Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.003
Filtrar
1.
Curr Top Dev Biol ; 159: 232-271, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38729677

RESUMO

The anterior-to-posterior (head-to-tail) body axis is extraordinarily diverse among vertebrates but conserved within species. Body axis development requires a population of axial progenitors that resides at the posterior of the embryo to sustain elongation and is then eliminated once axis extension is complete. These progenitors occupy distinct domains in the posterior (tail-end) of the embryo and contribute to various lineages along the body axis. The subset of axial progenitors with neuromesodermal competency will generate both the neural tube (the precursor of the spinal cord), and the trunk and tail somites (producing the musculoskeleton) during embryo development. These axial progenitors are called Neuromesodermal Competent cells (NMCs) and Neuromesodermal Progenitors (NMPs). NMCs/NMPs have recently attracted interest beyond the field of developmental biology due to their clinical potential. In the mouse, the maintenance of neuromesodermal competency relies on a fine balance between a trio of known signals: Wnt/ß-catenin, FGF signalling activity and suppression of retinoic acid signalling. These signals regulate the relative expression levels of the mesodermal transcription factor Brachyury and the neural transcription factor Sox2, permitting the maintenance of progenitor identity when co-expressed, and either mesoderm or neural lineage commitment when the balance is tilted towards either Brachyury or Sox2, respectively. Despite important advances in understanding key genes and cellular behaviours involved in these fate decisions, how the balance between mesodermal and neural fates is achieved remains largely unknown. In this chapter, we provide an overview of signalling and gene regulatory networks in NMCs/NMPs. We discuss mutant phenotypes associated with axial defects, hinting at the potential significant role of lesser studied proteins in the maintenance and differentiation of the progenitors that fuel axial elongation.


Assuntos
Padronização Corporal , Mesoderma , Animais , Padronização Corporal/genética , Mesoderma/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Proteínas com Domínio T/genética , Diferenciação Celular , Cabeça/embriologia
2.
Curr Top Dev Biol ; 159: 372-405, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38729682

RESUMO

The Segmentation Clock is a tissue-level patterning system that enables the segmentation of the vertebral column precursors into transient multicellular blocks called somites. This patterning system comprises a set of elements that are essential for correct segmentation. Under the so-called "Clock and Wavefront" model, the system consists of two elements, a genetic oscillator that manifests itself as traveling waves of gene expression, and a regressing wavefront that transforms the temporally periodic signal encoded in the oscillations into a permanent spatially periodic pattern of somite boundaries. Over the last twenty years, every new discovery about the Segmentation Clock has been tightly linked to the nomenclature of the "Clock and Wavefront" model. This constrained allocation of discoveries into these two elements has generated long-standing debates in the field as what defines molecularly the wavefront and how and where the interaction between the two elements establishes the future somite boundaries. In this review, we propose an expansion of the "Clock and Wavefront" model into three elements, "Clock", "Wavefront" and signaling gradients. We first provide a detailed description of the components and regulatory mechanisms of each element, and we then examine how the spatiotemporal integration of the three elements leads to the establishment of the presumptive somite boundaries. To be as exhaustive as possible, we focus on the Segmentation Clock in zebrafish. Furthermore, we show how this three-element expansion of the model provides a better understanding of the somite formation process and we emphasize where our current understanding of this patterning system remains obscure.


Assuntos
Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma , Somitos , Animais , Padronização Corporal/genética , Somitos/embriologia , Somitos/metabolismo , Mesoderma/embriologia , Mesoderma/metabolismo , Mesoderma/citologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Transdução de Sinais , Relógios Biológicos/genética
3.
FASEB J ; 38(9): e23632, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38686936

RESUMO

The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper MD development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.


Assuntos
Ductos Paramesonéfricos , Oviductos , Análise de Célula Única , Transcriptoma , Útero , Animais , Feminino , Camundongos , Útero/metabolismo , Útero/citologia , Ductos Paramesonéfricos/metabolismo , Oviductos/metabolismo , Oviductos/citologia , Perfilação da Expressão Gênica , Animais Recém-Nascidos , Diferenciação Celular , Mesoderma/metabolismo , Mesoderma/citologia , Células Epiteliais/metabolismo , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica no Desenvolvimento
4.
Int J Oral Sci ; 16(1): 33, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654018

RESUMO

Precise orchestration of cell fate determination underlies the success of scaffold-based skeletal regeneration. Despite extensive studies on mineralized parenchymal tissue rebuilding, regenerating and maintaining undifferentiated mesenchyme within calvarial bone remain very challenging with limited advances yet. Current knowledge has evidenced the indispensability of rebuilding suture mesenchymal stem cell niches to avoid severe brain or even systematic damage. But to date, the absence of promising therapeutic biomaterials/scaffolds remains. The reason lies in the shortage of fundamental knowledge and methodological evidence to understand the cellular fate regulations of scaffolds. To address these issues, in this study, we systematically investigated the cellular fate determinations and transcriptomic mechanisms by distinct types of commonly used calvarial scaffolds. Our data elucidated the natural processes without scaffold transplantation and demonstrated how different scaffolds altered in vivo cellular responses. A feasible scaffold, polylactic acid electrospinning membrane (PLA), was next identified to precisely control mesenchymal ingrowth and self-renewal to rebuild non-osteogenic suture-like tissue at the defect center, meanwhile supporting proper osteointegration with defect bony edges. Especially, transcriptome analysis and cellular mechanisms underlying the well-orchestrated cell fate determination of PLA were deciphered. This study for the first time cellularly decoded the fate regulations of scaffolds in suture-bony composite defect healing, offering clinicians potential choices for regenerating such complicated injuries.


Assuntos
Regeneração Óssea , Alicerces Teciduais , Transcriptoma , Animais , Regeneração Óssea/fisiologia , Poliésteres , Crânio/cirurgia , Células-Tronco Mesenquimais , Mesoderma/citologia , Diferenciação Celular , Engenharia Tecidual/métodos , Suturas Cranianas , Materiais Biocompatíveis
5.
PLoS Biol ; 22(4): e3002590, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38683849

RESUMO

Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.


Assuntos
Encéfalo , Diferenciação Celular , Pericitos , Fatores de Transcrição , Proteínas de Peixe-Zebra , Peixe-Zebra , Pericitos/metabolismo , Pericitos/citologia , Animais , Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Encéfalo/metabolismo , Encéfalo/embriologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Diferenciação Celular/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Regulação da Expressão Gênica no Desenvolvimento , Crista Neural/metabolismo , Crista Neural/citologia , Mesoderma/metabolismo , Mesoderma/citologia , Transdução de Sinais , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética
6.
PLoS Biol ; 22(4): e3002611, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38683880

RESUMO

As tissues grow and change shape during animal development, they physically pull and push on each other, and these mechanical interactions can be important for morphogenesis. During Drosophila gastrulation, mesoderm invagination temporally overlaps with the convergence and extension of the ectodermal germband; the latter is caused primarily by Myosin II-driven polarised cell intercalation. Here, we investigate the impact of mesoderm invagination on ectoderm extension, examining possible mechanical and mechanotransductive effects on Myosin II recruitment and polarised cell intercalation. We find that the germband ectoderm is deformed by the mesoderm pulling in the orthogonal direction to germband extension (GBE), showing mechanical coupling between these tissues. However, we do not find a significant change in Myosin II planar polarisation in response to mesoderm invagination, nor in the rate of junction shrinkage leading to neighbour exchange events. We conclude that the main cellular mechanism of axis extension, polarised cell intercalation, is robust to the mesoderm invagination pull. We find, however, that mesoderm invagination slows down the rate of anterior-posterior cell elongation that contributes to axis extension, counteracting the tension from the endoderm invagination, which pulls along the direction of GBE.


Assuntos
Drosophila melanogaster , Ectoderma , Gastrulação , Mesoderma , Miosina Tipo II , Animais , Mesoderma/embriologia , Mesoderma/citologia , Gastrulação/fisiologia , Ectoderma/citologia , Ectoderma/embriologia , Ectoderma/metabolismo , Miosina Tipo II/metabolismo , Drosophila melanogaster/embriologia , Polaridade Celular , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrião não Mamífero , Morfogênese , Padronização Corporal/fisiologia , Drosophila/embriologia
7.
J Chin Med Assoc ; 87(5): 488-497, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38451105

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) have promising potential in clinical application, whereas their limited amount and sources hinder their bioavailability. Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have become prominent options in regenerative medicine as both possess the ability to differentiate into MSCs. METHODS: Recently, our research team has successfully developed human leukocyte antigen (HLA)-homozygous iPSC cell lines with high immune compatibility, covering 13.5% of the Taiwanese population. As we deepen our understanding of the differences between these ESCs and HLA-homozygous iPSCs, our study focused on morphological observations and flow cytometry analysis of specific surface marker proteins during the differentiation of ESCs and iPSCs into MSCs. RESULTS: The results showed no significant differences between the two pluripotent stem cells, and both of them demonstrated the equivalent ability to further differentiate into adipose, cartilage, and bone cells. CONCLUSION: Our research revealed that these iPSCs with high immune compatibility exhibit the same differentiation potential as ESCs, enhancing the future applicability of highly immune-compatible iPSCs.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias , Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes Induzidas/citologia , Humanos , Células-Tronco Embrionárias/citologia , Células-Tronco Mesenquimais , Mesoderma/citologia , Células Cultivadas
8.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38355799

RESUMO

The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.


Assuntos
Animais Recém-Nascidos , Embrião de Mamíferos , Desenvolvimento Embrionário , Gástrula , Análise de Célula Única , Imagem com Lapso de Tempo , Animais , Feminino , Camundongos , Gravidez , Animais Recém-Nascidos/embriologia , Animais Recém-Nascidos/genética , Diferenciação Celular/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário/genética , Gástrula/citologia , Gástrula/embriologia , Gastrulação/genética , Rim/citologia , Rim/embriologia , Mesoderma/citologia , Mesoderma/enzimologia , Neurônios/citologia , Neurônios/metabolismo , Retina/citologia , Retina/embriologia , Somitos/citologia , Somitos/embriologia , Fatores de Tempo , Fatores de Transcrição/genética , Transcrição Gênica , Especificidade de Órgãos/genética
9.
Nature ; 626(7998): 367-376, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38092041

RESUMO

Implantation of the human embryo begins a critical developmental stage that comprises profound events including axis formation, gastrulation and the emergence of haematopoietic system1,2. Our mechanistic knowledge of this window of human life remains limited due to restricted access to in vivo samples for both technical and ethical reasons3-5. Stem cell models of human embryo have emerged to help unlock the mysteries of this stage6-16. Here we present a genetically inducible stem cell-derived embryoid model of early post-implantation human embryogenesis that captures the reciprocal codevelopment of embryonic tissue and the extra-embryonic endoderm and mesoderm niche with early haematopoiesis. This model is produced from induced pluripotent stem cells and shows unanticipated self-organizing cellular programmes similar to those that occur in embryogenesis, including the formation of amniotic cavity and bilaminar disc morphologies as well as the generation of an anterior hypoblast pole and posterior domain. The extra-embryonic layer in these embryoids lacks trophoblast and shows advanced multilineage yolk sac tissue-like morphogenesis that harbours a process similar to distinct waves of haematopoiesis, including the emergence of erythroid-, megakaryocyte-, myeloid- and lymphoid-like cells. This model presents an easy-to-use, high-throughput, reproducible and scalable platform to probe multifaceted aspects of human development and blood formation at the early post-implantation stage. It will provide a tractable human-based model for drug testing and disease modelling.


Assuntos
Desenvolvimento Embrionário , Camadas Germinativas , Hematopoese , Saco Vitelino , Humanos , Implantação do Embrião , Endoderma/citologia , Endoderma/embriologia , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Saco Vitelino/citologia , Saco Vitelino/embriologia , Mesoderma/citologia , Mesoderma/embriologia , Células-Tronco Pluripotentes Induzidas/citologia , Âmnio/citologia , Âmnio/embriologia , Corpos Embrioides/citologia , Linhagem da Célula , Biologia do Desenvolvimento/métodos , Biologia do Desenvolvimento/tendências
10.
J Biol Chem ; 299(2): 102804, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36529290

RESUMO

Fibroblast growth factor (FGF) is a multifunctional protein that exhibits a wide range of biological effects. Most commonly, it acts as a mitogen, but it also has regulatory, morphological, and endocrine effects. The four receptor subtypes of FGF are activated by more than 20 different FGF ligands. FGF2, one of the FGF ligands, is an essential factor for cell culture in stem cells for regenerative medicine; however, recombinant FGF2 is extremely unstable. Here, we successfully generated homobivalent agonistic single-domain antibodies (variable domain of heavy chain of heavy chain antibodies referred to as VHHs) that bind to domain III and induce activation of the FGF receptor 1 and thus transduce intracellular signaling. This agonistic VHH has similar biological activity (EC50) as the natural FGF2 ligand. Furthermore, we determined that the agonistic VHH could support the proliferation of human-induced pluripotent stem cells (PSCs) and human mesenchymal stem cells, which are PSCs for regenerative medicine. In addition, the agonistic VHH could maintain the ability of mesenchymal stem cells to differentiate into adipocytes or osteocytes, indicating that it could maintain the properties of PSCs. These results suggest that the VHH agonist may function as an FGF2 mimetic in cell preparation of stem cells for regenerative medicine with better cost effectiveness.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Domínios Proteicos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Anticorpos de Domínio Único , Humanos , Adipócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Ligantes , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Osteócitos/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/agonistas , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Medicina Regenerativa , Transdução de Sinais/efeitos dos fármacos , Anticorpos de Domínio Único/metabolismo , Anticorpos de Domínio Único/farmacologia
11.
Biochimie ; 207: 33-48, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36427681

RESUMO

Mesenchymal stem/stromal cells (MSCs) are multipotent somatic cells that have been widely explored in the field of regenerative medicine. MSCs possess the ability to secrete soluble factors as well as lipid bound extracellular vesicles (EVs). MSCs have gained increased interest and attention as a result of their therapeutic properties, which are thought to be attributed to their secretome. However, while the use of MSCs as whole cells pose heterogeneity concerns and survival issues post-transplantation, such limitations are absent in cell-free EV-based treatments. EVs derived from MSCs are promising therapeutic agents for a range of clinical conditions and disorders owing to their immunomodulatory, pro-regenerative, anti-inflammatory, and antifibrotic activity. Recent successes with preclinical studies using EVs for repair and regeneration of damaged tissues such as cardiac tissue, lung, liver, pancreas, bone, skin, cornea, and blood diseases are discussed in this review. We also discuss delivery strategies of EVs using biomaterials as delivery vehicles through systemic or local administration. Despite its effectiveness in preclinical investigations, the application of MSC-EV in clinical settings will necessitate careful consideration surrounding issues such as: i) scalability and isolation, ii) biodistribution, iii) targeting specific tissues, iv) quantification and characterization, and v) safety and efficacy of dosage. The future of EVs in regenerative medicine is promising yet still needs further investigation on enhancing the efficacy, scalability, and potency for clinical applications.


Assuntos
Vesículas Extracelulares , Mesoderma , Regeneração , Medicina Regenerativa , Células-Tronco , Vesículas Extracelulares/classificação , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Medicina Regenerativa/métodos , Medicina Regenerativa/normas , Medicina Regenerativa/tendências , Mesoderma/citologia , Células-Tronco/citologia , Humanos , Animais , Biotecnologia/métodos , Biotecnologia/normas , Biotecnologia/tendências
12.
Nature ; 612(7941): 732-738, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36517595

RESUMO

Our understanding of human early development is severely hampered by limited access to embryonic tissues. Due to their close evolutionary relationship with humans, nonhuman primates are often used as surrogates to understand human development but currently suffer from a lack of in vivo datasets, especially from gastrulation to early organogenesis during which the major embryonic cell types are dynamically specified. To fill this gap, we collected six Carnegie stage 8-11 cynomolgus monkey (Macaca fascicularis) embryos and performed in-depth transcriptomic analyses of 56,636 single cells. Our analyses show transcriptomic features of major perigastrulation cell types, which help shed light on morphogenetic events including primitive streak development, somitogenesis, gut tube formation, neural tube patterning and neural crest differentiation in primates. In addition, comparative analyses with mouse embryos and human embryoids uncovered conserved and divergent features of perigastrulation development across species-for example, species-specific dependency on Hippo signalling during presomitic mesoderm differentiation-and provide an initial assessment of relevant stem cell models of human early organogenesis. This comprehensive single-cell transcriptome atlas not only fills the knowledge gap in the nonhuman primate research field but also serves as an invaluable resource for understanding human embryogenesis and developmental disorders.


Assuntos
Gastrulação , Macaca fascicularis , Organogênese , Análise de Célula Única , Animais , Humanos , Camundongos , Gastrulação/genética , Macaca fascicularis/embriologia , Macaca fascicularis/genética , Organogênese/genética , Corpos Embrioides , Perfilação da Expressão Gênica , Linha Primitiva/citologia , Linha Primitiva/embriologia , Tubo Neural/citologia , Tubo Neural/embriologia , Crista Neural/citologia , Crista Neural/embriologia , Via de Sinalização Hippo , Mesoderma/citologia , Mesoderma/embriologia , Células-Tronco
13.
Development ; 149(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35156681

RESUMO

Axolotls are an important model organism for multiple types of regeneration, including functional spinal cord regeneration. Remarkably, axolotls can repair their spinal cord after a small lesion injury and can also regenerate their entire tail following amputation. Several classical signaling pathways that are used during development are reactivated during regeneration, but how this is regulated remains a mystery. We have previously identified miR-200a as a key factor that promotes successful spinal cord regeneration. Here, using RNA-seq analysis, we discovered that the inhibition of miR-200a results in an upregulation of the classical mesodermal marker brachyury in spinal cord cells after injury. However, these cells still express the neural stem cell marker sox2. In vivo cell tracking allowed us to determine that these cells can give rise to cells of both the neural and mesoderm lineage. Additionally, we found that miR-200a can directly regulate brachyury via a seed sequence in the 3'UTR of the gene. Our data indicate that miR-200a represses mesodermal cell fate after a small lesion injury in the spinal cord when only glial cells and neurons need to be replaced.


Assuntos
MicroRNAs/metabolismo , Regeneração da Medula Espinal/genética , Medula Espinal/metabolismo , Regiões 3' não Traduzidas , Ambystoma mexicanum/metabolismo , Animais , Antagomirs/metabolismo , Diferenciação Celular , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Medula Espinal/citologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Cauda/fisiologia , Via de Sinalização Wnt , beta Catenina/antagonistas & inibidores , beta Catenina/química , beta Catenina/metabolismo
14.
Development ; 149(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35179180

RESUMO

Specification of primordial germ cells requires a proportion of the cells in the posterior of the epiblast to reacquire pluripotency. A new paper in Development describes how OVOL2 is involved in regulating the balance between mesodermal fate and germ cell fate during gastrulation. We caught up with the first author, Yuki Naitou, and corresponding author, Katsuhiko Hayashi (Osaka University), to find out more about the paper and their future research.


Assuntos
Células Germinativas/metabolismo , Pesquisadores/psicologia , Fatores de Transcrição/metabolismo , Animais , Autoria , Transição Epitelial-Mesenquimal , Gastrulação , Células Germinativas/citologia , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Humanos , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
15.
Dev Cell ; 57(3): 398-414.e5, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-35134346

RESUMO

The postnatal development and maturation of the liver, the major metabolic organ, are inadequately understood. We have analyzed 52,834 single-cell transcriptomes and identified 31 cell types or states in mouse livers at postnatal days 1, 3, 7, 21, and 56. We observe unexpectedly high levels of hepatocyte heterogeneity in the developing liver and the progressive construction of the zonated metabolic functions from pericentral to periportal hepatocytes, which is orchestrated with the development of sinusoid endothelial, stellate, and Kupffer cells. Trajectory and gene regulatory analyses capture 36 transcription factors, including a circadian regulator, Bhlhe40, in programming liver development. Remarkably, we identified a special group of macrophages enriched at day 7 with a hybrid phenotype of macrophages and endothelial cells, which may regulate sinusoidal construction and Treg-cell function. This study provides a comprehensive atlas that covers all hepatic cell types and is instrumental for further dissection of liver development, metabolism, and disease.


Assuntos
Perfilação da Expressão Gênica , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Análise de Célula Única , Animais , Animais Recém-Nascidos , Comunicação Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Hematopoese , Hepatócitos/citologia , Hepatócitos/metabolismo , Macrófagos/metabolismo , Mesoderma/citologia , RNA-Seq , Fatores de Tempo , Fatores de Transcrição/metabolismo
16.
Cell Mol Life Sci ; 79(3): 158, 2022 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-35220463

RESUMO

Calvarial bone is one of the most complex sequences of developmental events in embryology, featuring a uniquely transient, pluripotent stem cell-like population known as the cranial neural crest (CNC). The skull is formed through intramembranous ossification with distinct tissue lineages (e.g. neural crest derived frontal bone and mesoderm derived parietal bone). Due to CNC's vast cell fate potential, in response to a series of inductive secreted cues including BMP/TGF-ß, Wnt, FGF, Notch, Hedgehog, Hippo and PDGF signaling, CNC enables generations of a diverse spectrum of differentiated cell types in vivo such as osteoblasts and chondrocytes at the craniofacial level. In recent years, since the studies from a genetic mouse model and single-cell sequencing, new discoveries are uncovered upon CNC patterning, differentiation, and the contribution to the development of cranial bones. In this review, we summarized the differences upon the potential gene regulatory network to regulate CNC derived osteogenic potential in mouse and human, and highlighted specific functions of genetic molecules from multiple signaling pathways and the crosstalk, transcription factors and epigenetic factors in orchestrating CNC commitment and differentiation into osteogenic mesenchyme and bone formation. Disorders in gene regulatory network in CNC patterning indicate highly close relevance to clinical birth defects and diseases, providing valuable transgenic mouse models for subsequent discoveries in delineating the underlying molecular mechanisms. We also emphasized the potential regenerative alternative through scientific discoveries from CNC patterning and genetic molecules in interfering with or alleviating clinical disorders or diseases, which will be beneficial for the molecular targets to be integrated for novel therapeutic strategies in the clinic.


Assuntos
Diferenciação Celular , Redes Reguladoras de Genes/genética , Osteogênese , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Crista Neural/citologia , Crista Neural/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
17.
Stem Cell Reports ; 17(2): 427-442, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35090587

RESUMO

Elucidating regulatory relationships between transcription factors (TFs) and target genes is fundamental to understanding how cells control their identity and behavior. Unfortunately, existing computational gene regulatory network (GRN) reconstruction methods are imprecise, computationally burdensome, and fail to reveal dynamic regulatory topologies. Here, we present Epoch, a reconstruction tool that uses single-cell transcriptomics to accurately infer dynamic networks. We apply Epoch to identify the dynamic networks underpinning directed differentiation of mouse embryonic stem cells (ESCs) guided by multiple signaling pathways, and we demonstrate that modulating these pathways drives topological changes that bias cell fate potential. We also find that Peg3 rewires the pluripotency network to favor mesoderm specification. By integrating signaling pathways with GRNs, we trace how Wnt activation and PI3K suppression govern mesoderm and endoderm specification, respectively. Finally, we identify regulatory circuits of patterning and axis formation that distinguish in vitro and in vivo mesoderm specification.


Assuntos
Redes Reguladoras de Genes/genética , Camadas Germinativas/metabolismo , Animais , Diferenciação Celular , Endoderma/citologia , Endoderma/metabolismo , Camadas Germinativas/citologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/genética , Análise de Célula Única , Proteínas Wnt/metabolismo
18.
Development ; 149(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-35020897

RESUMO

The patterned array of basal, intermediate and superficial cells in the urothelium of the mature ureter arises from uncommitted epithelial progenitors of the distal ureteric bud. Urothelial development requires signaling input from surrounding mesenchymal cells, which, in turn, depend on cues from the epithelial primordium to form a layered fibro-muscular wall. Here, we have identified FGFR2 as a crucial component in this reciprocal signaling crosstalk in the murine ureter. Loss of Fgfr2 in the ureteric epithelium led to reduced proliferation, stratification, intermediate and basal cell differentiation in this tissue, and affected cell survival and smooth muscle cell differentiation in the surrounding mesenchyme. Loss of Fgfr2 impacted negatively on epithelial expression of Shh and its mesenchymal effector gene Bmp4. Activation of SHH or BMP4 signaling largely rescued the cellular defects of mutant ureters in explant cultures. Conversely, inhibition of SHH or BMP signaling in wild-type ureters recapitulated the mutant phenotype in a dose-dependent manner. Our study suggests that FGF signals from the mesenchyme enhance, via epithelial FGFR2, the SHH-BMP4 signaling axis to drive urothelial and mesenchymal development in the early ureter.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Proteínas Hedgehog/metabolismo , Organogênese , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Ureter/metabolismo , Animais , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Ureter/embriologia , Urotélio/citologia , Urotélio/metabolismo
19.
Nature ; 602(7895): 129-134, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35082446

RESUMO

Differentiation proceeds along a continuum of increasingly fate-restricted intermediates, referred to as canalization1,2. Canalization is essential for stabilizing cell fate, but the mechanisms that underlie robust canalization are unclear. Here we show that the BRG1/BRM-associated factor (BAF) chromatin-remodelling complex ATPase gene Brm safeguards cell identity during directed cardiogenesis of mouse embryonic stem cells. Despite the establishment of a well-differentiated precardiac mesoderm, Brm-/- cells predominantly became neural precursors, violating germ layer assignment. Trajectory inference showed a sudden acquisition of a non-mesodermal identity in Brm-/- cells. Mechanistically, the loss of Brm prevented de novo accessibility of primed cardiac enhancers while increasing the expression of neurogenic factor POU3F1, preventing the binding of the neural suppressor REST and shifting the composition of BRG1 complexes. The identity switch caused by the Brm mutation was overcome by increasing BMP4 levels during mesoderm induction. Mathematical modelling supports these observations and demonstrates that Brm deletion affects cell fate trajectory by modifying saddle-node bifurcations2. In the mouse embryo, Brm deletion exacerbated mesoderm-deleted Brg1-mutant phenotypes, severely compromising cardiogenesis, and reveals an in vivo role for Brm. Our results show that Brm is a compensable safeguard of the fidelity of mesoderm chromatin states, and support a model in which developmental canalization is not a rigid irreversible path, but a highly plastic trajectory.


Assuntos
Diferenciação Celular , Linhagem da Célula , Mesoderma/citologia , Mesoderma/metabolismo , Miócitos Cardíacos/citologia , Fatores de Transcrição/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Cromatina/genética , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Embrião de Mamíferos , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Miocárdio/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fenótipo , Proteínas Repressoras/metabolismo , Células-Tronco/citologia , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
20.
Dev Biol ; 483: 76-88, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34973174

RESUMO

The chick limb bud has plasticity to reconstruct a normal skeletal pattern after a part of mesenchymal mass is excised to make a hole in its early stage of development. To understand the details of hole closure and re-establishment of normal limb axes to reconstruct a normal limb skeleton, we focused on cellular and molecular changes during hole repair and limb restoration. We excised a cube-shaped mass of mesenchymal cells from the medial region of chick hindlimb bud (stage 23) and observed the following morphogenesis. The hole had closed by 15 â€‹h after excision, followed by restoration of the limb bud morphology, and the cartilage pattern was largely restored by 48 â€‹h. Lineage analysis of the mesenchymal cells showed that cells at the anterior and posterior margins of the hole were adjoined at the hole closure site, whereas cells at the proximal and distal margins were not. To investigate cell polarity during hole repair, we analyzed intracellular positioning of the Golgi apparatus relative to the nuclei. We found that the Golgi apparatus tended to be directed toward the hole among cells at the anterior and posterior margins but not among cells at identical positions in normal limb buds or cells at the proximal and distal hole margins. In the manipulated limb buds, the frequency of cell proliferation was maintained compared with the control side. Tbx3 expression, which was usually restricted to anterior and posterior margins of the limb bud, was temporarily expanded medially and then reverted to a normal pattern as limb reconstruction proceeded, with Tbx3 negative cells reappearing in the medial regions of the limb buds. Thus, mesenchymal hole closure and limb reconstruction are mainly mediated by cells at the anterior and posterior hole margins. These results suggest that adjustment of cellular properties along the anteroposterior axis is crucial to restore limb damage and reconstruct normal skeletal patterns.


Assuntos
Padronização Corporal/fisiologia , Botões de Extremidades/citologia , Botões de Extremidades/embriologia , Células-Tronco Mesenquimais/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Esqueleto/embriologia , Animais , Proteínas Aviárias/metabolismo , Núcleo Celular/metabolismo , Polaridade Celular/fisiologia , Proliferação de Células/fisiologia , Embrião de Galinha , Extremidades/embriologia , Complexo de Golgi/metabolismo , Membro Posterior/embriologia , Transdução de Sinais/fisiologia , Esqueleto/citologia , Esqueleto/metabolismo , Proteínas com Domínio T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...