal proteins provide crucial signals for mesoderm and endoderm induction. In zebrafish embryos, the nodal genes ndr1/squint and ndr2/cyclops are implicated in mesendoderm induction. It remains elusive how ndr1 and ndr2 expression is regulated spatiotemporally. Here we investigated regulation of ndr1 and ndr2 expression using Mhwa mutants that lack the maternal dorsal determinant Hwa with deficiency in ß-catenin signaling, Meomesa mutants that lack maternal Eomesodermin A (Eomesa), Meomesa;Mhwa double mutants, and the Nodal signaling inhibitor SB431542. We show that ndr1 and ndr2 expression is completely abolished in Meomesa;Mhwa mutant embryos, indicating an essential role of maternal eomesa and hwa. Hwa-activated ß-catenin signaling plays a major role in activation of ndr1 expression in the dorsal blastodermal margin, while eomesa is mostly responsible for ndr1 expression in the lateroventral margin and Nodal signaling contributes to ventral expansion of the ndr1 expression domain. However, ndr2 expression mainly depends on maternal eomesa with minor or negligible contribution of maternal hwa and Nodal autoregulation. These mechanisms may help understand regulation of Nodal expression in other species.

11.
J Mol Cell Biol ; 12(2): 138-151, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31291647

RESUMO

The transforming growth factor-ß (TGF-ß) family controls embryogenesis, stem cell differentiation, and tissue homeostasis. However, how post-translation modifications contribute to fine-tuning of TGF-ß family signaling responses is not well understood. Inhibitory (I)-Smads can antagonize TGF-ß/Smad signaling by recruiting Smurf E3 ubiquitin ligases to target the active TGF-ß receptor for proteasomal degradation. A proteomic interaction screen identified Vpr binding protein (VprBP) as novel binding partner of Smad7. Mis-expression studies revealed that VprBP negatively controls Smad2 phosphorylation, Smad2-Smad4 interaction, as well as TGF-ß target gene expression. VprBP was found to promote Smad7-Smurf1-TßRI complex formation and induce proteasomal degradation of TGF-ß type I receptor (TßRI). Moreover, VprBP appears to stabilize Smurf1 by suppressing Smurf1 poly-ubiquitination. In multiple adult and mouse embryonic stem cells, depletion of VprBP promotes TGF-ß or Activin-induced responses. In the mouse embryo VprBP expression negatively correlates with mesoderm marker expression, and VprBP attenuated mesoderm induction during zebrafish embryogenesis. Our findings thereby uncover a novel regulatory mechanism by which Smurf1 controls the TGF-ß and Activin cascade and identify VprBP as a critical determinant of embryonic mesoderm induction.


Assuntos
Ativinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteólise , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ativinas/farmacologia , Animais , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Células HaCaT , Células Hep G2 , Humanos , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/genética , Proteína Smad7/genética , Proteína Smad7/metabolismo , Transfecção , Fator de Crescimento Transformador beta/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética , Peixe-Zebra/embriologia
12.
Transl Cancer Res ; 9(1): 346-357, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35117188

RESUMO

BACKGROUND: The mesoderm induction early response 1, family member 3 (MIER3) gene has been recognized as potentially being associated with cancer. However, in relation to the development of non-small cell lung cancer (NSCLC), the expression pattern and the role of MIER3 are yet to be reported. The aim of this research was to investigate the rate of expression of MIER3 in NSCLC cells and tissues and to investigate the role of MIER3 in NSCLC. METHODS: Seventeen patients received NSCLC tissues and corresponding healthy tissues. MTT assay was used to determine cell proliferation. For detecting mRNA and protein expression, we used both quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot method. To measure cell apoptosis and cell cycle distribution, we applied the flow cytometry technique. We used a wound-healing assay and a Transwell invasion assay to study cell migration and invasion. RESULTS: In comparison with adjacent normal tissues, the expression of MIER3 was down-regulated in NSCLC tissues. In addition, the level of MIER3 in NSCLC cell lines was also lower than in pulmonary epithelial cell BEAS-2B. Moreover, when MIER3 was overexpressed, cell proliferation, migration, and invasion were significantly inhibited, apoptosis increased, and cell cycle arrest was induced in A549 and H460 cells. MIER3 overexpression also suppressed tumor growth in NSCLC xenograft mouse models. Furthermore, our study demonstrated that MIER3 down-regulated the Wnt/ß-catenin signaling pathway in NSCLC cells. More importantly, MIER3 decreased the activity of histone acetyltransferase (HAT) p300, which may have contributed to its regulation on ß-catenin and tumorigenesis. CONCLUSIONS: The data suggests MIER3 takes on the tumor-suppressor role in the progression of NSCLC and, therefore, could prove to be a valuable clinical marker in the prognosis of the disease.

13.
Cell Rep ; 21(1): 37-46, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28978482

RESUMO

The early Xenopus laevis embryo is replete with dynamic spatial waves. One such wave, the cell division wave, emerges from the collective cell division timing of first tens and later hundreds of cells throughout the embryo. Here, we show that cell division waves do not propagate between neighboring cells and do not rely on cell-to-cell coupling to maintain their division timing. Instead, intrinsic variation in division period autonomously and gradually builds these striking patterns of cell division. Disrupting this pattern of division by placing embryos in a temperature gradient resulted in highly asynchronous entry to the midblastula transition and misexpression of the mesodermal marker Xbra. Remarkably, this gene expression defect is corrected during involution, resulting in delayed yet normal Xbra expression and viable embryos. This implies the existence of a previously unknown mechanism for normalizing mesodermal gene expression during involution.


Assuntos
Relógios Biológicos/genética , Mesoderma/metabolismo , Proteínas com Domínio T/genética , Proteínas de Xenopus/genética , Xenopus laevis/embriologia , Animais , Divisão Celular , Temperatura Baixa , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/citologia , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
14.
Dev Growth Differ ; 37(3): 303-309, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37281913

RESUMO

Single animal hemisphere blastomeres isolated from the eight-cell stage Xenopus embryos differentiate into mesoderm when treated with activin A, whereas when cultured without activin they form atypical epidermis. The mesoderm tissue induced by activin is different between dorsal and ventral blastomeres. In the present study, the duration and timing of activin treatment was varied, in order to identify the critical stage when animal blastomeres acquire competence to respond to activin A. It was shown that the critical time was 45 min after blastomere isolation, which corresponds approximately to NF stage 6 (32-cell stage) of normal development. The competence gradually increased during the morula stages.

15.
Dev Growth Differ ; 38(6): 625-634, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37281281

RESUMO

The kidney has been used as a model organ to analyze organogenesis. In in vitro experiments using Xenopus blastula ectoderm, the development of pronephric tubules (the prototype of the kidney) may be induced by treatment with activin A and retinoic acid (RA). The present study examined whether pronephric tubules induced in ectodermal explants exhibited similar characteristics to those of normal embryos at the molecular level. The experimental conditions required for high frequency induction (100%) of pronephric tubule formation from presumptive ectoderm without the development of muscle and notochord were determined. The developmental expression of the pronephros marker genes Xlim-1 and Xlcaax-1 was examined in induced pronephric tubules. After treatment with 10 ng/mL activin A and 10-4 mol/L RA, only pronephric tubules were induced at a high frequency. Induced pronephric tubules showed the same timing and patterns of expression for the marker genes Xlim-1 and Xlcaax-1 as normal embryos. These results suggest that the in vitro development of pronephric tubules induced in the presumptive ectoderm by activin A and RA parallels normal development at the molecular level.

16.
Dev Growth Differ ; 38(6): 577-582, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37281037

RESUMO

Mitogen-activated protein kinase (MAPK) was originally identified as a serine/threonine protein kinase that is rapidly activated in response to various growth factors and tumor promoters in mammalian cultured cells. The kinase cascade including MAPK and its direct activator, MAPK kinase (MAPKK), is now believed to transmit various extracellular signals into their intracellular targets in eukaryotic cells. It has been reported that activation of MAPKK and MAPK occurs during the meiotic maturation of oocytes in several species, including Xenopus laevis. Studies with neutralizing antibodies against MAPKK, MAPK phosphatases and constitutively active MAPKK or MAPK have revealed a crucial role of the MAPKK/MAPK cascade in a number of developmental processes in Xenopus oocytes and embryos.

17.
Dev Growth Differ ; 37(5): 581-588, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37281476

RESUMO

Mesoderm of early vertebrate embryos gradually acquires dorsal-ventral polarity during embryogenesis. This specification of mesoderm is thought to be regulated by several polypeptide growth factors. Bone morphogenetic protein (BMP), a member of the TGF-ß family, is one of the regulators suggested to be involved in the formation of ventral mesoderm. In this paper, the nature of the endogenous BMP signal in dorsal-ventral specification was assessed in early Xenopus embryos using a dominant negative mutant of the Xenopus BMP receptor. In ectodermal explant assays, disruption of endogenous BMP signaling by the mutant receptor changed the competence of the explant cells to mesoderm-inducing factors, activin and basic fibroblast growth factor (bFGF), and led to formation of neural tissue without mesoderm induction. This result suggests that endogenous BMP acts as a ventral mesoderm modifier rather than a ventral mesoderm inducer, and that interactions between endogenous BMP and mesoderm-inducing factors may be important in dorsal-ventral patterning of embryonic mesoderm. In addition, the induction of neural tissue by inhibition of the BMP signaling pathway also suggests involvement of BMP in neural induction.

18.
Proc Jpn Acad Ser B Phys Biol Sci ; 82(7): 197-207, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25792783

RESUMO

Mesoderm induction as a result of the interaction between endoderm and ectoderm is one of the most crucial events in vertebrate development. We identified activin as a strong mesoderm-inducing factor in an animal cap assay, an in vitro assay system using amphibian pluripotential cell mass. Activin induces mesodermal tisswes including most dorsal mesodermal tissue, notochord (which has important roles in neural induction, somite segmentation, and endodermal organogenesis), and its effects are concentration-dependent. Animal cap cells treated with high concentrations of activin differentiate into anterior endoderm, which can act as an organizer, or center of body patterning. We have established an in vitro induction system for 22 different organs and tissues using animal cap cells, and have isolated many organ-specific genes. With these useful methods, and analysis of newly isolated tissue- and organ-specific genes, the molecular biological "road map" for organogenesis is being established.

19.
Rouxs Arch Dev Biol ; 201(1): 18-29, 1992 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28305608

RESUMO

During the years 1973 and 1974, non-dorsal/ventral (non-d/v)-polarized blastulae were made of animal (upper) ectodermal caps and vegetal (lower) endodermal yolk masses by means of the destruction of the d/v polarity of the yolk mass endoderm by disaggregation, stirring and subsequent reaggregation. A simultaneous dis- and reaggregation of the upper ectodermal cap, as performed in the 1970 and 1971 experiments, led to the same results. The destruction of d/v polarity in the reconstituted blastulae, under maintenance of their animal-vegetal polarity, did not therefore prevent dorsal axis formation. The formation of single, double, triple or multiple dorsal axes in these reconstituted embryos must be based upon the self-organizing capacity of the induced meso-endoderm, since the alternative assumption of the re-establishment of mesoderm-inducing centre(s) in the reaggregated yolk mass endoderm seems very unlikely. The formation of single, double, triple or multiple axes must be the result of a competition between mesodermal aggregation centres arising in an initially evenly-induced zone of meso-endoderm. These initial centres which compete for like cells, attract each other, resulting, e.g. in their fusion into a single dorsal axis system, or in the survival of two, three or more centres in an initially balanced spatial configuration. Intensification of the meso-endoderm-inducing capacity, by using dorsal instead of entire yolk endoderm mass and by using two instead of one dorsal yolk mass, led to a more complete type of dorsal axis formation and to a higher number of axes formed (a higher percentage of triple and multiple axes than double and single ones) in one and the same reaggregate. The possible nature of the self-organizing capacity of the induced meso-endoderm has briefly been discussed, emphasizing the fundamental significance of the self-organizing capacity of embryonic anlagen in developmental processes.

20.
Rouxs Arch Dev Biol ; 198(1): 8-13, 1989 May.
Artigo em Inglês | MEDLINE | ID: mdl-28305777

RESUMO

We have compared the chemical properties and biological activities of the mesoderm-inducing factor that is secreted by the Xenopus XTC cell line with the vegetalizing factor from chicken embryos. The inducing activity of the factors was tested in different concentrations on totipotent ectoderm either by implantation into early gastrulae of Triturm alpestris or by application of solutions to isolated ectoderm of early gastrulae of Xenopus laevis. Both factors have similar properties. They are not irreversibly inactivated after treatment with 6 M urea or with phenol at 60° C. Reduction with thioglycolic acid inactivates the factors completely. The inducing activity of XTC-conditioned medium decreases only slightly after treatment with 50% formic acid. The apparent molecular mass and the isoelectric point of the factors are similar. The XTC factor was partially purified by size-exclusion and reversed-phase high-pressure liquid chromatography and by isoelectric focusing. The possible relationship of these factors to transforming growth factor ß is discussed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA