Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Cell Biol ; 26(5): 719-730, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38594587

RESUMO

During embryonic development, blood cells emerge from specialized endothelial cells, named haemogenic endothelial cells (HECs). As HECs are rare and only transiently found in early developing embryos, it remains difficult to distinguish them from endothelial cells. Here we performed transcriptomic analysis of 28- to 32-day human embryos and observed that the expression of Fc receptor CD32 (FCGR2B) is highly enriched in the endothelial cell population that contains HECs. Functional analyses using human embryonic and human pluripotent stem cell-derived endothelial cells revealed that robust multilineage haematopoietic potential is harboured within CD32+ endothelial cells and showed that 90% of CD32+ endothelial cells are bona fide HECs. Remarkably, these analyses indicated that HECs progress through different states, culminating in FCGR2B expression, at which point cells are irreversibly committed to a haematopoietic fate. These findings provide a precise method for isolating HECs from human embryos and human pluripotent stem cell cultures, thus allowing the efficient generation of haematopoietic cells in vitro.


Assuntos
Desenvolvimento Embrionário , Receptores de IgG , Humanos , Desenvolvimento Embrionário/genética , Receptores de IgG/metabolismo , Receptores de IgG/genética , Hemangioblastos/metabolismo , Hemangioblastos/citologia , Diferenciação Celular , Células Endoteliais/metabolismo , Células Endoteliais/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/citologia , Linhagem da Célula , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Transcriptoma , Perfilação da Expressão Gênica , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/citologia
2.
J Cell Physiol ; 238(1): 179-194, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36436185

RESUMO

Hemogenic endothelial (HE) cells are specialized endothelial cells to give rise to hematopoietic stem/progenitor cells during hematopoietic development. The underlying mechanisms that regulate endothelial-to-hematopoietic transition (EHT) of human HE cells are not fully understand. Here, we identified platelet endothelial aggregation receptor-1 (PEAR1) as a novel regulator of early hematopoietic development in human pluripotent stem cells (hPSCs). We found that the expression of PEAP1 was elevated during hematopoietic development. A subpopulation of PEAR1+ cells overlapped with CD34+ CD144+ CD184+ CD73- arterial-type HE cells. Transcriptome analysis by RNA sequencing indicated that TAL1/SCL, GATA2, MYB, RUNX1 and other key transcription factors for hematopoietic development were mainly expressed in PEAR1+ cells, whereas the genes encoding for niche-related signals, such as fibronectin, vitronectin, bone morphogenetic proteins and jagged1, were highly expressed in PEAR1- cells. The isolated PEAR1+ cells exhibited significantly greater EHT capacity on endothelial niche, compared with the PEAR1- cells. Colony-forming unit (CFU) assays demonstrated the multilineage hematopoietic potential of PEAR1+ -derived hematopoietic cells. Furthermore, PEAR1 knockout in hPSCs by CRISPR/Cas9 technology revealed that the hematopoietic differentiation was impaired, resulting in decreased EHT capacity, decreased expression of hematopoietic-related transcription factors, and increased expression of niche-related signals. In summary, this study revealed a novel role of PEAR1 in balancing intrinsic and extrinsic signals for early hematopoietic fate decision.


Assuntos
Hemangioblastos , Hematopoese , Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes , Receptores de Superfície Celular , Humanos , Diferenciação Celular , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes/citologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição/metabolismo
3.
Blood ; 139(19): 2942-2957, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35245372

RESUMO

The hematopoietic stem cells (HSCs) that produce blood for the lifetime of an animal arise from RUNX1+ hemogenic endothelial cells (HECs) in the embryonic vasculature through a process of endothelial-to-hematopoietic transition (EHT). Studies have identified inflammatory mediators and fluid shear forces as critical environmental stimuli for EHT, raising the question of how such diverse inputs are integrated to drive HEC specification. Endothelial cell MEKK3-KLF2/4 signaling can be activated by both fluid shear forces and inflammatory mediators, and it plays roles in cardiovascular development and disease that have been linked to both stimuli. Here we demonstrate that MEKK3 and KLF2/4 are required in endothelial cells for the specification of RUNX1+ HECs in both the yolk sac and dorsal aorta of the mouse embryo and for their transition to intraaortic hematopoietic cluster (IAHC) cells. The inflammatory mediators lipopolysaccharide and interferon-γ increase RUNX1+ HECs in an MEKK3-dependent manner. Maternal administration of catecholamines that stimulate embryo cardiac function and accelerate yolk sac vascular remodeling increases EHT by wild-type but not MEKK3-deficient endothelium. These findings identify MEKK-KLF2/4 signaling as an essential pathway for EHT and provide a molecular basis for the integration of diverse environmental inputs, such as inflammatory mediators and hemodynamic forces, during definitive hematopoiesis.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core , Hemangioblastos , Hematopoese , Animais , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Endotélio/metabolismo , Hemangioblastos/citologia , Hemangioblastos/metabolismo , Hemodinâmica , Mediadores da Inflamação/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , MAP Quinase Quinase Quinase 3/metabolismo , Camundongos
4.
Development ; 149(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35043940

RESUMO

Hemogenic endothelial (HE) cells in the dorsal aorta undergo an endothelial-to-hematopoietic transition (EHT) to form multipotent progenitors, lympho-myeloid biased progenitors (LMPs), pre-hematopoietic stem cells (pre-HSCs) and adult-repopulating HSCs. These briefly accumulate in intra-arterial hematopoietic clusters (IAHCs) before being released into the circulation. It is generally assumed that the number of IAHC cells correlates with the number of HSCs. Here, we show that changes in the number of IAHC cells, LMPs and HSCs can be uncoupled. Mutations impairing MyD88-dependent toll-like receptor (TLR) signaling decreased the number of IAHC cells and LMPs, but increased the number of HSCs in the aorta-gonad-mesonephros region of mouse embryos. TLR4-deficient embryos generated normal numbers of HE cells, but IAHC cell proliferation decreased. Loss of MyD88-dependent TLR signaling in innate immune myeloid cells had no effect on IAHC cell numbers. Instead, TLR4 deletion in endothelial cells (ECs) recapitulated the phenotype observed with germline deletion, demonstrating that MyD88-dependent TLR signaling in ECs and/or in IAHCs regulates the numbers of LMPs and HSCs.


Assuntos
Embrião de Mamíferos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Embrião de Mamíferos/citologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Hemangioblastos/citologia , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/citologia , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Receptores Toll-Like/metabolismo
5.
Blood ; 139(3): 343-356, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-34517413

RESUMO

In vitro generation and expansion of hematopoietic stem cells (HSCs) holds great promise for the treatment of any ailment that relies on bone marrow or blood transplantation. To achieve this, it is essential to resolve the molecular and cellular pathways that govern HSC formation in the embryo. HSCs first emerge in the aorta-gonad-mesonephros (AGM) region, where a rare subset of endothelial cells, hemogenic endothelium (HE), undergoes an endothelial-to-hematopoietic transition (EHT). Here, we present full-length single-cell RNA sequencing (scRNA-seq) of the EHT process with a focus on HE and dorsal aorta niche cells. By using Runx1b and Gfi1/1b transgenic reporter mouse models to isolate HE, we uncovered that the pre-HE to HE continuum is specifically marked by angiotensin-I converting enzyme (ACE) expression. We established that HE cells begin to enter the cell cycle near the time of EHT initiation when their morphology still resembles endothelial cells. We further demonstrated that RUNX1 AGM niche cells consist of vascular smooth muscle cells and PDGFRa+ mesenchymal cells and can functionally support hematopoiesis. Overall, our study provides new insights into HE differentiation toward HSC and the role of AGM RUNX1+ niche cells in this process. Our expansive scRNA-seq datasets represents a powerful resource to investigate these processes further.


Assuntos
Embrião de Mamíferos/embriologia , Hemangioblastos/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Animais , Diferenciação Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Mesonefro/citologia , Mesonefro/embriologia , Mesonefro/metabolismo , Camundongos , Análise de Célula Única , Transcriptoma , Peixe-Zebra
6.
STAR Protoc ; 2(4): 100884, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34693362

RESUMO

The murine mid-gestational placenta has been identified as a hematopoietic site during embryonic development. Here, we describe a protocol for isolation and characterization of the hemogenic endothelial (HE) cells from mouse placenta. We also describe techniques for dissection of placental tissues and for the optimization of tissue digestion and antibody conjugation conditions to identify HE cells via fluorescence-activated cell sorting. For details on the usage and application of this protocol, please refer to Liang et al. (2021).


Assuntos
Citometria de Fluxo/métodos , Hemangioblastos/citologia , Placenta/citologia , Animais , Técnicas de Cultura de Células , Feminino , Camundongos , Gravidez
7.
Sci Rep ; 11(1): 17589, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34475502

RESUMO

During hematopoietic development, definitive hematopoietic cells are derived from hemogenic endothelial (HE) cells through a process known as endothelial to hematopoietic transition (EHT). During EHT, transitioning cells proliferate and undergo progressive changes in gene expression culminating in the new cell identity with corresponding changes in function, phenotype and morphology. However, the metabolic pathways fueling this transition remain unclear. We show here that glutamine is a crucial regulator of EHT and a rate limiting metabolite in the hematopoietic differentiation of HE cells. Intriguingly, different hematopoietic lineages require distinct derivatives of glutamine. While both derivatives, α-ketoglutarate and nucleotides, are required for early erythroid differentiation of HE during glutamine deprivation, lymphoid differentiation relies on α-ketoglutarate alone. Furthermore, treatment of HE cells with α-ketoglutarate in glutamine-free conditions pushes their differentiation towards lymphoid lineages both in vitro and in vivo, following transplantation into NSG mice. Thus, we report an essential role for glutamine metabolism during EHT, regulating both the emergence and the specification of hematopoietic cells through its various derivatives.


Assuntos
Glutamina/metabolismo , Hemangioblastos/citologia , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Hematopoese , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID
8.
Cell Rep ; 36(11): 109675, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525376

RESUMO

During embryogenesis, waves of hematopoietic progenitors develop from hemogenic endothelium (HE) prior to the emergence of self-renewing hematopoietic stem cells (HSCs). Although previous studies have shown that yolk-sac-derived erythromyeloid progenitors and HSCs emerge from distinct populations of HE, it remains unknown whether the earliest lymphoid-competent progenitors, multipotent progenitors, and HSCs originate from common HE. In this study, we demonstrate by clonal assays and single-cell transcriptomics that rare HE with functional HSC potential in the early murine embryo are distinct from more abundant HE with multilineage hematopoietic potential that fail to generate HSCs. Specifically, HSC-competent HE are characterized by expression of CXCR4 surface marker and by higher expression of genes tied to arterial programs regulating HSC dormancy and self-renewal. Taken together, these findings suggest a revised model of developmental hematopoiesis in which the initial populations of multipotent progenitors and HSCs arise independently from HE with distinct phenotypic and transcriptional properties.


Assuntos
Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Multipotentes/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular/genética , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Feminino , Hemangioblastos/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/citologia , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transcrição Gênica
9.
Dev Cell ; 56(14): 2121-2133.e6, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34197725

RESUMO

Macrophages play pivotal roles in immunity, hematopoiesis, and tissue homeostasis. In mammals, macrophages have been shown to originate from yolk-sac-derived erythro-myeloid progenitors and aorta-gonad-mesonephros (AGM)-derived hematopoietic stem cells. However, whether macrophages can arise from other embryonic sites remains unclear. Here, using single-cell RNA sequencing, we profile the transcriptional landscape of mouse fetal placental hematopoiesis. We uncover and experimentally validate that a CD44+ subpopulation of placental endothelial cells (ECs) exhibits hemogenic potential. Importantly, lineage tracing using the newly generated Hoxa13 reporter line shows that Hoxa13-labeled ECs can produce placental macrophages, named Hofbauer cell (HBC)-like cells. Furthermore, we identify two subtypes of HBC-like cells, and cell-cell interaction analysis identifies their potential roles in angiogenesis and antigen presentation, separately. Our study provides a comprehensive understanding of placental hematopoiesis and highlights the placenta as a source of macrophages, which has important implications for both basic and translational research.


Assuntos
Linhagem da Célula , Hemangioblastos/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Placenta/citologia , Animais , Feminino , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Gravidez , Análise de Célula Única , Transcriptoma
10.
Cells Dev ; 166: 203685, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33994358

RESUMO

Embryonic stem cells (ESCs) have been shown to have an ability to form a large number of functional endothelial cells in vitro, but generating organ-specific endothelial cells remains a challenge. Sonic hedgehog (SHH) pathway is one of the crucial developmental pathways that control differentiation of many embryonic cell types such as neuroectodermal, primitive gut tube and developing limb buds; SHH pathway is important for functioning of adult cell of skin, bone, liver as well as it regulates haematopoiesis. Misregulation of SHH pathway leads to cancers such as hepatic, pancreatic, basal cell carcinoma, medulloblastoma, etc. However, its role in differentiation of human ESCs into endothelial cells has not been completely elucidated. Here, we examined the role of SHH signalling pathway in endothelial differentiation of hESCs by growing them in the presence of an SHH agonist (purmorphamine) and an SHH antagonist (SANT-1) for a period of 6 days. Interestingly, we found that activation of SHH pathway led to a higher expression of set of transcription factors such as BRACHYURY, GATA2 and RUNX1, thus favouring hemogenic endothelium; whereas inhibition of SHH pathway led to a reduced expression of set of markers such as RUNX1 and BRACHURY, and an increased expression of set of markers - NFATC1, c-KIT, GATA4, CD31 & CD34, thus favouring endocardiogenic endothelium. The results of this study have revealed the previously unreported deterministic role of SHH pathway in specification of endothelial cells differentiated from human ESCs into hemogenic vs. endocardiogenic lineage; this finding could have major implications for clinical applications.


Assuntos
Padronização Corporal , Diferenciação Celular , Proteínas Hedgehog/metabolismo , Hemangioblastos/citologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Transdução de Sinais , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Padronização Corporal/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/antagonistas & inibidores , Humanos , Mesoderma/metabolismo , Modelos Biológicos , Transdução de Sinais/genética
11.
Biochem Biophys Res Commun ; 558: 161-167, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33930817

RESUMO

Current understanding of hematopoietic stem cell (HSC) development comes from mouse models is considered to be evolutionarily conserved in human. However, the cross-species comparison of the transcriptomic profiles of developmental HSCs at single-cell level is still lacking. Here, we performed integrative transcriptomic analysis of a series of key cell populations during HSC development in human and mouse, including HSC-primed hemogenic endothelial cells and pre-HSCs in mid-gestational aorta-gonad-mesonephros (AGM) region, and mature HSCs in fetal liver and adult bone marrow. We demonstrated the general similarity of transcriptomic characteristics between corresponding cell populations of the two species. Of note, one of the previously transcriptomically defined hematopoietic stem progenitor cell (HSPC) populations with certain arterial characteristics in AGM region of human embryos showed close transcriptomic similarity to pre-HSCs in mouse embryos. On the other hand, the other two HSPC populations in human AGM region displayed molecular similarity with fetal liver HSPCs, suggesting the maturation in AGM before HSCs colonizing the fetal liver in human, which was different to that in mouse. Finally, we re-clustered cells based on the integrated dataset and illustrated the evolutionarily conserved molecular signatures of major cell populations. Our results revealed transcriptomic conservation of critical cell populations and molecular characteristics during HSC development between human and mouse, providing a resource and theoretic basis for future studies on mammalian HSC development and regeneration by using mouse models.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Transcriptoma , Animais , Diferenciação Celular/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos/citologia , Hemangioblastos/metabolismo , Hematopoese/genética , Humanos , Mesonefro/citologia , Mesonefro/metabolismo , Camundongos , Família Multigênica , Análise de Célula Única/métodos , Especificidade da Espécie
12.
Nat Cell Biol ; 23(1): 61-74, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33420489

RESUMO

Extra-embryonic mesoderm (ExM)-composed of the earliest cells that traverse the primitive streak-gives rise to the endothelium as well as haematopoietic progenitors in the developing yolk sac. How a specific subset of ExM becomes committed to a haematopoietic fate remains unclear. Here we demonstrate using an embryonic stem cell model that transient expression of the T-box transcription factor Eomesodermin (Eomes) governs haemogenic competency of ExM. Eomes regulates the accessibility of enhancers that the transcription factor stem cell leukaemia (SCL) normally utilizes to specify primitive erythrocytes and is essential for the normal development of Runx1+ haemogenic endothelium. Single-cell RNA sequencing suggests that Eomes loss of function profoundly blocks the formation of blood progenitors but not specification of Flk-1+ haematoendothelial progenitors. Our findings place Eomes at the top of the transcriptional hierarchy regulating early blood formation and suggest that haemogenic competence is endowed earlier during embryonic development than was previously appreciated.


Assuntos
Células-Tronco Embrionárias/citologia , Hemangioblastos/citologia , Mesoderma/citologia , Proteínas com Domínio T/fisiologia , Saco Vitelino/citologia , Animais , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Hemangioblastos/metabolismo , Masculino , Mesoderma/metabolismo , Camundongos Knockout , Gravidez , RNA-Seq , Análise de Célula Única , Proteína 1 de Leucemia Linfocítica Aguda de Células T/genética , Proteína 1 de Leucemia Linfocítica Aguda de Células T/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Saco Vitelino/metabolismo
13.
Exp Hematol ; 95: 46-57.e8, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33395577

RESUMO

We report that a sheep fetal liver provides a microenvironment for generating hematopoietic cells with long-term engrafting capacity and multilineage differentiation potential from human induced pluripotent stem cell (iPSC)-derived hemogenic endothelial cells (HEs). Despite the promise of iPSCs for making any cell types, generating hematopoietic stem and progenitor cells (HSPCs) is still a challenge. We hypothesized that the hematopoietic microenvironment, which exists in fetal liver but is lacking in vitro, turns iPSC-HEs into HSPCs. To test this, we transplanted CD45-negative iPSC-HEs into fetal sheep liver, in which HSPCs first grow. Within 2 months, the transplanted cells became CD45 positive and differentiated into multilineage blood cells in the fetal liver. Then, CD45-positive cells translocated to the bone marrow and were maintained there for 3 years with the capability of multilineage differentiation, indicating that hematopoietic cells with long-term engraftment potential were generated. Moreover, human hematopoietic cells were temporally enriched by xenogeneic donor-lymphocyte infusion into the sheep. This study could serve as a foundation to generate HSPCs from iPSCs.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Ovinos/embriologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Microambiente Celular , Ensaio de Unidades Formadoras de Colônias , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Feminino , Técnicas Genéticas , Sobrevivência de Enxerto , Hemangioblastos/citologia , Xenoenxertos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Antígenos Comuns de Leucócito/análise , Fígado/embriologia , Subpopulações de Linfócitos , Gravidez , Especificidade da Espécie
14.
Cell Rep ; 32(6): 108004, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32783932

RESUMO

During embryogenesis, lymphoid tissue inducer (LTi) cells are essential for lymph node organogenesis. These cells are part of the innate lymphoid cell (ILC) family. Although their earliest embryonic hematopoietic origin is unclear, other innate immune cells have been shown to be derived from early hemogenic endothelium in the yolk sac as well as the aorta-gonad-mesonephros. A proper model to discriminate between these locations was unavailable. In this study, using a Cxcr4-CreERT2 lineage tracing model, we identify a major contribution from embryonic hemogenic endothelium, but not the yolk sac, toward LTi progenitors. Conversely, embryonic LTi cells are replaced by hematopoietic stem cell-derived cells in adults. We further show that, in the fetal liver, common lymphoid progenitors differentiate into highly dynamic alpha-lymphoid precursor cells that, at this embryonic stage, preferentially mature into LTi precursors and establish their functional LTi cell identity only after reaching the periphery.


Assuntos
Hemangioblastos/metabolismo , Hematopoese/fisiologia , Tecido Linfoide/embriologia , Receptores CXCR4/metabolismo , Animais , Desenvolvimento Embrionário/fisiologia , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunidade Inata , Fígado/embriologia , Linfócitos/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Saco Vitelino/embriologia
15.
Blood ; 136(7): 845-856, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32392346

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) in the bone marrow are derived from a small population of hemogenic endothelial (HE) cells located in the major arteries of the mammalian embryo. HE cells undergo an endothelial to hematopoietic cell transition, giving rise to HSPCs that accumulate in intra-arterial clusters (IAC) before colonizing the fetal liver. To examine the cell and molecular transitions between endothelial (E), HE, and IAC cells, and the heterogeneity of HSPCs within IACs, we profiled ∼40 000 cells from the caudal arteries (dorsal aorta, umbilical, vitelline) of 9.5 days post coitus (dpc) to 11.5 dpc mouse embryos by single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin sequencing. We identified a continuous developmental trajectory from E to HE to IAC cells, with identifiable intermediate stages. The intermediate stage most proximal to HE, which we term pre-HE, is characterized by increased accessibility of chromatin enriched for SOX, FOX, GATA, and SMAD motifs. A developmental bottleneck separates pre-HE from HE, with RUNX1 dosage regulating the efficiency of the pre-HE to HE transition. A distal candidate Runx1 enhancer exhibits high chromatin accessibility specifically in pre-HE cells at the bottleneck, but loses accessibility thereafter. Distinct developmental trajectories within IAC cells result in 2 populations of CD45+ HSPCs; an initial wave of lymphomyeloid-biased progenitors, followed by precursors of hematopoietic stem cells (pre-HSCs). This multiomics single-cell atlas significantly expands our understanding of pre-HSC ontogeny.


Assuntos
Diferenciação Celular , Endotélio/embriologia , Hemangioblastos/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Diferenciação Celular/genética , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Embrião de Mamíferos , Endotélio/citologia , Endotélio/metabolismo , Feminino , Dosagem de Genes/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos/citologia , Hematopoese/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , RNA-Seq/métodos
16.
Genesis ; 58(6): e23363, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32302038

RESUMO

Hox genes encode transcription factors that have been implicated in embryonic, adult and disease processes. The earliest developmental program known to be directed by Hox genes is the timing of ingression of presumptive axial mesoderm during gastrulation. We previously used morpholino (MO)-based knockdown to implicate the zebrafish hoxd4a gene in the specification of the hemangioblast, an event occurring at pre-gastrulation stages, well before the earliest known Hox gene function. The precise time at which hoxd4a function is required for this specification is not defined. We therefore fused the hoxd4a coding region to the human estrogen receptor (hERT2 ). Following co-injection of anti-hoxd4a MO with mRNA encoding the Hoxd4a-ERT2 fusion protein, hemangioblast specification was fully rescued when embryos were exposed to the estrogen analog 4-hydroxy-tamoxifen (4-OHT) at 4 hr post-fertilization (hpf), but only poorly at 6 hpf and not at all at 8 hpf, thereby defining a pre-gastrulation role for Hoxd4a, the earliest developmental function of a vertebrate Hox gene so far described. Both DNA binding and interaction with cofactor Pbx were further shown to be required for rescue of the morphant phenotype. Confirmation of the morphant phenotype was sought via the generation of hoxd4a null mutants using CRISPR/Cas9 technology. Null mutants of hoxd4a up to the third generation (F3 ) failed to recapitulate the morphant phenotype, and were largely refractory to the effects of injected anti-hoxd4a MO suggesting the action of genetic compensation.


Assuntos
Diferenciação Celular , Gastrulação , Genes Homeobox , Hemangioblastos/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos/citologia , Humanos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Peixe-Zebra
17.
Cell Res ; 30(5): 376-392, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32203131

RESUMO

Hematopoietic stem cells (HSCs) in adults are believed to be born from hemogenic endothelial cells (HECs) in mid-gestational embryos. Due to the rare and transient nature, the HSC-competent HECs have never been stringently identified and accurately captured, let alone their genuine vascular precursors. Here, we first used high-precision single-cell transcriptomics to unbiasedly examine the relevant EC populations at continuous developmental stages with intervals of 0.5 days from embryonic day (E) 9.5 to E11.0. As a consequence, we transcriptomically identified two molecularly different arterial EC populations and putative HSC-primed HECs, whose number peaked at E10.0 and sharply decreased thereafter, in the dorsal aorta of the aorta-gonad-mesonephros (AGM) region. Combining computational prediction and in vivo functional validation, we precisely captured HSC-competent HECs by the newly constructed Neurl3-EGFP reporter mouse model, and realized the enrichment further by a combination of surface markers (Procr+Kit+CD44+, PK44). Surprisingly, the endothelial-hematopoietic dual potential was rarely but reliably witnessed in the cultures of single HECs. Noteworthy, primitive vascular ECs from E8.0 experienced two-step fate choices to become HSC-primed HECs, namely an initial arterial fate choice followed by a hemogenic fate conversion. This finding resolves several previously observed contradictions. Taken together, comprehensive understanding of endothelial evolutions and molecular programs underlying HSC-primed HEC specification in vivo will facilitate future investigations directing HSC production in vitro.


Assuntos
Aorta/embriologia , Hemangioblastos/citologia , Hematopoese , Transcriptoma , Animais , Células Cultivadas , Embrião de Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Análise de Célula Única
18.
Nat Cell Biol ; 22(1): 60-73, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31907413

RESUMO

Defining the ontogeny of the human adaptive immune system during embryogenesis has implications for understanding childhood diseases including leukaemias and autoimmune conditions. Using RAG1:GFP human pluripotent stem cell reporter lines, we examined human T-cell genesis from pluripotent-stem-cell-derived haematopoietic organoids. Under conditions favouring T-cell development, RAG1+ cells progressively upregulated a cohort of recognized T-cell-associated genes, arresting development at the CD4+CD8+ stage. Sort and re-culture experiments showed that early RAG1+ cells also possessed B-cell, myeloid and erythroid potential. Flow cytometry and single-cell-RNA-sequencing data showed that early RAG1+ cells co-expressed the endothelial/haematopoietic progenitor markers CD34, VECAD and CD90, whereas imaging studies identified RAG1+ cells within CD31+ endothelial structures that co-expressed SOX17+ or the endothelial marker CAV1. Collectively, these observations provide evidence for a wave of human T-cell development that originates directly from haemogenic endothelium via a RAG1+ intermediate with multilineage potential.


Assuntos
Endotélio/citologia , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Linhagem Celular , Desenvolvimento Embrionário/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Organoides/citologia
19.
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
20.
FEBS Lett ; 593(23): 3266-3287, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31557312

RESUMO

Transcription factor (TF)-based reprogramming of somatic tissues holds great promise for regenerative medicine. Previously, we demonstrated that the TFs GATA2, GFI1B, and FOS convert mouse and human fibroblasts to hemogenic endothelial-like precursors that generate hematopoietic stem progenitor (HSPC)-like cells over time. This conversion is lacking in robustness both in yield and biological function. Herein, we show that inclusion of GFI1 to the reprogramming cocktail significantly expands the HSPC-like population. AFT024 coculture imparts functional potential to these cells and allows quantification of stem cell frequency. Altogether, we demonstrate an improved human hemogenic induction protocol that could provide a valuable human in vitro model of hematopoiesis for disease modeling and a platform for cell-based therapeutics. DATABASE: Gene expression data are available in the Gene Expression Omnibus (GEO) database under the accession number GSE130361.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular/genética , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Animais , Técnicas de Cocultura/métodos , Fibroblastos/citologia , Fibroblastos/metabolismo , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hemangioblastos/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...