Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Development ; 150(1)2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36602140

RESUMO

Recent studies have highlighted the crucial role of the aorta microenvironment in the generation of the first haematopoietic stem cells (HSCs) from specialized haemogenic endothelial cells (HECs). Despite more than two decades of investigations, we require a better understanding of the cellular and molecular events driving aorta formation and polarization, which will be pivotal to establish the mechanisms that operate during HEC specification and HSC competency. Here, we outline the early mechanisms involved in vertebrate aorta formation by comparing four different species: zebrafish, chicken, mouse and human. We highlight how this process, which is tightly controlled in time and space, requires a coordinated specification of several cell types, in particular endothelial cells originating from distinct mesodermal tissues. We also discuss how molecular signals originating from the aorta environment result in its polarization, creating a unique entity for HSC generation.


Assuntos
Hemangioblastos , Peixe-Zebra , Animais , Humanos , Camundongos , Peixe-Zebra/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Hemangioblastos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Aorta , Diferenciação Celular , Hematopoese
2.
Blood ; 136(7): 831-844, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32457985

RESUMO

The defined location of a stem cell within a niche regulates its fate, behavior, and molecular identity via a complex extrinsic regulation that is far from being fully elucidated. To explore the molecular characteristics and key components of the aortic microenvironment, where the first hematopoietic stem cells are generated during development, we performed genome-wide RNA tomography sequencing on zebrafish, chicken, mouse, and human embryos. The resulting anterior-posterior and dorsal-ventral transcriptional maps provided a powerful resource for exploring genes and regulatory pathways active in the aortic microenvironment. By performing interspecies comparative RNA sequencing analyses and functional assays, we explored the complexity of the aortic microenvironment landscape and the fine-tuning of various factors interacting to control hematopoietic stem cell generation, both in time and space in vivo, including the ligand-receptor couple ADM-RAMP2 and SVEP1. Understanding the regulatory function of the local environment will pave the way for improved stem cell production in vitro and clinical cell therapy.


Assuntos
Aorta/embriologia , Células-Tronco Hematopoéticas/citologia , RNA/análise , Nicho de Células-Tronco/genética , Tomografia , Animais , Animais Geneticamente Modificados , Aorta/citologia , Rastreamento de Células/métodos , Embrião de Galinha , Embrião de Mamíferos , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , RNA/genética , Análise de Sequência de RNA/métodos , Análise de Célula Única , Especificidade da Espécie , Tomografia/métodos , Tomografia/veterinária , Peixe-Zebra/embriologia , Peixe-Zebra/genética
3.
Development ; 144(13): 2352-2363, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28526756

RESUMO

Hematopoietic stem cells (HSCs), which are responsible for blood cell production, are generated during embryonic development. Human and chicken embryos share features that position the chicken as a reliable and accessible alternative model to study developmental hematopoiesis. However, the existence of HSCs has never been formally proven in chicken embryos. Here, we have established a complete cartography and quantification of hematopoietic cells in the aorta during development. We demonstrate the existence of bona fide HSCs, originating from the chicken embryo aorta (and not the yolk sac, allantois or head), through an in vivo transplantation assay. Embryos transplanted in ovo with GFP embryonic tissues on the chorio-allantoic membrane provided multilineage reconstitution in adulthood. Historically, most breakthrough discoveries in the field of developmental hematopoiesis were first made in birds and later extended to mammals. Our study sheds new light on the avian model as a valuable system to study HSC production and regulation in vivo.


Assuntos
Linhagem da Célula , Galinhas/metabolismo , Células-Tronco Hematopoéticas/citologia , Animais , Aorta/citologia , Aorta/embriologia , Linfócitos B/citologia , Proliferação de Células , Sobrevivência Celular , Embrião de Galinha , Membrana Corioalantoide/transplante , Desenvolvimento Embrionário , Proteínas de Fluorescência Verde/metabolismo , Hematopoese , Saco Vitelino/embriologia
4.
Development ; 143(8): 1302-12, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26952980

RESUMO

Adult-type hematopoietic stem and progenitor cells are formed during ontogeny from a specialized subset of endothelium, termed the hemogenic endothelium, via an endothelial-to-hematopoietic transition (EHT) that occurs in the embryonic aorta and the associated arteries. Despite efforts to generate models, little is known about the mechanisms that drive endothelial cells to the hemogenic fate and about the subsequent molecular control of the EHT. Here, we have designed a stromal line-free controlled culture system utilizing the embryonic pre-somitic mesoderm to obtain large numbers of endothelial cells that subsequently commit into hemogenic endothelium before undergoing EHT. Monitoring the culture for up to 12 days using key molecular markers reveals stepwise commitment into the blood-forming system that is reminiscent of the cellular and molecular changes occurring during hematopoietic development at the level of the aorta. Long-term single-cell imaging allows tracking of the EHT of newly formed blood cells from the layer of hemogenic endothelial cells. By modifying the culture conditions, it is also possible to modulate the endothelial cell commitment or the EHT or to produce smooth muscle cells at the expense of endothelial cells, demonstrating the versatility of the cell culture system. This method will improve our understanding of the precise cellular changes associated with hemogenic endothelium commitment and EHT and, by unfolding these earliest steps of the hematopoietic program, will pave the way for future ex vivo production of blood cells.


Assuntos
Técnicas de Cultura de Células , Endotélio Vascular/citologia , Hemangioblastos/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Animais , Adesão Celular , Coturnix , Meios de Cultura , Mesoderma/citologia , Transcriptoma
5.
Development ; 139(2): 277-87, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22129828

RESUMO

We have combined the use of mouse genetic strains and the mouse-into-chicken chimera system to determine precisely the sequence of forelimb colonization by presomitic mesoderm (PSM)-derived myoblasts and angioblasts, and the possible role of this latter cell type in myoblast guidance. By creating a new Flk1/Pax3 double reporter mouse line, we have established the precise timetable for angioblast and myoblast delamination/migration from the somite to the limb bud. This timetable was conserved when mouse PSM was grafted into a chicken host, which further validates the experimental model. The use of Pax3(GFP/GFP) knockout mice showed that establishment of vascular endothelial and smooth muscle cells (SMCs) is not compromised by the absence of Pax3. Of note, Pax3(GFP/GFP) knockout mouse PSM-derived cells can contribute to aortic, but not to limb, SMCs that are derived from the somatopleure. Finally, using the Flk1(lacZ)(/)(lacZ) knockout mouse, we show that, in the absence of angioblast and vascular network formation, myoblasts are prevented from migrating into the limb. Taken together, our study establishes for the first time the time schedule for endothelial and skeletal muscle cell colonization in the mouse limb bud and establishes the absolute requirement of endothelial cells for myoblast delamination and migration to the limb. It also reveals that cells delaminating from the somites display marked differentiation traits, suggesting that if a common progenitor exists, its lifespan is extremely short and restricted to the somite.


Assuntos
Vasos Sanguíneos/citologia , Movimento Celular/fisiologia , Membro Anterior/embriologia , Botões de Extremidades/citologia , Mesoderma/citologia , Mioblastos/fisiologia , Somitos/citologia , Animais , Diferenciação Celular/fisiologia , Embrião de Galinha , Quimera/embriologia , Membro Anterior/citologia , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados , Fatores de Tempo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , beta-Galactosidase
6.
Haematologica ; 97(7): 975-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22271899

RESUMO

CD105 is an auxiliary receptor for the transforming growth factor beta superfamily, highly expressed on proliferating endothelial cells and adult hematopoietic stem cells. Because CD105 mRNA expression was reported in the developing aortic region, we further characterized its expression profile in the aorta and examined the hematopoietic potential of CD105(+) cells. Aortic endothelial cells, intra-aortic hematopoietic cell clusters and the purified cell fraction enriched in progenitor/hematopoietic stem cell activity expressed CD105. Aortic hematopoietic short-term clonogenic progenitors were highly enriched in the CD105(intermediate) population whereas more immature long-term progenitors/hematopoietic stem cells are contained within the CD105(high) population. This places CD105 on the short list of molecules discriminating short-term versus long-term progenitors in the aorta. Furthermore, decreasing transforming growth factor beta signaling increases the number of clonogenic progenitors. This suggests that CD105 expression level defines a hierarchy among aortic hematopoietic cells allowing purification of clonogenic versus more immature hematopoietic progenitors, and that the transforming growth factor beta pathway plays a critical role in this process.


Assuntos
Antígenos CD/genética , Aorta/citologia , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Receptores de Superfície Celular/genética , Animais , Antígenos CD/metabolismo , Aorta/metabolismo , Proliferação de Células , Embrião de Mamíferos , Endoglina , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Gravidez , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Fatores de Tempo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
7.
Cell Rep ; 39(11): 110957, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35705037

RESUMO

Hematopoietic stem cells (HSCs) express a large variety of cell surface receptors that are associated with acquisition of self-renewal and multipotent properties. Correct expression of these receptors depends on a delicate balance between cell surface trafficking, recycling, and degradation and is controlled by the microtubule network and Golgi apparatus, whose roles have hardly been explored during embryonic/fetal hematopoiesis. Here we show that, in the absence of CLASP2, a microtubule-associated protein, the overall production of HSCs is reduced, and the produced HSCs fail to self-renew and maintain their stemness throughout mouse and zebrafish development. This phenotype can be attributed to decreased cell surface expression of the hematopoietic receptor c-Kit, which originates from increased lysosomal degradation in combination with a reduction in trafficking to the plasma membrane. A dysfunctional Golgi apparatus in CLASP2-deficient HSCs seems to be the underlying cause of the c-Kit expression and signaling imbalance.


Assuntos
Células-Tronco Hematopoéticas , Peixe-Zebra , Animais , Camundongos , Hematopoese/genética , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo
8.
Front Immunol ; 12: 790379, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899758

RESUMO

The journey of a hematopoietic stem cell (HSC) involves the passage through successive anatomical sites where HSCs are in direct contact with their surrounding microenvironment, also known as niche. These spatial and temporal cellular interactions throughout development are required for the acquisition of stem cell properties, and for maintaining the HSC pool through balancing self-renewal, quiescence and lineage commitment. Understanding the context and consequences of these interactions will be imperative for our understanding of HSC biology and will lead to the improvement of in vitro production of HSCs for clinical purposes. The aorta-gonad-mesonephros (AGM) region is in this light of particular interest since this is the cradle of HSC emergence during the embryonic development of all vertebrate species. In this review, we will focus on the developmental origin of HSCs and will discuss the novel technological approaches and recent progress made to identify the cellular composition of the HSC supportive niche and the underlying molecular events occurring in the AGM region.


Assuntos
Genômica/tendências , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Análise de Célula Única/tendências , Nicho de Células-Tronco , Animais , Aorta/embriologia , Técnicas de Cultura de Células/tendências , Linhagem da Célula , Células Cultivadas , Difusão de Inovações , Perfilação da Expressão Gênica/tendências , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Humanos , Mesonefro/embriologia , Fenótipo , Proteômica/tendências , Transdução de Sinais , Transcriptoma
9.
Nat Commun ; 12(1): 3851, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34158501

RESUMO

Positional information driving limb muscle patterning is contained in connective tissue fibroblasts but not in myogenic cells. Limb muscles originate from somites, while connective tissues originate from lateral plate mesoderm. With cell and genetic lineage tracing we challenge this model and identify an unexpected contribution of lateral plate-derived fibroblasts to the myogenic lineage, preferentially at the myotendinous junction. Analysis of single-cell RNA-sequencing data from whole limbs at successive developmental stages identifies a population displaying a dual muscle and connective tissue signature. BMP signalling is active in this dual population and at the tendon/muscle interface. In vivo and in vitro gain- and loss-of-function experiments show that BMP signalling regulates a fibroblast-to-myoblast conversion. These results suggest a scenario in which BMP signalling converts a subset of lateral plate mesoderm-derived cells to a myogenic fate in order to create a boundary of fibroblast-derived myonuclei at the myotendinous junction that controls limb muscle patterning.


Assuntos
Padronização Corporal/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/metabolismo , Somitos/metabolismo , Animais , Linhagem da Célula/genética , Células Cultivadas , Embrião de Galinha , Extremidades/embriologia , Fibroblastos/citologia , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Desenvolvimento Muscular/genética , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somitos/citologia , Somitos/embriologia
10.
Nat Cell Biol ; 21(11): 1334-1345, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31685991

RESUMO

It is well established that haematopoietic stem and progenitor cells (HSPCs) are generated from a transient subset of specialized endothelial cells termed haemogenic, present in the yolk sac, placenta and aorta, through an endothelial-to-haematopoietic transition (EHT). HSPC generation via EHT is thought to be restricted to the early stages of development. By using experimental embryology and genetic approaches in birds and mice, respectively, we document here the discovery of a bone marrow haemogenic endothelium in the late fetus/young adult. These cells are capable of de novo producing a cohort of HSPCs in situ that harbour a very specific molecular signature close to that of aortic endothelial cells undergoing EHT or their immediate progenies, i.e., recently emerged HSPCs. Taken together, our results reveal that HSPCs can be generated de novo past embryonic stages. Understanding the molecular events controlling this production will be critical for devising innovative therapies.


Assuntos
Células da Medula Óssea/metabolismo , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Animais , Animais Geneticamente Modificados , Aorta/citologia , Aorta/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Galinhas , Embrião de Mamíferos , Embrião não Mamífero , Feminino , Feto , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Hemangioblastos/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Heterozigoto , Homozigoto , Masculino , Camundongos , Gravidez , Saco Vitelino/citologia , Saco Vitelino/crescimento & desenvolvimento , Saco Vitelino/metabolismo
11.
Cell Rep ; 24(1): 130-141, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29972775

RESUMO

Microglia, the tissue-resident macrophages of the CNS, represent major targets for therapeutic intervention in a wide variety of neurological disorders. Efficient reprogramming protocols to generate microglia-like cells in vitro using patient-derived induced pluripotent stem cells will, however, require a precise understanding of the cellular and molecular events that instruct microglial cell fates. This remains a challenge since the developmental origin of microglia during embryogenesis is controversial. Here, using genetic tracing in zebrafish, we uncover primitive macrophages as the unique source of embryonic microglia. We also demonstrate that this initial population is transient, with primitive microglia later replaced by definitive microglia that persist throughout adulthood. The adult wave originates from cmyb-dependent hematopoietic stem cells. Collectively, our work challenges the prevailing model establishing erythro-myeloid progenitors as the sole and direct microglial precursor and provides further support for the existence of multiple waves of microglia, which originate from distinct hematopoietic precursors.


Assuntos
Embrião não Mamífero/citologia , Macrófagos/citologia , Microglia/metabolismo , Proteínas Proto-Oncogênicas c-myb/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Desenvolvimento Embrionário , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Cinética , Macrófagos/metabolismo
12.
Nat Commun ; 9(1): 2517, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29955049

RESUMO

Haematopoietic stem cells (HSCs) are generated from haemogenic endothelial (HE) cells via the formation of intra-aortic haematopoietic clusters (IAHCs) in vertebrate embryos. The molecular events controlling endothelial specification, endothelial-to-haematopoietic transition (EHT) and IAHC formation, as it occurs in vivo inside the aorta, are still poorly understood. To gain insight in these processes, we performed single-cell RNA-sequencing of non-HE cells, HE cells, cells undergoing EHT, IAHC cells, and whole IAHCs isolated from mouse embryo aortas. Our analysis identified the genes and transcription factor networks activated during the endothelial-to-haematopoietic switch and IAHC cell maturation toward an HSC fate. Our study provides an unprecedented complete resource to study in depth HSC generation in vivo. It will pave the way for improving HSC production in vitro to address the growing need for tailor-made HSCs to treat patients with blood-related disorders.


Assuntos
Aorta/metabolismo , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Transcriptoma , Animais , Aorta/citologia , Aorta/crescimento & desenvolvimento , Diferenciação Celular , Embrião de Mamíferos , Feminino , Ontologia Genética , Redes Reguladoras de Genes , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Anotação de Sequência Molecular , Análise de Célula Única
14.
Exp Hematol ; 42(8): 661-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997246

RESUMO

The chicken embryo has a long history as a key model in developmental biology. Because of its distinctive developmental characteristics, it has contributed to major breakthroughs in the field of hematopoiesis. Among these, the discovery of B lymphocytes and the three rounds of thymus colonization; the embryonic origin of hematopoietic stem cells and the traffic between different hematopoietic organs; and the existence of two distinct endothelial cell lineages one angioblastic, restricted to endothelial cell production, and another, hemangioblastic, able to produce both endothelial and hematopoietic cells, should be cited. The avian model has also contributed to substantiate the endothelial-to-hematopoietic transition associated with aortic hematopoiesis and the existence of the allantois as a hematopoietic organ. Because the immune system develops relatively late in aves, the avian embryo is used to probe the tissue-forming potential of mouse tissues through mouse-into-chicken chimeras, providing insights into early mouse development by circumventing the lethality associated with some genetic strains. Finally, the avian embryo can be used to investigate the differentiation potential of human ES cells in the context of a whole organism. The combinations of classic approaches with the development of powerful genetic tools make the avian embryo a great and versatile model.


Assuntos
Hematopoese , Animais , Aorta/fisiologia , Embrião de Galinha , Endotélio Vascular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Modelos Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA