Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 150(23)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37840454

RESUMO

The emergence of definitive human haematopoietic stem cells (HSCs) from Carnegie Stage (CS) 14 to CS17 in the aorta-gonad-mesonephros (AGM) region is a tightly regulated process. Previously, we conducted spatial transcriptomic analysis of the human AGM region at the end of this period (CS16/CS17) and identified secreted factors involved in HSC development. Here, we extend our analysis to investigate the progression of dorso-ventral polarised signalling around the dorsal aorta over the entire period of HSC emergence. Our results reveal a dramatic increase in ventral signalling complexity from the CS13-CS14 transition, coinciding with the first appearance of definitive HSCs. We further observe stage-specific changes in signalling up to CS17, which may underpin the step-wise maturation of HSCs described in the mouse model. The data-rich resource is also presented in an online interface enabling in silico analysis of molecular interactions between spatially defined domains of the AGM region. This resource will be of particular interest for researchers studying mechanisms underlying human HSC development as well as those developing in vitro methods for the generation of clinically relevant HSCs from pluripotent stem cells.


Assuntos
Células-Tronco Hematopoéticas , Transdução de Sinais , Camundongos , Animais , Humanos , Transdução de Sinais/genética , Comunicação Celular , Perfilação da Expressão Gênica , Aorta , Mesonefro , Gônadas , Hematopoese/genética
2.
Blood ; 128(12): 1567-77, 2016 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-27421959

RESUMO

The first definitive hematopoietic stem cells (dHSCs) in the mouse emerge in the dorsal aorta of the embryonic day (E) 10.5 to 11 aorta-gonad-mesonephros (AGM) region. Notch signaling is essential for early HSC development but is dispensable for the maintenance of adult bone marrow HSCs. How Notch signaling regulates HSC formation in the embryo is poorly understood. We demonstrate here that Notch signaling is active in E10.5 HSC precursors and involves both Notch1 and Notch2 receptors, but is gradually downregulated while they progress toward dHSCs at E11.5. This downregulation is accompanied by gradual functional loss of Notch dependency. Thus, as early as at final steps in the AGM region, HSCs begin acquiring the Notch independency characteristic of adult bone marrow HSCs as part of the maturation program. Our data indicate that fine stage-dependent tuning of Notch signaling may be required for the generation of definitive HSCs from pluripotent cells.


Assuntos
Aorta/embriologia , Embrião de Mamíferos/citologia , Gônadas/embriologia , Células-Tronco Hematopoéticas/citologia , Mesonefro/embriologia , Receptor Notch2/metabolismo , Células Estromais/citologia , Animais , Aorta/metabolismo , Células Cultivadas , Embrião de Mamíferos/metabolismo , Gônadas/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Mesonefro/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais , Células Estromais/metabolismo
3.
PLoS Genet ; 11(10): e1005595, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26496642

RESUMO

Upon apoptotic stimuli, epithelial cells compensate the gaps left by dead cells by activating proliferation. This has led to the proposal that dying cells signal to surrounding living cells to maintain homeostasis. Although the nature of these signals is not clear, reactive oxygen species (ROS) could act as a signaling mechanism as they can trigger pro-inflammatory responses to protect epithelia from environmental insults. Whether ROS emerge from dead cells and what is the genetic response triggered by ROS is pivotal to understand regeneration of Drosophila imaginal discs. We genetically induced cell death in wing imaginal discs, monitored the production of ROS and analyzed the signals required for repair. We found that cell death generates a burst of ROS that propagate to the nearby surviving cells. Propagated ROS activate p38 and induce tolerable levels of JNK. The activation of JNK and p38 results in the expression of the cytokines Unpaired (Upd), which triggers the JAK/STAT signaling pathway required for regeneration. Our findings demonstrate that this ROS/JNK/p38/Upd stress responsive module restores tissue homeostasis. This module is not only activated after cell death induction but also after physical damage and reveals one of the earliest responses for imaginal disc regeneration.


Assuntos
Proteínas de Drosophila/genética , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Regeneração/genética , Fatores de Transcrição/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Animais , Apoptose/genética , Proliferação de Células/genética , Proteínas de Drosophila/biossíntese , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Discos Imaginais/crescimento & desenvolvimento , Proteínas Quinases JNK Ativadas por Mitógeno/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Estresse Fisiológico/genética , Fatores de Transcrição/biossíntese , Asas de Animais/crescimento & desenvolvimento , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
4.
Curr Opin Cell Biol ; 73: 133-140, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34717142

RESUMO

The development of the vertebrate body axis relies on the activity of different populations of axial progenitors, including neuromesodermal progenitors. Currently, the term 'Neuromesodermal progenitors' is associated with various definitions. Here, we use distinct terminologies to highlight advances in our understanding of this cell type at both the single-cell and population levels. We discuss how these recent insights prompt new opportunities to address a range of biomedical questions spanning cancer metastasis, congenital disorders, cellular metabolism, regenerative medicine, and evolution. Finally, we outline some of the major unanswered questions and propose future directions at the forefront of neuromesodermal research.


Assuntos
Padronização Corporal , Mesoderma , Consenso
5.
Elife ; 92020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32597756

RESUMO

Formation of the vertebrate postcranial body axis follows two sequential but distinct phases. The first phase generates pre-sacral structures (the so-called primary body) through the activity of the primitive streak on axial progenitors within the epiblast. The embryo then switches to generate the secondary body (post-sacral structures), which depends on axial progenitors in the tail bud. Here we show that the mammalian tail bud is generated through an independent functional developmental module, concurrent but functionally different from that generating the primary body. This module is triggered by convergent Tgfbr1 and Snai1 activities that promote an incomplete epithelial to mesenchymal transition on a subset of epiblast axial progenitors. This EMT is functionally different from that coordinated by the primitive streak, as it does not lead to mesodermal differentiation but brings axial progenitors into a transitory state, keeping their progenitor activity to drive further axial body extension.


Assuntos
Padronização Corporal , Transição Epitelial-Mesenquimal , Mesoderma/embriologia , Camundongos/embriologia , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Fatores de Transcrição da Família Snail/genética , Animais , Embrião de Mamíferos/embriologia , Camundongos/genética , Camundongos Transgênicos , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Cauda/embriologia
6.
Cell Stem Cell ; 27(5): 822-839.e8, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32946788

RESUMO

Hematopoietic stem cells (HSCs) first emerge in the embryonic aorta-gonad-mesonephros (AGM) region. Studies of model organisms defined intersecting signaling pathways that converge to promote HSC emergence predominantly in the ventral domain of the dorsal aorta. Much less is known about mechanisms driving HSC development in humans. Here, to identify secreted signals underlying human HSC development, we combined spatial transcriptomics analysis of dorsoventral polarized signaling in the aorta with gene expression profiling of sorted cell populations and single cells. Our analysis revealed a subset of aortic endothelial cells with a downregulated arterial signature and a predicted lineage relationship with the emerging HSC/progenitor population. Analysis of the ventrally polarized molecular landscape identified endothelin 1 as an important secreted regulator of human HSC development. The obtained gene expression datasets will inform future studies on mechanisms of HSC development in vivo and on generation of clinically relevant HSCs in vitro.


Assuntos
Células Endoteliais , Transcriptoma , Gônadas , Hematopoese , Células-Tronco Hematopoéticas , Humanos , Mesonefro , Transcriptoma/genética
7.
Stem Cell Reports ; 8(6): 1549-1562, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28479304

RESUMO

During development, hematopoietic stem cells (HSCs) emerge in the aorta-gonad-mesonephros (AGM) region through a process of multi-step maturation and expansion. While proliferation of adult HSCs is implicated in the balance between self-renewal and differentiation, very little is known about the proliferation status of nascent HSCs in the AGM region. Using Fucci reporter mice that enable in vivo visualization of cell-cycle status, we detect increased proliferation during pre-HSC expansion followed by a slowing down of cycling once cells start to acquire a definitive HSC state, similar to fetal liver HSCs. We observe time-specific changes in intra-aortic hematopoietic clusters corresponding to HSC maturation stages. The proliferative architecture of the clusters is maintained in an orderly anatomical manner with slowly cycling cells at the base and more actively proliferating cells at the more apical part of the cluster, which correlates with c-KIT expression levels, thus providing an anatomical basis for the role of SCF in HSC maturation.


Assuntos
Aorta/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Genes Reporter , Gônadas/metabolismo , Células-Tronco Hematopoéticas/citologia , Antígenos Comuns de Leucócito/metabolismo , Mesonefro/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA