Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Development ; 151(18)2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39315665

RESUMO

The intricate dynamics of Hes expression across diverse cell types in the developing vertebrate embryonic tail have remained elusive. To address this, we have developed an endogenously tagged Hes1-Achilles mouse line, enabling precise quantification of dynamics at the single-cell resolution across various tissues. Our findings reveal striking disparities in Hes1 dynamics between presomitic mesoderm (PSM) and preneural tube (pre-NT) cells. While pre-NT cells display variable, low-amplitude oscillations, PSM cells exhibit synchronized, high-amplitude oscillations. Upon the induction of differentiation, the oscillation amplitude increases in pre-NT cells. Additionally, our study of Notch inhibition on Hes1 oscillations unveils distinct responses in PSM and pre-NT cells, corresponding to differential Notch ligand expression dynamics. These findings suggest the involvement of separate mechanisms driving Hes1 oscillations. Thus, Hes1 demonstrates dynamic behaviour across adjacent tissues of the embryonic tail, yet the varying oscillation parameters imply differences in the information that can be transmitted by these dynamics.


Assuntos
Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma , Análise de Célula Única , Fatores de Transcrição HES-1 , Animais , Fatores de Transcrição HES-1/metabolismo , Fatores de Transcrição HES-1/genética , Camundongos , Mesoderma/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Embrião de Mamíferos/metabolismo , Receptores Notch/metabolismo , Diferenciação Celular , Padronização Corporal , Somitos/metabolismo , Somitos/embriologia , Desenvolvimento Embrionário/genética , Cauda/embriologia
2.
Development ; 151(3)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38223992

RESUMO

The generation of the post-cranial embryonic body relies on the coordinated production of spinal cord neurectoderm and presomitic mesoderm cells from neuromesodermal progenitors (NMPs). This process is orchestrated by pro-neural and pro-mesodermal transcription factors that are co-expressed in NMPs together with Hox genes, which are essential for axial allocation of NMP derivatives. NMPs reside in a posterior growth region, which is marked by the expression of Wnt, FGF and Notch signalling components. Although the importance of Wnt and FGF in influencing the induction and differentiation of NMPs is well established, the precise role of Notch remains unclear. Here, we show that the Wnt/FGF-driven induction of NMPs from human embryonic stem cells (hESCs) relies on Notch signalling. Using hESC-derived NMPs and chick embryo grafting, we demonstrate that Notch directs a pro-mesodermal character at the expense of neural fate. We show that Notch also contributes to activation of HOX gene expression in human NMPs, partly in a non-cell-autonomous manner. Finally, we provide evidence that Notch exerts its effects via the establishment of a negative-feedback loop with FGF signalling.


Assuntos
Padronização Corporal , Genes Homeobox , Animais , Embrião de Galinha , Humanos , Padronização Corporal/genética , Diferenciação Celular/genética , Mesoderma/metabolismo , Medula Espinal , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento
3.
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
4.
Development ; 148(11)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34061173

RESUMO

During early embryogenesis, the vertebrate embryo extends from anterior to posterior because of the progressive addition of cells from a posteriorly localized neuromesodermal progenitor (NMp) population. An autoregulatory loop between Wnt and Brachyury/Tbxt is required for NMps to retain mesodermal potential and, hence, normal axis development. We recently showed that Hox13 genes help to support body axis formation and to maintain the autoregulatory loop, although the direct Hox13 target genes were unknown. Here, using a new method for identifying in vivo transcription factor-binding sites, we identified more than 500 potential Hox13 target genes in zebrafish. Importantly, we found two highly conserved Hox13-binding elements far from the tbxta transcription start site that also contain a conserved Tcf7/Lef1 (Wnt response) site. We show that the proximal of the two elements is sufficient to confer somitogenesis-stage expression to a tbxta promoter that, on its own, only drives NMp expression during gastrulation. Importantly, elimination of this proximal element produces shortened embryos due to aberrant formation of the most posterior somites. Our study provides a potential direct connection between Hox13 and regulation of the Wnt/Brachyury loop.


Assuntos
Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Animais , Sítios de Ligação , Padronização Corporal , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Somitos/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , Peixe-Zebra/embriologia
5.
Development ; 148(4)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33593754

RESUMO

The generation of the components that make up the embryonic body axis, such as the spinal cord and vertebral column, takes place in an anterior-to-posterior (head-to-tail) direction. This process is driven by the coordinated production of various cell types from a pool of posteriorly-located axial progenitors. Here, we review the key features of this process and the biology of axial progenitors, including neuromesodermal progenitors, the common precursors of the spinal cord and trunk musculature. We discuss recent developments in the in vitro production of axial progenitors and their potential implications in disease modelling and regenerative medicine.


Assuntos
Biologia , Padronização Corporal , Gastrulação/fisiologia , Camadas Germinativas/embriologia , Animais , Ectoderma/embriologia , Endoderma/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/inervação , Humanos , Técnicas In Vitro , Mesoderma/embriologia , Mesoderma/inervação , Músculo Esquelético , Células-Tronco
6.
Development ; 148(6)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33658223

RESUMO

The anteroposterior axial identity of motor neurons (MNs) determines their functionality and vulnerability to neurodegeneration. Thus, it is a crucial parameter in the design of strategies aiming to produce MNs from human pluripotent stem cells (hPSCs) for regenerative medicine/disease modelling applications. However, the in vitro generation of posterior MNs corresponding to the thoracic/lumbosacral spinal cord has been challenging. Although the induction of cells resembling neuromesodermal progenitors (NMPs), the bona fide precursors of the spinal cord, offers a promising solution, the progressive specification of posterior MNs from these cells is not well defined. Here, we determine the signals guiding the transition of human NMP-like cells toward thoracic ventral spinal cord neurectoderm. We show that combined WNT-FGF activities drive a posterior dorsal pre-/early neural state, whereas suppression of TGFß-BMP signalling pathways promotes a ventral identity and neural commitment. Based on these results, we define an optimised protocol for the generation of thoracic MNs that can efficiently integrate within the neural tube of chick embryos. We expect that our findings will facilitate the comparison of hPSC-derived spinal cord cells of distinct axial identities.


Assuntos
Diferenciação Celular/genética , Mesoderma/crescimento & desenvolvimento , Células-Tronco Neurais/metabolismo , Medula Espinal/crescimento & desenvolvimento , Animais , Padronização Corporal/genética , Proteínas Morfogenéticas Ósseas/genética , Linhagem da Célula/genética , Embrião de Galinha , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Mesoderma/metabolismo , Neurônios Motores/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Pluripotentes/citologia , Transdução de Sinais/genética , Medula Espinal/metabolismo , Fator de Crescimento Transformador beta/genética , Proteínas Wnt/genética
7.
Dev Growth Differ ; 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39287331

RESUMO

Chicken embryos have many advantages in the study of amniote embryonic development. In particular, culture techniques developed for early-stage embryos have promoted the advancement of modern developmental studies using chicken embryos. However, the standard technique involves placing chicken embryos in the ventral-upward (ventral-up) orientation, limiting manipulation of the epiblast at the dorsal surface, which is the primary source of ectodermal and mesodermal tissues. To circumvent this limitation, we developed chicken embryo cultures in the dorsal-up orientation and exploited this technique to address diverse issues. In this article, we first review the history of chicken embryo culture techniques to evaluate the advantages and limitations of the current standard technique. Then, the dorsal-up technique is discussed. These technological discussions are followed by three different examples of experimental analyses using dorsal-up cultures to illustrate their advantages: (1) EdU labeling of epiblast cells to assess potential variation in the cell proliferation rate; (2) migration behaviors of N1 enhancer-active epiblast cells revealed by tracking cells with focal fluorescent dye labeling in dorsal-up embryo culture; and (3) neural crest development of mouse neural stem cells in chicken embryos.

8.
Mol Biol Rep ; 51(1): 441, 2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38520606

RESUMO

BACKGROUND: Identification of novel cell-based therapy sources has been of great interest in recent years to provide alternative and available therapy options in clinics. Conditioned medium (CM) can be a valuable supply for growth factors, cytokines and chemokines as a source of stem cell secretome. Exploring the role of new CM sources for tissue regeneration might be a promising approach for therapeutic purposes. METHODS AND RESULTS: In the current study, neuromesodermal progenitors (NMPs) derived from induced pluripotent stem cells (iPSCs) were used to collect CM. Fibroblast derived iPSCs were successfully differentiated into NMPs and NMPs were characterized by double positive T/Bra and Sox2 staining. CM was collected from NMPs, and the content was characterized by membrane analysis. In vitro wound healing assay was used as a model system to observe potential activity of CM on cell migration. Fibroblasts, keratinocytes and endothelial cells were used to evaluate the effect of NMP-derived CM (NMP-CM) on cell migration in vitro. Several important proteins related to wound healing such as ANGPT 1, ANGPT 2, MCP-1, PDGF-AA, SDF-1α, TIMP-1 and TIMP-2 were increased in NMP-CM. NMP-CM increased cell proliferation and migration in vitro. CONCLUSIONS: In vitro data obtained from three distinct cell types suggest a promising role of NMP-CM on cell migration. NMP-CM can be used for wound management in the further future after detailed in vitro and in vivo research.


Assuntos
Células-Tronco Pluripotentes Induzidas , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Endoteliais/metabolismo , Queratinócitos/metabolismo , Movimento Celular , Proliferação de Células
9.
Dev Biol ; 489: 110-117, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35718236

RESUMO

The production of the tissues that make up the mammalian embryonic trunk takes place in a head-tail direction, via the differentiation of posteriorly-located axial progenitor populations. These include bipotent neuromesodermal progenitors (NMPs), which generate both spinal cord neurectoderm and presomitic mesoderm, the precursor of the musculoskeleton. Over the past few years, a number of studies have described the derivation of NMP-like cells from mouse and human pluripotent stem cells (PSCs). In turn, these have greatly facilitated the establishment of PSC differentiation protocols aiming to give rise efficiently to posterior mesodermal and neural cell types, which have been particularly challenging to produce using previous approaches. Moreover, the advent of 3-dimensional-based culture systems incorporating distinct axial progenitor-derived cell lineages has opened new avenues toward the functional dissection of early patterning events and cell vs non-cell autonomous effects. Here, we provide a brief overview of the applications of these cell types in disease modelling and cell therapy and speculate on their potential uses in the future.


Assuntos
Padronização Corporal , Células-Tronco Neurais , Animais , Padronização Corporal/fisiologia , Diferenciação Celular/fisiologia , Linhagem da Célula , Humanos , Mamíferos , Mesoderma , Camundongos
10.
Development ; 147(22)2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33154036

RESUMO

The early vertebrate embryo extends from anterior to posterior due to the addition of neural and mesodermal cells from a neuromesodermal progenitor (NMp) population located at the most posterior end of the embryo. In order to produce mesoderm throughout this time, the NMps produce their own niche, which is high in Wnt and low in retinoic acid. Using a loss-of-function approach, we demonstrate here that the two most abundant Hox13 genes in zebrafish have a novel role in providing robustness to the NMp niche by working in concert with the niche-establishing factor Brachyury to allow mesoderm formation. Mutants lacking both hoxa13b and hoxd13a in combination with reduced Brachyury activity have synergistic posterior body defects, in the strongest case producing embryos with severe mesodermal defects that phenocopy brachyury null mutants. Our results provide a new way of understanding the essential role of the Hox13 genes in early vertebrate development.This article has an associated 'The people behind the papers' interview.


Assuntos
Embrião não Mamífero/embriologia , Proteínas de Homeodomínio/metabolismo , Mesoderma/embriologia , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Proteínas de Homeodomínio/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
11.
Stem Cells ; 40(2): 175-189, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35257173

RESUMO

Hox genes play key roles in the anterior-posterior (AP) specification of all 3 germ layers during different developmental stages. It is only partially understood how they function in widely different developmental contexts, particularly with regards to extracellular signaling, and to what extent their function can be harnessed to guide cell specification in vitro. Here, we addressed the role of Hoxb1 in 2 distinct developmental contexts; in mouse embryonic stem cells (mES)-derived neuromesodermal progenitors (NMPs) and hindbrain neural progenitors. We found that Hoxb1 promotes NMP survival through the upregulation of Fgf8, Fgf17, and other components of Fgf signaling as well as the repression of components of the apoptotic pathway. Additionally, it upregulates other anterior Hox genes suggesting that it plays an active role in the early steps of AP specification. In neural progenitors, Hoxb1 synergizes with shh to repress anterior and dorsal neural markers, promote the expression of ventral neural markers and direct the specification of facial branchiomotorneuron (FBM)-like progenitors. Hoxb1 and shh synergize in regulating the expression of diverse signals and signaling molecules, including the Ret tyrosine kinase receptor. Finally, Hoxb1 synergizes with exogenous Glial cell line-derived neurotrophic factor (GDNF) to strengthen Ret expression and further promote the generation of FBM-like progenitors. Facial branchiomotorneuron-like progenitors survived for at least 6 months and differentiated into postmitotic neurons after orthotopic transplantation near the facial nucleus of adult mice. These results suggested that the patterning activity of Hox genes in combination with downstream signaling molecules can be harnessed for the generation of defined neural populations and transplantations with implications for neurodegenerative diseases.


Assuntos
Proteínas de Homeodomínio/metabolismo , Rombencéfalo , Animais , Diferenciação Celular/genética , Sobrevivência Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos , Rombencéfalo/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
12.
Development ; 146(12)2019 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-31152001

RESUMO

The mammalian embryo's caudal lateral epiblast (CLE) harbours bipotent progenitors, called neural mesodermal progenitors (NMPs), that contribute to the spinal cord and the paraxial mesoderm throughout axial elongation. Here, we performed a single cell analysis of different in vitro NMP populations produced either from embryonic stem cells (ESCs) or epiblast stem cells (EpiSCs) and compared them with E8.25 CLE mouse embryos. In our analysis of this region, our findings challenge the notion that NMPs can be defined by the exclusive co-expression of Sox2 and T at mRNA level. We analyse the in vitro NMP-like populations using a purpose-built support vector machine (SVM) based on the embryo CLE and use it as a classification model to compare the in vivo and in vitro populations. Our results show that NMP differentiation from ESCs leads to heterogeneous progenitor populations with few NMP-like cells, as defined by the SVM algorithm, whereas starting with EpiSCs yields a high proportion of cells with the embryo NMP signature. We find that the population from which the Epi-NMPs are derived in culture contains a node-like population, which suggests that this population probably maintains the expression of T in vitro and thereby a source of NMPs. In conclusion, differentiation of EpiSCs into NMPs reproduces events in vivo and suggests a sequence of events for the emergence of the NMP population.


Assuntos
Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Mesoderma/embriologia , Células-Tronco Pluripotentes/citologia , Células-Tronco/citologia , Animais , Padronização Corporal/genética , Diferenciação Celular , Linhagem da Célula , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Células-Tronco Neurais/citologia , Medula Espinal/embriologia , Máquina de Vetores de Suporte , Transcriptoma
13.
Development ; 146(10)2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31023877

RESUMO

The caudal lateral epiblast of mammalian embryos harbours bipotent progenitors that contribute to the spinal cord and the paraxial mesoderm in concert with the body axis elongation. These progenitors, called neural mesodermal progenitors (NMPs), are identified as cells that co-express Sox2 and T/brachyury, a criterion used to derive NMP-like cells from embryonic stem cells in vitro However, unlike embryonic NMPs, these progenitors do not self-renew. Here, we find that the protocols that yield NMP-like cells in vitro initially produce a multipotent population that, in addition to NMPs, generates progenitors for the lateral plate and intermediate mesoderm. We show that epiblast stem cells (EpiSCs) are an effective source of these multipotent progenitors, which are further differentiated by a balance between BMP and Nodal signalling. Importantly, we show that NMP-like cells derived from EpiSCs exhibit limited self-renewal in vitro and a gene expression signature like their embryonic counterparts.


Assuntos
Padronização Corporal/fisiologia , Camadas Germinativas/citologia , Células-Tronco Neurais/metabolismo , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula , Camadas Germinativas/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Células-Tronco Neurais/citologia , Fatores de Transcrição SOXB1/metabolismo
14.
Development ; 146(14)2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31235634

RESUMO

Bi-potential neuromesodermal progenitors (NMPs) produce both neural and paraxial mesodermal progenitors in the trunk and tail during vertebrate body elongation. We show that Sall4, a pluripotency-related transcription factor gene, has multiple roles in regulating NMPs and their descendants in post-gastrulation mouse embryos. Sall4 deletion using TCre caused body/tail truncation, reminiscent of early depletion of NMPs, suggesting a role of Sall4 in NMP maintenance. This phenotype became significant at the time of the trunk-to-tail transition, suggesting that Sall4 maintenance of NMPs enables tail formation. Sall4 mutants exhibit expanded neural and reduced mesodermal tissues, indicating a role of Sall4 in NMP differentiation balance. Mechanistically, we show that Sall4 promotion of WNT/ß-catenin signaling contributes to NMP maintenance and differentiation balance. RNA-Seq and SALL4 ChIP-Seq analyses support the notion that Sall4 regulates both mesodermal and neural development. Furthermore, in the mesodermal compartment, genes regulating presomitic mesoderm differentiation are downregulated in Sall4 mutants. In the neural compartment, we show that differentiation of NMPs towards post-mitotic neuron is accelerated in Sall4 mutants. Our results collectively provide evidence supporting the role of Sall4 in regulating NMPs and their descendants.


Assuntos
Padronização Corporal/genética , Linhagem da Célula/genética , Proteínas de Ligação a DNA/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Células-Tronco Neurais/citologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/genética , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Mesoderma/metabolismo , Camundongos , Células-Tronco Neurais/fisiologia , Gravidez , Via de Sinalização Wnt/fisiologia
15.
Development ; 145(14)2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30061166

RESUMO

The Myc transcriptional regulators are implicated in a range of cellular functions, including proliferation, cell cycle progression, metabolism and pluripotency maintenance. Here, we investigated the expression, regulation and function of the Myc family during mouse embryonic axis elongation and segmentation. Expression of both cMyc (Myc - Mouse Genome Informatics) and MycN in the domains in which neuromesodermal progenitors (NMPs) and underlying caudal pre-somitic mesoderm (cPSM) cells reside is coincident with WNT and FGF signals, factors known to maintain progenitors in an undifferentiated state. Pharmacological inhibition of Myc activity downregulates expression of WNT/FGF components. In turn, we find that cMyc expression is WNT, FGF and Notch protein regulated, placing it centrally in the signalling circuit that operates in the tail end that both sustains progenitors and drives maturation of the PSM into somites. Interfering with Myc function in the PSM, where it displays oscillatory expression, delays the timing of segmentation clock oscillations and thus of somite formation. In summary, we identify Myc as a component that links NMP maintenance and PSM maturation during the body axis elongation stages of mouse embryogenesis.


Assuntos
Relógios Biológicos/genética , Padronização Corporal/genética , Proteínas CLOCK/genética , Mesoderma/citologia , Mesoderma/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais/genética , Células-Tronco/metabolismo , Animais , Proteínas CLOCK/metabolismo , Diferenciação Celular , Regulação para Baixo/genética , Embrião de Mamíferos/metabolismo , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/embriologia , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Somitos/embriologia , Somitos/metabolismo , Células-Tronco/citologia , Cauda/embriologia , Proteínas Wnt/metabolismo
16.
Development ; 145(19)2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30201686

RESUMO

The vertebrate body forms by continuous generation of new tissue from progenitors at the posterior end of the embryo. The study of these axial progenitors has proved to be challenging in vivo largely because of the lack of unique molecular markers to identify them. Here, we elucidate the expression pattern of the transcription factor Nkx1-2 in the mouse embryo and show that it identifies axial progenitors throughout body axis elongation, including neuromesodermal progenitors and early neural and mesodermal progenitors. We create a tamoxifen-inducible Nkx1-2CreERT2 transgenic mouse and exploit the conditional nature of this line to uncover the lineage contributions of Nkx1-2-expressing cells at specific stages. We show that early Nkx1-2-expressing epiblast cells contribute to all three germ layers, mostly neuroectoderm and mesoderm, excluding notochord. Our data are consistent with the presence of some self-renewing axial progenitors that continue to generate neural and mesoderm tissues from the tail bud. This study identifies Nkx1-2-expressing cells as the source of most trunk and tail tissues in the mouse and provides a useful tool to genetically label and manipulate axial progenitors in vivo.


Assuntos
Linhagem da Célula , Proteínas de Homeodomínio/metabolismo , Integrases/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco/citologia , Cauda/embriologia , Tronco/embriologia , Fatores de Transcrição/metabolismo , Animais , Padronização Corporal , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Genes Reporter , Mesoderma/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Cauda/citologia
17.
Dev Biol ; 441(1): 127-131, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29964026

RESUMO

In mouse, retinoic acid (RA) is required for the early phase of body axis extension controlled by a population of neuromesodermal progenitors (NMPs) in the trunk called expanding-NMPs, but not for the later phase of body axis extension controlled by a population of NMPs in the tail called depleting-NMPs. Recent observations suggest that zebrafish utilize depleting-NMPs but not expanding-NMPs for body axis extension. In zebrafish, a role for RA in body axis extension was not supported by previous studies on aldh1a2 (raldh2) mutants lacking RA synthesis. Here, by treating zebrafish embryos with an RA synthesis inhibitor, we also found that body axis extension and somitogenesis was not perturbed, although loss of pectoral fin and cardiac edema were observed consistent with previous studies. The conclusion that zebrafish diverges from mouse in not requiring RA for body axis extension is consistent with zebrafish lacking early expanding-NMPs to generate the trunk. We suggest that RA control of body axis extension was added to higher vertebrates during evolution of expanding-NMPs.


Assuntos
Embrião de Mamíferos/embriologia , Embrião não Mamífero/embriologia , Mesoderma/embriologia , Células-Tronco Neurais/metabolismo , Tretinoína/metabolismo , Peixe-Zebra/embriologia , Animais , Embrião de Mamíferos/citologia , Embrião não Mamífero/citologia , Mesoderma/citologia , Camundongos , Células-Tronco Neurais/citologia , Especificidade da Espécie
18.
Development ; 143(2): 244-54, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26674311

RESUMO

Vertebrate body axis formation depends on a population of bipotential neuromesodermal cells along the posterior wall of the tailbud that make a germ layer decision after gastrulation to form spinal cord and mesoderm. Despite exhibiting germ layer plasticity, these cells never give rise to midline tissues of the notochord, floor plate and dorsal endoderm, raising the question of whether midline tissues also arise from basal posterior progenitors after gastrulation. We show in zebrafish that local posterior signals specify germ layer fate in two basal tailbud midline progenitor populations. Wnt signaling induces notochord within a population of notochord/floor plate bipotential cells through negative transcriptional regulation of sox2. Notch signaling, required for hypochord induction during gastrulation, continues to act in the tailbud to specify hypochord from a notochord/hypochord bipotential cell population. Our results lend strong support to a continuous allocation model of midline tissue formation in zebrafish, and provide an embryological basis for zebrafish and mouse bifurcated notochord phenotypes as well as the rare human congenital split notochord syndrome. We demonstrate developmental equivalency between the tailbud progenitor cell populations. Midline progenitors can be transfated from notochord to somite fate after gastrulation by ectopic expression of msgn1, a master regulator of paraxial mesoderm fate, or if transplanted into the bipotential progenitors that normally give rise to somites. Our results indicate that the entire non-epidermal posterior body is derived from discrete, basal tailbud cell populations. These cells remain receptive to extracellular cues after gastrulation and continue to make basic germ layer decisions.


Assuntos
Células-Tronco/citologia , Cauda/citologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Transdução de Sinais , Células-Tronco/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
19.
Proc Natl Acad Sci U S A ; 113(51): E8267-E8276, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27930333

RESUMO

TET-family dioxygenases catalyze conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) and oxidized methylcytosines in DNA. Here, we show that mouse embryonic stem cells (mESCs), either lacking Tet3 alone or with triple deficiency of Tet1/2/3, displayed impaired adoption of neural cell fate and concomitantly skewed toward cardiac mesodermal fate. Conversely, ectopic expression of Tet3 enhanced neural differentiation and limited cardiac mesoderm specification. Genome-wide analyses showed that Tet3 mediates cell-fate decisions by inhibiting Wnt signaling, partly through promoter demethylation and transcriptional activation of the Wnt inhibitor secreted frizzled-related protein 4 (Sfrp4). Tet1/2/3-deficient embryos (embryonic day 8.0-8.5) showed hyperactivated Wnt signaling, as well as aberrant differentiation of bipotent neuromesodermal progenitors (NMPs) into mesoderm at the expense of neuroectoderm. Our data demonstrate a key role for TET proteins in modulating Wnt signaling and establishing the proper balance between neural and mesodermal cell fate determination in mouse embryos and ESCs.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Mesoderma/embriologia , Proteínas Proto-Oncogênicas/metabolismo , Via de Sinalização Wnt , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/química , Animais , Diferenciação Celular , Linhagem da Célula , Metilação de DNA , Dioxigenases , Células-Tronco Embrionárias/citologia , Epigênese Genética , Estudo de Associação Genômica Ampla , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Placa Neural/embriologia
20.
Semin Cell Dev Biol ; 49: 59-67, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26658097

RESUMO

The formation of the vertebrate body depends on the precise timing and coordination of molecular and morphological events. During vertebrate embryogenesis, the paraxial mesoderm is segmented into structures called somites in a progressive fashion from the anterior to the posterior at the same time as the entire body axis elongates in the posterior direction. Evidence from several vertebrate species indicates that new paraxial mesoderm is continuously induced from neuromesodermal progenitors at the posterior-most end of the embryo. The newly forming mesoderm exists in a specialized environment called the mesodermal progenitor niche. This review will discuss how the progenitor niche coordinates the continuous addition of new mesoderm to the body axis with proper segmentation of this mesoderm upon exit from the niche. I will focus on evidence that the t-box transcription factor Brachyury and its downstream transcriptional targets serve as the primary factors coordinating mesoderm specification with somitogenesis. I will end with a discussion of recent exciting work regarding the cell-cycle and migratory behavior of mesodermal cells as they exit the progenitor niche, which may serve to further integrate new mesoderm production with proper segmentation.


Assuntos
Padronização Corporal , Mesoderma/embriologia , Animais , Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/citologia , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa