Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 861
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 187(9): 2129-2142.e17, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38670071

RESUMO

Interspecies blastocyst complementation (IBC) provides a unique platform to study development and holds the potential to overcome worldwide organ shortages. Despite recent successes, brain tissue has not been achieved through IBC. Here, we developed an optimized IBC strategy based on C-CRISPR, which facilitated rapid screening of candidate genes and identified that Hesx1 deficiency supported the generation of rat forebrain tissue in mice via IBC. Xenogeneic rat forebrain tissues in adult mice were structurally and functionally intact. Cross-species comparative analyses revealed that rat forebrain tissues developed at the same pace as the mouse host but maintained rat-like transcriptome profiles. The chimeric rate of rat cells gradually decreased as development progressed, suggesting xenogeneic barriers during mid-to-late pre-natal development. Interspecies forebrain complementation opens the door for studying evolutionarily conserved and divergent mechanisms underlying brain development and cognitive function. The C-CRISPR-based IBC strategy holds great potential to broaden the study and application of interspecies organogenesis.


Assuntos
Prosencéfalo , Animais , Prosencéfalo/metabolismo , Prosencéfalo/embriologia , Camundongos , Ratos , Blastocisto/metabolismo , Feminino , Sistemas CRISPR-Cas/genética , Transcriptoma , Organogênese , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Masculino , Camundongos Endogâmicos C57BL
2.
Cell ; 187(13): 3194-3219, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38906095

RESUMO

Developing functional organs from stem cells remains a challenging goal in regenerative medicine. Existing methodologies, such as tissue engineering, bioprinting, and organoids, only offer partial solutions. This perspective focuses on two promising approaches emerging for engineering human organs from stem cells: stem cell-based embryo models and interspecies organogenesis. Both approaches exploit the premise of guiding stem cells to mimic natural development. We begin by summarizing what is known about early human development as a blueprint for recapitulating organogenesis in both embryo models and interspecies chimeras. The latest advances in both fields are discussed before highlighting the technological and knowledge gaps to be addressed before the goal of developing human organs could be achieved using the two approaches. We conclude by discussing challenges facing embryo modeling and interspecies organogenesis and outlining future prospects for advancing both fields toward the generation of human tissues and organs for basic research and translational applications.


Assuntos
Quimera , Organogênese , Animais , Humanos , Quimera/embriologia , Implantação do Embrião , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Células-Tronco Embrionárias , Modelos Biológicos , Organoides , Medicina Regenerativa , Engenharia Tecidual/métodos
3.
Cell ; 186(6): 1212-1229.e21, 2023 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-36827974

RESUMO

Mitochondrial activity differs markedly between organs, but it is not known how and when this arises. Here we show that cell lineage-specific expression profiles involving essential mitochondrial genes emerge at an early stage in mouse development, including tissue-specific isoforms present before organ formation. However, the nuclear transcriptional signatures were not independent of organelle function. Genetically disrupting intra-mitochondrial protein synthesis with two different mtDNA mutations induced cell lineage-specific compensatory responses, including molecular pathways not previously implicated in organellar maintenance. We saw downregulation of genes whose expression is known to exacerbate the effects of exogenous mitochondrial toxins, indicating a transcriptional adaptation to mitochondrial dysfunction during embryonic development. The compensatory pathways were both tissue and mutation specific and under the control of transcription factors which promote organelle resilience. These are likely to contribute to the tissue specificity which characterizes human mitochondrial diseases and are potential targets for organ-directed treatments.


Assuntos
Mitocôndrias , Organogênese , Animais , Feminino , Humanos , Camundongos , Gravidez , Linhagem da Célula , DNA Mitocondrial/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais , Especificidade de Órgãos , Desenvolvimento Embrionário , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo
4.
Cell ; 186(10): 2092-2110.e23, 2023 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-37172563

RESUMO

The third and fourth weeks of gestation in primates are marked by several developmental milestones, including gastrulation and the formation of organ primordia. However, our understanding of this period is limited due to restricted access to in vivo embryos. To address this gap, we developed an embedded 3D culture system that allows for the extended ex utero culture of cynomolgus monkey embryos for up to 25 days post-fertilization. Morphological, histological, and single-cell RNA-sequencing analyses demonstrate that ex utero cultured monkey embryos largely recapitulated key events of in vivo development. With this platform, we were able to delineate lineage trajectories and genetic programs involved in neural induction, lateral plate mesoderm differentiation, yolk sac hematopoiesis, primitive gut, and primordial germ-cell-like cell development in monkeys. Our embedded 3D culture system provides a robust and reproducible platform for growing monkey embryos from blastocysts to early organogenesis and studying primate embryogenesis ex utero.


Assuntos
Embrião de Mamíferos , Desenvolvimento Embrionário , Animais , Macaca fascicularis , Blastocisto , Organogênese , Primatas
5.
Cell ; 186(18): 3776-3792.e16, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37478861

RESUMO

In vitro stem cell models that replicate human gastrulation have been generated, but they lack the essential extraembryonic cells needed for embryonic development, morphogenesis, and patterning. Here, we describe a robust and efficient method that prompts human extended pluripotent stem cells to self-organize into embryo-like structures, termed peri-gastruloids, which encompass both embryonic (epiblast) and extraembryonic (hypoblast) tissues. Although peri-gastruloids are not viable due to the exclusion of trophoblasts, they recapitulate critical stages of human peri-gastrulation development, such as forming amniotic and yolk sac cavities, developing bilaminar and trilaminar embryonic discs, specifying primordial germ cells, initiating gastrulation, and undergoing early neurulation and organogenesis. Single-cell RNA-sequencing unveiled transcriptomic similarities between advanced human peri-gastruloids and primary peri-gastrulation cell types found in humans and non-human primates. This peri-gastruloid platform allows for further exploration beyond gastrulation and may potentially aid in the development of human fetal tissues for use in regenerative medicine.


Assuntos
Implantação do Embrião , Gastrulação , Células-Tronco Pluripotentes , Animais , Feminino , Humanos , Gravidez , Diferenciação Celular , Embrião de Mamíferos , Desenvolvimento Embrionário , Organogênese , Células-Tronco Pluripotentes/metabolismo , Primatas
6.
Cell ; 185(11): 1960-1973.e11, 2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35551765

RESUMO

During vertebrate embryogenesis, cell collectives engage in coordinated behavior to form tissue structures of increasing complexity. In the avian skin, assembly into follicles depends on intrinsic mechanical forces of the dermis, but how cell mechanics initiate pattern formation is not known. Here, we reconstitute the initiation of follicle patterning ex vivo using only freshly dissociated avian dermal cells and collagen. We find that contractile cells physically rearrange the extracellular matrix (ECM) and that ECM rearrangement further aligns cells. This exchange transforms a mechanically unlinked collective of dermal cells into a continuum, with coherent, long-range order. Combining theory with experiment, we show that this ordered cell-ECM layer behaves as an active contractile fluid that spontaneously forms regular patterns. Our study illustrates a role for mesenchymal dynamics in generating cell-level ordering and tissue-level patterning through a fluid instability-processes that may be at play across morphological symmetry-breaking contexts.


Assuntos
Matriz Extracelular , Folículo Piloso , Animais , Colágeno , Pele , Vertebrados
7.
Cell ; 185(10): 1777-1792.e21, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35512705

RESUMO

Spatially resolved transcriptomic technologies are promising tools to study complex biological processes such as mammalian embryogenesis. However, the imbalance between resolution, gene capture, and field of view of current methodologies precludes their systematic application to analyze relatively large and three-dimensional mid- and late-gestation embryos. Here, we combined DNA nanoball (DNB)-patterned arrays and in situ RNA capture to create spatial enhanced resolution omics-sequencing (Stereo-seq). We applied Stereo-seq to generate the mouse organogenesis spatiotemporal transcriptomic atlas (MOSTA), which maps with single-cell resolution and high sensitivity the kinetics and directionality of transcriptional variation during mouse organogenesis. We used this information to gain insight into the molecular basis of spatial cell heterogeneity and cell fate specification in developing tissues such as the dorsal midbrain. Our panoramic atlas will facilitate in-depth investigation of longstanding questions concerning normal and abnormal mammalian development.


Assuntos
Organogênese , Transcriptoma , Animais , DNA/genética , Embrião de Mamíferos , Feminino , Perfilação da Expressão Gênica/métodos , Mamíferos/genética , Camundongos , Organogênese/genética , Gravidez , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma/genética
8.
Cell ; 183(3): 702-716.e14, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33125890

RESUMO

The cellular complexity and scale of the early liver have constrained analyses examining its emergence during organogenesis. To circumvent these issues, we analyzed 45,334 single-cell transcriptomes from embryonic day (E)7.5, when endoderm progenitors are specified, to E10.5 liver, when liver parenchymal and non-parenchymal cell lineages emerge. Our data detail divergence of vascular and sinusoidal endothelia, including a distinct transcriptional profile for sinusoidal endothelial specification by E8.75. We characterize two distinct mesothelial cell types as well as early hepatic stellate cells and reveal distinct spatiotemporal distributions for these populations. We capture transcriptional profiles for hepatoblast specification and migration, including the emergence of a hepatomesenchymal cell type and evidence for hepatoblast collective cell migration. Further, we identify cell-cell interactions during the organization of the primitive sinusoid. This study provides a comprehensive atlas of liver lineage establishment from the endoderm and mesoderm through to the organization of the primitive sinusoid at single-cell resolution.


Assuntos
Linhagem da Célula/genética , Fígado/citologia , Fígado/metabolismo , Análise de Célula Única , Transcriptoma/genética , Animais , Movimento Celular , Embrião de Mamíferos/citologia , Endotélio/citologia , Mesoderma/citologia , Camundongos , Transdução de Sinais , Células-Tronco/citologia
9.
Cell ; 175(3): 859-876.e33, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30318151

RESUMO

The mouse embryo has long been central to the study of mammalian development; however, elucidating the cell behaviors governing gastrulation and the formation of tissues and organs remains a fundamental challenge. A major obstacle is the lack of live imaging and image analysis technologies capable of systematically following cellular dynamics across the developing embryo. We developed a light-sheet microscope that adapts itself to the dramatic changes in size, shape, and optical properties of the post-implantation mouse embryo and captures its development from gastrulation to early organogenesis at the cellular level. We furthermore developed a computational framework for reconstructing long-term cell tracks, cell divisions, dynamic fate maps, and maps of tissue morphogenesis across the entire embryo. By jointly analyzing cellular dynamics in multiple embryos registered in space and time, we built a dynamic atlas of post-implantation mouse development that, together with our microscopy and computational methods, is provided as a resource. VIDEO ABSTRACT.


Assuntos
Linhagem da Célula , Gastrulação , Organogênese , Análise de Célula Única/métodos , Animais , Camundongos , Camundongos Endogâmicos C57BL , Modelos Estatísticos , Imagem Óptica/métodos
10.
Annu Rev Cell Dev Biol ; 34: 427-450, 2018 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-30125139

RESUMO

The nephron is a multifunctional filtration device equipped with an array of sophisticated sensors. For appropriate physiological function in the human and mouse, nephrons must be stereotypically arrayed in large numbers, and this essential structural property that defines the kidney is determined during its fetal development. This review explores the process of nephron determination in the fetal kidney, providing an overview of the foundational literature in the field as well as exploring new developments in this dynamic research area. Mechanisms that ensure that a large number of nephrons can be formed from a small initial number of progenitor cells are central to this process, and the question of how the nephron progenitor cell population balances epithelial differentiation with renewal in the progenitor state is a subject of particular interest. Key growth factor signaling pathways and transcription factor networks are discussed.


Assuntos
Diferenciação Celular/genética , Desenvolvimento Fetal/genética , Rim/crescimento & desenvolvimento , Néfrons/crescimento & desenvolvimento , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Rim/metabolismo , Néfrons/metabolismo , Organogênese/genética , Transdução de Sinais/genética , Células-Tronco/citologia
11.
Annu Rev Cell Dev Biol ; 33: 203-217, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28806099

RESUMO

As chimeras transform from beasts of Greek mythology into tools of contemporary bioscience, secrets of developmental biology and evolutionary divergence are being revealed. Recent advances in stem cell biology and interspecies chimerism have generated new models with extensive basic and translational applications, including generation of transplantable, patient-specific organs.


Assuntos
Quimera/metabolismo , Mamíferos/metabolismo , Animais , Humanos , Organogênese , Especificidade da Espécie
12.
Physiol Rev ; 103(3): 1899-1964, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-36656056

RESUMO

The teeth are vertebrate-specific, highly specialized organs performing fundamental functions of mastication and speech, the maintenance of which is crucial for orofacial homeostasis and is further linked to systemic health and human psychosocial well-being. However, with limited ability for self-repair, the teeth can often be impaired by traumatic, inflammatory, and progressive insults, leading to high prevalence of tooth loss and defects worldwide. Regenerative medicine holds the promise to achieve physiological restoration of lost or damaged organs, and in particular an evolving framework of developmental engineering has pioneered functional tooth regeneration by harnessing the odontogenic program. As a key event of tooth morphogenesis, mesenchymal condensation dictates dental tissue formation and patterning through cellular self-organization and signaling interaction with the epithelium, which provides a representative to decipher organogenetic mechanisms and can be leveraged for regenerative purposes. In this review, we summarize how mesenchymal condensation spatiotemporally assembles from dental stem cells (DSCs) and sequentially mediates tooth development. We highlight condensation-mimetic engineering efforts and mechanisms based on ex vivo aggregation of DSCs, which have achieved functionally robust and physiologically relevant tooth regeneration after implantation in animals and in humans. The discussion of this aspect will add to the knowledge of development-inspired tissue engineering strategies and will offer benefits to propel clinical organ regeneration.


Assuntos
Regeneração Óssea , Mesoderma , Odontogênese , Engenharia Tecidual , Perda de Dente , Dente , Dente/crescimento & desenvolvimento , Engenharia Tecidual/métodos , Humanos , Animais , Mesoderma/crescimento & desenvolvimento , Perda de Dente/terapia
13.
Immunity ; 54(12): 2795-2811.e9, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34788601

RESUMO

Lymphangitis and the formation of tertiary lymphoid organs (TLOs) in the mesentery are features of Crohn's disease. Here, we examined the genesis of these TLOs and their impact on disease progression. Whole-mount and intravital imaging of the ileum and ileum-draining collecting lymphatic vessels (CLVs) draining to mesenteric lymph nodes from TNFΔARE mice, a model of ileitis, revealed TLO formation at valves of CLVs. TLOs obstructed cellular and molecular outflow from the gut and were sites of lymph leakage and backflow. Tumor necrosis factor (TNF) neutralization begun at early stages of TLO formation restored lymph transport. However, robustly developed, chronic TLOs resisted regression and restoration of flow after TNF neutralization. TNF stimulation of cultured lymphatic endothelial cells reprogrammed responses to oscillatory shear stress, preventing the induction of valve-associated genes. Disrupted transport of immune cells, driven by loss of valve integrity and TLO formation, may contribute to the pathology of Crohn's disease.


Assuntos
Doença de Crohn/imunologia , Células Endoteliais/imunologia , Íleo/imunologia , Linfa/metabolismo , Vasos Linfáticos/imunologia , Mesentério/imunologia , Estruturas Linfoides Terciárias/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Ileíte , Linfangite , Camundongos , Camundongos Knockout , Estresse Mecânico
14.
Development ; 151(2)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38265192

RESUMO

The autonomic nervous system innervates the pancreas by sympathetic, parasympathetic and sensory branches during early organogenesis, starting with neural crest cell invasion and formation of an intrinsic neuronal network. Several studies have demonstrated that signals from pancreatic neural crest cells direct pancreatic endocrinogenesis. Likewise, autonomic neurons have been shown to regulate pancreatic islet formation, and have also been implicated in type I diabetes. Here, we provide an overview of recent progress in mapping pancreatic innervation and understanding the interactions between pancreatic neurons, epithelial morphogenesis and cell differentiation. Finally, we discuss pancreas innervation as a factor in the development of diabetes.


Assuntos
Diabetes Mellitus , Ilhotas Pancreáticas , Humanos , Diferenciação Celular , Organogênese , Pâncreas
15.
Development ; 151(2)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38174902

RESUMO

To gain insight into the transcription programs activated during the formation of Drosophila larval structures, we carried out single cell RNA sequencing during two periods of Drosophila embryogenesis: stages 10-12, when most organs are first specified and initiate morphological and physiological specialization; and stages 13-16, when organs achieve their final mature architectures and begin to function. Our data confirm previous findings with regards to functional specialization of some organs - the salivary gland and trachea - and clarify the embryonic functions of another - the plasmatocytes. We also identify two early developmental trajectories in germ cells and uncover a potential role for proteolysis during germline stem cell specialization. We identify the likely cell type of origin for key components of the Drosophila matrisome and several commonly used Drosophila embryonic cell culture lines. Finally, we compare our findings with other recent related studies and with other modalities for identifying tissue-specific gene expression patterns. These data provide a useful community resource for identifying many new players in tissue-specific morphogenesis and functional specialization of developing organs.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Transcriptoma/genética , Organogênese , Proteínas de Drosophila/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento
16.
Plant Cell ; 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39012965

RESUMO

During nutrient scarcity, plants can adapt their developmental strategy to maximize their chance of survival. Such plasticity in development is underpinned by hormonal regulation, which mediates the relationship between environmental cues and developmental outputs. In legumes, endosymbiosis with nitrogen fixing bacteria (rhizobia) is a key adaptation for supplying the plant with nitrogen in the form of ammonium. Rhizobia are housed in lateral root-derived organs termed nodules that maintain an environment conducive to Nitrogenase in these bacteria. Several phytohormones are important for regulating the formation of nodules, with both positive and negative roles proposed for gibberellin (GA). In this study, we determine the cellular location and function of bioactive GA during nodule organogenesis using a genetically-encoded second generation GA biosensor, GIBBERELLIN PERCEPTION SENSOR 2 in Medicago truncatula. We find endogenous bioactive GA accumulates locally at the site of nodule primordia, increasing dramatically in the cortical cell layers, persisting through cell divisions and maintaining accumulation in the mature nodule meristem. We show, through mis-expression of GA catabolic enzymes that suppress GA accumulation, that GA acts as a positive regulator of nodule growth and development. Furthermore, increasing or decreasing GA through perturbation of biosynthesis gene expression can increase or decrease the size of nodules, respectively. This is unique from lateral root formation, a developmental program that shares common organogenesis regulators. We link GA to a wider gene regulatory program by showing that nodule-identity genes induce and sustain GA accumulation necessary for proper nodule formation.

17.
Immunity ; 48(6): 1258-1270.e6, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29884461

RESUMO

Thymus development is critical to the adaptive immune system, yet a comprehensive transcriptional framework capturing thymus organogenesis at single-cell resolution is still needed. We applied single-cell RNA sequencing (RNA-seq) to capture 8 days of thymus development, perturbations of T cell receptor rearrangement, and in vitro organ cultures, producing profiles of 24,279 cells. We resolved transcriptional heterogeneity of developing lymphocytes, and genetic perturbation confirmed T cell identity of conventional and non-conventional lymphocytes. We characterized maturation dynamics of thymic epithelial cells in vivo, classified cell maturation state in a thymic organ culture, and revealed the intrinsic capacity of thymic epithelium to preserve transcriptional regularity despite exposure to exogenous retinoic acid. Finally, by integrating the cell atlas with human genome-wide association study (GWAS) data and autoimmune-disease-related genes, we implicated embryonic thymus-resident cells as possible participants in autoimmune disease etiologies. This resource provides a single-cell transcriptional framework for biological discovery and molecular analysis of thymus organogenesis.


Assuntos
Diferenciação Celular/imunologia , Análise de Sequência de RNA/métodos , Linfócitos T/imunologia , Timo/embriologia , Animais , Doenças Autoimunes/imunologia , Embrião de Mamíferos , Perfilação da Expressão Gênica/métodos , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Organogênese/imunologia , Linfócitos T/citologia , Timo/citologia
18.
Proc Natl Acad Sci U S A ; 121(40): e2410269121, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39320918

RESUMO

Organ architecture is established during development through intricate cell-cell communication mechanisms, yet the specific signals mediating these communications often remain elusive. Here, we used the anterior pituitary gland that harbors different interdigitated hormone-secreting homotypic cell networks to dissect cell-cell communication mechanisms operating during late development. We show that blocking differentiation of corticotrope cells leads to pituitary hypoplasia with a major effect on somatotrope cells that directly contact corticotropes. Gene knockout of the corticotrope-restricted transcription factor Tpit results in fewer somatotropes, with less secretory granules and a loss of cell polarity, resulting in systemic growth retardation. Single-cell transcriptomic analyses identified FGF1 as a corticotrope-specific Tpit dosage-dependent target gene responsible for these phenotypes. Consistently, genetic ablation of FGF1 in mice phenocopies pituitary hypoplasia and growth impairment observed in Tpit-deficient mice. These findings reveal FGF1 produced by the corticotrope cell network as an essential paracrine signaling molecule participating in pituitary architecture and size.


Assuntos
Fator 1 de Crescimento de Fibroblastos , Camundongos Knockout , Comunicação Parácrina , Hipófise , Animais , Camundongos , Fator 1 de Crescimento de Fibroblastos/metabolismo , Fator 1 de Crescimento de Fibroblastos/genética , Hipófise/metabolismo , Hipófise/citologia , Corticotrofos/metabolismo , Transdução de Sinais , Adeno-Hipófise/metabolismo , Adeno-Hipófise/citologia , Diferenciação Celular , Somatotrofos/metabolismo , Comunicação Celular
19.
Genes Dev ; 33(5-6): 258-275, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824532

RESUMO

Pbx genes encode transcription factors that belong to the TALE (three-amino-acid loop extension) superclass of homeodomain proteins. We have witnessed a surge in information about the roles of this gene family as leading actors in the transcriptional control of development. PBX proteins represent a clear example of how transcription factors can regulate developmental processes by combinatorial properties, acting within multimeric complexes to implement activation or repression of transcription depending on their interaction partners. Here, we revisit long-emphasized functions of PBX transcription factors as cofactors for HOX proteins, major architects of the body plan. We further discuss new knowledge on roles of PBX proteins in different developmental contexts as upstream regulators of Hox genes-as factors that interact with non-HOX proteins and can work independently of HOX-as well as potential pioneer factors. Committed to building a perfect body, PBX proteins govern regulatory networks that direct essential morphogenetic processes and organogenesis in vertebrate development. Perturbations of PBX-dependent networks can cause human congenital disease and cancer.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Organogênese/genética , Vertebrados/embriologia , Vertebrados/genética , Animais , Genes Homeobox/genética , Humanos
20.
Development ; 150(5)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36912416

RESUMO

During mammalian development, gonadal sex determination results from the commitment of bipotential supporting cells to Sertoli or granulosa cell fates. Typically, this decision is coordinated across the gonad to ensure commitment to a single organ fate. When unified commitment fails in an XY mouse, an ovotestis forms in which supporting cells in the center of the gonad typically develop as Sertoli cells, while supporting cells in the poles develop as granulosa cells. This central bias for Sertoli cell fate was thought to result from the initial expression of the drivers of Sertoli cell fate, SRY and/or SOX9, in the central domain, followed by paracrine expansion to the poles. However, we show here that the earliest cells expressing SRY and SOX9 are widely distributed across the gonad. In addition, Sertoli cell fate does not spread among supporting cells through paracrine relay. Instead, we uncover a center-biased pattern of supporting cell precursor ingression that occurs in both sexes and results in increased supporting cell density in the central domain. Our findings prompt a new model of gonad patterning in which a density-dependent organizing principle dominates Sertoli cell fate stabilization.


Assuntos
Gônadas , Processos de Determinação Sexual , Feminino , Camundongos , Masculino , Animais , Gônadas/metabolismo , Células de Sertoli/metabolismo , Diferenciação Celular , Desenvolvimento Embrionário , Fatores de Transcrição SOX9/metabolismo , Testículo/metabolismo , Proteína da Região Y Determinante do Sexo/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Mamíferos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA