Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 32: 633-648, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27712101

RESUMEN

Biomechanical forces are emerging as critical regulators of embryogenesis, particularly in the developing cardiovascular system. From the onset of blood flow, the embryonic vasculature is continuously exposed to a variety of hemodynamic forces. These biomechanical stimuli are key determinants of vascular cell specification and remodeling and the establishment of vascular homeostasis. In recent years, major advances have been made in our understanding of mechano-activated signaling networks that control both spatiotemporal and structural aspects of vascular development. It has become apparent that a major site for mechanotransduction is situated at the interface of blood and the vessel wall and that this process is controlled by the vascular endothelium. In this review, we discuss the hemodynamic control of endothelial cell fates, focusing on arterial-venous specification, lymphatic development, and the endothelial-to-hematopoietic transition, and present some recent insights into the mechano-activated pathways driving these cell fate decisions in the developing embryo.


Asunto(s)
Linaje de la Célula , Desarrollo Embrionario , Células Endoteliales/citología , Hemodinámica , Animales , Humanos , Mecanotransducción Celular , Reología
2.
Genes Dev ; 35(21-22): 1398-1400, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725127

RESUMEN

Definitive long-term hematopoietic stem cells (LT-HSCs) arise during embryogenesis in a process termed endothelial-to-hematopoietic transition (EHT), in which specialized hemogenic endothelial cells (HECs) transform into hematopoietic cells. The transcription factor RUNX1 marks HECs and is essential for EHT. Ectopic RUNX1 expression in non-HECs is sufficient to convert them into HECs. However, the conversion efficiency depends on the developmental timing of expression. In this issue of Genes & Development, Howell and colleagues (pp. 1475-1489) leverage this observation to further understand how RUNX1 mediates EHT. They engineered mice that ectopically express RUNX1 in endothelial cells at different developmental time points and doses. They then performed chromatin accessibility and other analyses and correlate this with hemogenic potential. They found that RUNX1 collaborates with TGFß signaling transcription factors to drive chromatin accessibility changes that specify HECs. They also highlight interesting parallels between EHT and endothelial-to-mesenchymal transition (EndoMT), which occurs during cardiac development. The results of Howell and colleagues provide new mechanistic insights into EHT and take us one step closer to generating patient-specific LT-HSCs from induced pluripotent stem cells.


Asunto(s)
Hemangioblastos , Hematopoyesis , Animales , Adhesión Celular , Diferenciación Celular/genética , Hemangioblastos/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones
3.
Genes Dev ; 35(21-22): 1475-1489, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34675061

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are generated de novo in the embryo from hemogenic endothelial cells (HECs) via an endothelial-to-hematopoietic transition (EHT) that requires the transcription factor RUNX1. Ectopic expression of RUNX1 alone can efficiently promote EHT and HSPC formation from embryonic endothelial cells (ECs), but less efficiently from fetal or adult ECs. Efficiency correlated with baseline accessibility of TGFß-related genes associated with endothelial-to-mesenchymal transition (EndoMT) and participation of AP-1 and SMAD2/3 to initiate further chromatin remodeling along with RUNX1 at these sites. Activation of TGFß signaling improved the efficiency with which RUNX1 specified fetal ECs as HECs. Thus, the ability of RUNX1 to promote EHT depends on its ability to recruit the TGFß signaling effectors AP-1 and SMAD2/3, which in turn is determined by the changing chromatin landscape in embryonic versus fetal ECs. This work provides insight into regulation of EndoMT and EHT that will guide reprogramming efforts for clinical applications.


Asunto(s)
Hemangioblastos , Diferenciación Celular/genética , Cromatina/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Feto , Hemangioblastos/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
4.
EMBO J ; 43(9): 1722-1739, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38580775

RESUMEN

Understanding the regulatory mechanisms facilitating hematopoietic stem cell (HSC) specification during embryogenesis is important for the generation of HSCs in vitro. Megakaryocyte emerged from the yolk sac and produce platelets, which are involved in multiple biological processes, such as preventing hemorrhage. However, whether megakaryocytes regulate HSC development in the embryonic aorta-gonad-mesonephros (AGM) region is unclear. Here, we use platelet factor 4 (PF4)-Cre;Rosa-tdTomato+ cells to report presence of megakaryocytes in the HSC developmental niche. Further, we use the PF4-Cre;Rosa-DTA (DTA) depletion model to reveal that megakaryocytes control HSC specification in the mouse embryos. Megakaryocyte deficiency blocks the generation and maturation of pre-HSCs and alters HSC activity at the AGM. Furthermore, megakaryocytes promote endothelial-to-hematopoietic transition in a OP9-DL1 coculture system. Single-cell RNA-sequencing identifies megakaryocytes positive for the cell surface marker CD226 as the subpopulation with highest potential in promoting the hemogenic fate of endothelial cells by secreting TNFSF14. In line, TNFSF14 treatment rescues hematopoietic cell function in megakaryocyte-depleted cocultures. Taken together, megakaryocytes promote production and maturation of pre-HSCs, acting as a critical microenvironmental control factor during embryonic hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas , Megacariocitos , Animales , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular , Hematopoyesis/fisiología , Mesonefro/embriología , Mesonefro/metabolismo , Mesonefro/citología , Células Endoteliales/metabolismo , Células Endoteliales/citología , Técnicas de Cocultivo
5.
Development ; 150(11)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37272531

RESUMEN

Endothelial-to-hematopoietic transition (EHT) is crucial for hematopoietic stem cell (HSC) generation. During EHT, the morphology of hemogenic endothelial cells (HECs) changes from flat and adherent to spherical hematopoietic cells, which detach from the dorsal aorta. HECs attain a rounded shape in a mitosis-independent manner before cell adhesion termination, suggesting an atypical cell-rounding mechanism. However, the direct mechanisms underlying this change in cell morphology during EHT remain unclear. Here, we show that large vacuoles were transiently formed in avian HECs, and that aquaporin 1 (AQP1) was localized in the vacuole and plasma membranes. Overexpression of AQP1 in non-HECs induced ectopic vacuole expansion, cell rounding and subsequent cell detachment from the endothelium into the bloodstream, mimicking EHT. Loss of redundant AQP functions by CRISPR/Cas9 gene editing in HECs impeded the morphological EHT. Our findings provide the first evidence to indicate that morphological segregation of hematopoietic cells from endothelial cells is regulated by water influx into vacuoles. These findings provide important insights for further exploration of the mechanisms underlying cell/tissue morphogenesis through water-adoptive cellular responses.


Asunto(s)
Acuaporinas , Hemangioblastos , Vacuolas , Adhesión Celular , Diferenciación Celular/genética , Morfogénesis , Acuaporinas/metabolismo , Hematopoyesis/genética
6.
BMC Biol ; 22(1): 143, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937802

RESUMEN

BACKGROUND: The endothelial-to-hematopoietic transition (EHT) process during definitive hematopoiesis is highly conserved in vertebrates. Stage-specific expression of transposable elements (TEs) has been detected during zebrafish EHT and may promote hematopoietic stem cell (HSC) formation by activating inflammatory signaling. However, little is known about how TEs contribute to the EHT process in human and mouse. RESULTS: We reconstructed the single-cell EHT trajectories of human and mouse and resolved the dynamic expression patterns of TEs during EHT. Most TEs presented a transient co-upregulation pattern along the conserved EHT trajectories, coinciding with the temporal relaxation of epigenetic silencing systems. TE products can be sensed by multiple pattern recognition receptors, triggering inflammatory signaling to facilitate HSC emergence. Interestingly, we observed that hypoxia-related signals were enriched in cells with higher TE expression. Furthermore, we constructed the hematopoietic cis-regulatory network of accessible TEs and identified potential TE-derived enhancers that may boost the expression of specific EHT marker genes. CONCLUSIONS: Our study provides a systematic vision of how TEs are dynamically controlled to promote the hematopoietic fate decisions through transcriptional and cis-regulatory networks, and pre-train the immunity of nascent HSCs.


Asunto(s)
Elementos Transponibles de ADN , Hematopoyesis , Células Madre Hematopoyéticas , Análisis de la Célula Individual , Animales , Elementos Transponibles de ADN/genética , Análisis de la Célula Individual/métodos , Ratones , Hematopoyesis/genética , Humanos , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales/metabolismo
7.
Annu Rev Physiol ; 83: 17-37, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33035429

RESUMEN

Embryonic definitive hematopoiesis generates hematopoietic stem and progenitor cells (HSPCs) essential for establishment and maintenance of the adult blood system. This process requires the specification of a subset of vascular endothelial cells to become blood-forming, or hemogenic, and the subsequent endothelial-to-hematopoietic transition to generate HSPCs therefrom. The mechanisms that regulate these processes are under intensive investigation, as their recapitulation in vitro from human pluripotent stem cells has the potential to generate autologous HSPCs for clinical applications. In this review, we provide an overview of hemogenic endothelial cell development and highlight the molecular events that govern hemogenic specification of vascular endothelial cells and the generation of multilineage HSPCs from hemogenic endothelium. We also discuss the impact of hemogenic endothelial cell development on adult hematopoiesis.


Asunto(s)
Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Diferenciación Celular/fisiología , Hemangioblastos/fisiología , Hematopoyesis/fisiología , Humanos
8.
Dev Biol ; 501: 92-103, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37353106

RESUMEN

During embryonic development, primitive and definitive waves of hematopoiesis take place to provide proper blood cells for each developmental stage, with the possible involvement of epigenetic factors. We previously found that lysine-specific demethylase 1 (LSD1/KDM1A) promotes primitive hematopoietic differentiation by shutting down the gene expression program of hemangioblasts in an Etv2/Etsrp-dependent manner. In the present study, we demonstrated that zebrafish LSD1 also plays important roles in definitive hematopoiesis in the development of hematopoietic stem and progenitor cells. A combination of genetic approaches and imaging analyses allowed us to show that LSD1 promotes the egress of hematopoietic stem and progenitor cells into the bloodstream during the endothelial-to-hematopoietic transition. Analysis of compound mutant lines with Etv2/Etsrp mutant zebrafish revealed that, unlike in primitive hematopoiesis, this function of LSD1 was independent of Etv2/Etsrp. The phenotype of LSD1 mutant zebrafish during the endothelial-to-hematopoietic transition was similar to that of previously reported compound knockout mice of Gfi1/Gfi1b, which forms a complex with LSD1 and represses endothelial genes. Moreover, co-knockdown of zebrafish Gfi1/Gfi1b genes inhibited the development of hematopoietic stem and progenitor cells. We therefore hypothesize that the shutdown of the Gfi1/Gfi1b-target genes during the endothelial-to-hematopoietic transition is one of the key evolutionarily conserved functions of LSD1 in definitive hematopoiesis.


Asunto(s)
Células Madre , Pez Cebra , Animales , Ratones , Diferenciación Celular , Hematopoyesis/genética , Histona Demetilasas/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
EMBO Rep ; 23(2): e54384, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34914165

RESUMEN

During embryonic development, hematopoiesis occurs through primitive and definitive waves, giving rise to distinct blood lineages. Hematopoietic stem cells (HSCs) emerge from hemogenic endothelial (HE) cells, through endothelial-to-hematopoietic transition (EHT). In the adult, HSC quiescence, maintenance, and differentiation are closely linked to changes in metabolism. However, metabolic processes underlying the emergence of HSCs from HE cells remain unclear. Here, we show that the emergence of blood is regulated by multiple metabolic pathways that induce or modulate the differentiation toward specific hematopoietic lineages during human EHT. In both in vitro and in vivo settings, steering pyruvate use toward glycolysis or OXPHOS differentially skews the hematopoietic output of HE cells toward either an erythroid fate with primitive phenotype, or a definitive lymphoid fate, respectively. We demonstrate that glycolysis-mediated differentiation of HE toward primitive erythroid hematopoiesis is dependent on the epigenetic regulator LSD1. In contrast, OXPHOS-mediated differentiation of HE toward definitive hematopoiesis is dependent on cholesterol metabolism. Our findings reveal that during EHT, metabolism is a major regulator of primitive versus definitive hematopoietic differentiation.


Asunto(s)
Hemangioblastos , Diferenciación Celular , Linaje de la Célula/genética , Femenino , Hemangioblastos/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Embarazo , Piruvatos/metabolismo
10.
J Cell Physiol ; 238(1): 179-194, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36436185

RESUMEN

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.


Asunto(s)
Hemangioblastos , Hematopoyesis , Células Madre Hematopoyéticas , Células Madre Pluripotentes , Receptores de Superficie Celular , Humanos , Diferenciación Celular , Hemangioblastos/citología , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes/citología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Transcripción/metabolismo
11.
Cell Mol Life Sci ; 78(9): 4143-4160, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33559689

RESUMEN

In vitro generation of hematopoietic cells and especially hematopoietic stem cells (HSCs) from human pluripotent stem cells (PSCs) are subject to intensive research in recent decades, as these cells hold great potential for regenerative medicine and autologous cell replacement therapies. Despite many attempts, in vitro, de novo generation of bona fide HSCs remains challenging, and we are still far away from their clinical use, due to insufficient functionality and quantity of the produced HSCs. The challenges of generating PSC-derived HSCs are already apparent in early stages of hemato-endothelial specification with the limitation of recapitulating complex, dynamic processes of embryonic hematopoietic ontogeny in vitro. Further, these current shortcomings imply the incompleteness of our understanding of human ontogenetic processes from embryonic mesoderm over an intermediate, specialized hemogenic endothelium (HE) to their immediate progeny, the HSCs. In this review, we examine the recent investigations of hemato-endothelial ontogeny and recently reported progress for the conversion of PSCs and other promising somatic cell types towards HSCs with the focus on the crucial and inevitable role of the HE to achieve the long-standing goal-to generate therapeutically applicable PSC-derived HSCs in vitro.


Asunto(s)
Endotelio/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes/citología , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Endotelio/citología , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo
12.
EMBO J ; 35(21): 2315-2331, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27638855

RESUMEN

During development, hematopoietic stem cells (HSCs) emerge from aortic endothelial cells (ECs) through an intermediate stage called hemogenic endothelium by a process known as endothelial-to-hematopoietic transition (EHT). While Notch signaling, including its upstream regulator Vegf, is known to regulate this process, the precise molecular control and temporal specificity of Notch activity remain unclear. Here, we identify the zebrafish transcriptional regulator evi1 as critically required for Notch-mediated EHT In vivo live imaging studies indicate that evi1 suppression impairs EC progression to hematopoietic fate and therefore HSC emergence. evi1 is expressed in ECs and induces these effects cell autonomously by activating Notch via pAKT Global or endothelial-specific induction of notch, vegf, or pAKT can restore endothelial Notch and HSC formations in evi1 morphants. Significantly, evi1 overexpression induces Notch independently of Vegf and rescues HSC numbers in embryos treated with a Vegf inhibitor. In sum, our results unravel evi1-pAKT as a novel molecular pathway that, in conjunction with the shh-vegf axis, is essential for activation of Notch signaling in VDA endothelial cells and their subsequent conversion to HSCs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proto-Oncogenes/fisiología , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Aorta/metabolismo , Proteínas de Unión al ADN/genética , Diaminas/farmacología , Embrión no Mamífero , Células Endoteliales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proto-Oncogenes/genética , Receptores Notch/metabolismo , Tiazoles/farmacología , Factores de Transcripción/genética , Pez Cebra , Proteínas de Pez Cebra/genética
13.
Dev Biol ; 434(2): 292-303, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29253505

RESUMEN

The embryonic aorta produces hematopoietic stem and progenitor cells from a hemogenic endothelium localized in the aortic floor through an endothelial to hematopoietic transition. It has been long proposed that the Bone Morphogenetic Protein (BMP)/Transforming Growth Factor ß (TGFß) signaling pathway was implicated in aortic hematopoiesis but the very nature of the signal was unknown. Here, using thorough expression analysis of the BMP/TGFß signaling pathway members in the endothelial and hematopoietic compartments of the aorta at pre-hematopoietic and hematopoietic stages, we show that the TGFß pathway is preferentially balanced with a prominent role of Alk1/TgfßR2/Smad1 and 5 on both chicken and mouse species. Functional analysis using embryonic stem cells mutated for Acvrl1 revealed an enhanced propensity to produce hematopoietic cells. Collectively, we reveal that TGFß through the Alk1/TgfßR2 receptor axis is acting on endothelial cells to produce hematopoiesis.


Asunto(s)
Aorta/embriología , Proteínas Aviares/metabolismo , Endotelio Vascular/embriología , Hematopoyesis Extramedular/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Aorta/citología , Embrión de Pollo , Pollos , Endotelio Vascular/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo
14.
Development ; 142(15): 2719-24, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26243871

RESUMEN

Endothelial-to-hematopoietic transition (EHT) occurs within a population of hemogenic endothelial cells during embryogenesis, and leads to the formation of the adult hematopoietic system. Currently, the prospective identification of specific endothelial cells that will undergo EHT, and the cellular events enabling this transition, are not known. We set out to define precisely the morphological events of EHT, and to correlate cellular morphology with the expression of the transcription factors RUNX1 and SOX17. A novel strategy was developed to allow for correlation of immunofluorescence data with the ultrastructural resolution of scanning electron microscopy. The approach can identify single endothelial cells undergoing EHT, as identified by the ratio of RUNX1 to SOX17 immunofluorescence levels, and the morphological changes associated with the transition. Furthermore, this work details a new technical resource that is widely applicable for correlative analyses of single cells in their native tissue environments.


Asunto(s)
Diferenciación Celular/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Hemangioblastos/citología , Sistema Hematopoyético/embriología , Factores de Transcripción SOXF/metabolismo , Análisis de la Célula Individual/métodos , Animales , Técnica del Anticuerpo Fluorescente , Sistema Hematopoyético/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Microscopía Electrónica de Rastreo , Microscopía Fluorescente
15.
Proc Natl Acad Sci U S A ; 112(14): E1734-43, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25831528

RESUMEN

Lineage specification during development involves reprogramming of transcriptional states, but little is known about how this is regulated in vivo. The chromatin remodeler chomodomain helicase DNA-binding protein 1 (Chd1) promotes an elevated transcriptional output in mouse embryonic stem cells. Here we report that endothelial-specific deletion of Chd1 leads to loss of definitive hematopoietic progenitors, anemia, and lethality by embryonic day (E)15.5. Mutant embryos contain normal numbers of E10.5 intraaortic hematopoietic clusters that express Runx1 and Kit, but these clusters undergo apoptosis and fail to mature into blood lineages in vivo and in vitro. Hematopoietic progenitors emerging from the aorta have an elevated transcriptional output relative to structural endothelium, and this elevation is Chd1-dependent. In contrast, hematopoietic-specific deletion of Chd1 using Vav-Cre has no apparent phenotype. Our results reveal a new paradigm of regulation of a developmental transition by elevation of global transcriptional output that is critical for hemogenesis and may play roles in other contexts.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Transcripción Genética , Animales , Aorta/metabolismo , Apoptosis , Diferenciación Celular , Cromatina/metabolismo , Cruzamientos Genéticos , Proteínas de Unión al ADN/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Familia de Multigenes , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo
16.
Cell Mol Life Sci ; 73(8): 1547-67, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26849156

RESUMEN

Hemogenic endothelium is a specialized subset of developing vascular endothelium that acquires hematopoietic potential and can give rise to multilineage hematopoietic stem and progenitor cells during a narrow developmental window in tissues such as the extraembryonic yolk sac and embryonic aorta-gonad-mesonephros. Herein, we review current knowledge about the historical and developmental origins of hemogenic endothelium, the molecular events that govern hemogenic specification of vascular endothelial cells, the generation of multilineage hematopoietic stem and progenitor cells from hemogenic endothelium, and the potential for translational applications of knowledge gained from further study of these processes.


Asunto(s)
Endotelio Vascular/embriología , Hemangioblastos/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Desarrollo Embrionario , Humanos , Transducción de Señal/fisiología , Tretinoina/metabolismo
17.
Blood Sci ; 6(4): e00199, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39027902

RESUMEN

Hematopoietic stem progenitor cells (HSPCs) are derived from a specialized subset of endothelial cells named hemogenic endothelial cells (HECs) via a process of endothelial-to-hematopoietic transition during embryogenesis. Recently, with the usage of multiple single-cell technologies and advanced genetic lineage tracing techniques, namely, "TIF" approaches that combining transcriptome, immunophenotype and function/fate analyses, massive new insights have been achieved regarding the cellular and molecular evolution underlying the emergence of HSPCs from embryonic vascular beds. In this review, we focus on the most recent advances in the enrichment markers, functional characteristics, developmental paths, molecular controls, and the embryonic site-relevance of the key intermediate cell populations bridging embryonic vascular and hematopoietic systems, namely HECs and pre-hematopoietic stem cells, the immediate progenies of some HECs, in mouse and human embryos. Specifically, using expression analyses at both transcriptional and protein levels and especially efficient functional assays, we propose that the onset of Kit expression is at the HEC stage, which has previously been controversial.

18.
Dev Cell ; 59(8): 961-978.e7, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38508181

RESUMEN

Trans-differentiation represents a direct lineage conversion; however, insufficient characterization of this process hinders its potential applications. Here, to explore a potential universal principal for trans-differentiation, we performed single-cell transcriptomic analysis of endothelial-to-hematopoietic transition (EHT), endothelial-to-mesenchymal transition, and epithelial-to-mesenchymal transition in mouse embryos. We applied three scoring indexes of entropies, cell-type signature transcription factor expression, and critical transition signals to show common features underpinning the fate plasticity of transition states. Cross-model comparison identified inflammatory-featured transition states and a common trigger role of interleukin-33 in promoting fate conversions. Multimodal profiling (integrative transcriptomic and chromatin accessibility analysis) demonstrated the inflammatory regulation of hematopoietic specification. Furthermore, multimodal omics and fate-mapping analyses showed that endothelium-specific Spi1, as an inflammatory effector, governs appropriate chromatin accessibility and transcriptional programs to safeguard EHT. Overall, our study employs single-cell omics to identify critical transition states/signals and the common trigger role of inflammatory signaling in developmental-stress-induced fate conversions.


Asunto(s)
Transdiferenciación Celular , Embrión de Mamíferos , Inflamación , Transducción de Señal , Análisis de la Célula Individual , Animales , Ratones , Análisis de la Célula Individual/métodos , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Embrión de Mamíferos/metabolismo , Transición Epitelial-Mesenquimal , Regulación del Desarrollo de la Expresión Génica , Transcriptoma/genética , Células Endoteliales/metabolismo
19.
Elife ; 122024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38809590

RESUMEN

Hematopoietic stem cells emerge in the embryo from an aortic-derived tissue called the hemogenic endothelium (HE). The HE appears to give birth to cells of different nature and fate but the molecular principles underlying this complexity are largely unknown. Here we show, in the zebrafish embryo, that two cell types emerge from the aortic floor with radically different morphodynamics. With the support of live imaging, we bring evidence suggesting that the mechanics underlying the two emergence types rely, or not, on apicobasal polarity establishment. While the first type is characterized by reinforcement of apicobasal polarity and maintenance of the apical/luminal membrane until release, the second type emerges via a dynamic process reminiscent of trans-endothelial migration. Interfering with Runx1 function suggests that the balance between the two emergence types depends on tuning apicobasal polarity at the level of the HE. In support of this and unexpectedly, we show that Pard3ba - one of the four Pard3 proteins expressed in the zebrafish - is sensitive to interference with Runx1 activity, in aortic endothelial cells. This supports the idea of a signaling cross talk controlling cell polarity and its associated features, between aortic and hemogenic cells. In addition, using new transgenic fish lines that express Junctional Adhesion Molecules and functional interference, we bring evidence for the essential role of ArhGEF11/PDZ-RhoGEF in controlling the HE-endothelial cell dynamic interface, including cell-cell intercalation, which is ultimately required for emergence completion. Overall, we highlight critical cellular and dynamic events of the endothelial-to-hematopoietic transition that support emergence complexity, with a potential impact on cell fate.


In mammals and other animals with backbones, the cells that will make up blood and immune cells are generated during a very narrow timeframe in embryonic development. These cells, called hematopoietic stem cells and progenitors (or HSPCs for short), emerge from tissue known as hemogenic endothelium that makes up the floor of early blood vessels. For HPSCs to eventually specialise into different types of blood and immune cells, they require diverse migratory and homing properties that, ultimately, will determine the specific type of functions they exert. An important question for scientists studying the development of different blood and immune cell types is when this commitment to functional diversity is established. It could, for example, arise due to cells in the hemogenic endothelium having different origins. Alternatively, the signals that generate hemogenic endothelium cells could be responsible. It is also possible that both explanations are true, and that having different mechanisms involved ensures diversity in populations of HSPCs. To investigate differences between the HSPCs emerging from the hemogenic endothelium, Torcq et al. studied zebrafish embryos that had been modified so that one of the proteins involved in sensing cell polarity ­ where the top and bottom of the cell are located ­ was fluorescent. Live imaging of the embryos showed that two types of cells, with striking differences in morphology, emerge from the hemogenic tissue. In addition, one cell type displays the same polarity as the other vessel cells, whereas the other does not. Torcq et al. also present evidence suggesting that the signals responsible for controlling this cell polarity are provided by surrounding blood vessel cells, supporting the idea of an interplay between the different cell types. The finding that two different cell types emerge from the hemogenic endothelium, reveals a potential new source of diversity in HSPCs. Ultimately, this is expected to contribute to their functional complexity, resulting in both long-term stem cells that retain their full regenerative potential into adulthood and more specialized blood and immune cells.


Asunto(s)
Polaridad Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Células Madre Hematopoyéticas , Proteínas de Pez Cebra , Pez Cebra , Pez Cebra/embriología , Animales , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Hematopoyéticas/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hemangioblastos/metabolismo , Hemangioblastos/citología , Hemangioblastos/fisiología , Embrión no Mamífero/metabolismo , Animales Modificados Genéticamente
20.
Blood Cells Mol Dis ; 51(4): 206-12, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24095001

RESUMEN

Definitive hematopoietic cells are generated de novo during ontogeny from a specialized subset of endothelium, the so-called hemogenic endothelium. In this review we give a brief overview of the identification of hemogenic endothelium, explore its links with the HSC lineage, and summarize recent insights into the nature of hemogenic endothelium and the microenvironmental and intrinsic regulators contributing to its transition into blood. Ultimately, a better understanding of the processes controlling the transition of endothelium into blood will advance the generation and expansion of hematopoietic stem cells for therapeutic purposes.


Asunto(s)
Endotelio/fisiología , Hematopoyesis/fisiología , Animales , Linaje de la Célula , Transdiferenciación Celular , Microambiente Celular , Endotelio/embriología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Humanos , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA