Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.649
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 183(3): 702-716.e14, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33125890

RESUMEN

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


Asunto(s)
Linaje de la Célula/genética , Hígado/citología , Hígado/metabolismo , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Movimiento Celular , Embrión de Mamíferos/citología , Endotelio/citología , Mesodermo/citología , Ratones , Transducción de Señal , Células Madre/citología
2.
Cell ; 160(1-2): 241-52, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594182

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) can reconstitute and sustain the entire blood system. We generated a highly specific transgenic reporter of HSPCs in zebrafish. This allowed us to perform high-resolution live imaging on endogenous HSPCs not currently possible in mammalian bone marrow. Using this system, we have uncovered distinct interactions between single HSPCs and their niche. When an HSPC arrives in the perivascular niche, a group of endothelial cells remodel to form a surrounding pocket. This structure appears conserved in mouse fetal liver. Correlative light and electron microscopy revealed that endothelial cells surround a single HSPC attached to a single mesenchymal stromal cell. Live imaging showed that mesenchymal stromal cells anchor HSPCs and orient their divisions. A chemical genetic screen found that the compound lycorine promotes HSPC-niche interactions during development and ultimately expands the stem cell pool into adulthood. Our studies provide evidence for dynamic niche interactions upon stem cell colonization. PAPERFLICK:


Asunto(s)
Endotelio/fisiología , Células Madre Hematopoyéticas/citología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , División Celular , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/fisiología , Endotelio/citología , Células Madre Hematopoyéticas/fisiología , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Nicho de Células Madre , Células del Estroma/citología , Células del Estroma/metabolismo , Pez Cebra/fisiología
3.
Nature ; 621(7980): 813-820, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37587341

RESUMEN

Disruption of the lung endothelial-epithelial cell barrier following respiratory virus infection causes cell and fluid accumulation in the air spaces and compromises vital gas exchange function1. Endothelial dysfunction can exacerbate tissue damage2,3, yet it is unclear whether the lung endothelium promotes host resistance against viral pathogens. Here we show that the environmental sensor aryl hydrocarbon receptor (AHR) is highly active in lung endothelial cells and protects against influenza-induced lung vascular leakage. Loss of AHR in endothelia exacerbates lung damage and promotes the infiltration of red blood cells and leukocytes into alveolar air spaces. Moreover, barrier protection is compromised and host susceptibility to secondary bacterial infections is increased when endothelial AHR is missing. AHR engages tissue-protective transcriptional networks in endothelia, including the vasoactive apelin-APJ peptide system4, to prevent a dysplastic and apoptotic response in airway epithelial cells. Finally, we show that protective AHR signalling in lung endothelial cells is dampened by the infection itself. Maintenance of protective AHR function requires a diet enriched in naturally occurring AHR ligands, which activate disease tolerance pathways in lung endothelia to prevent tissue damage. Our findings demonstrate the importance of endothelial function in lung barrier immunity. We identify a gut-lung axis that affects lung damage following encounters with viral pathogens, linking dietary composition and intake to host fitness and inter-individual variations in disease outcome.


Asunto(s)
Células Endoteliales , Pulmón , Infecciones por Orthomyxoviridae , Receptores de Hidrocarburo de Aril , Animales , Humanos , Ratones , Apelina/metabolismo , Dieta , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/metabolismo , Células Epiteliales/metabolismo , Eritrocitos/metabolismo , Gripe Humana/inmunología , Gripe Humana/metabolismo , Intestinos/metabolismo , Leucocitos/metabolismo , Ligandos , Pulmón/inmunología , Pulmón/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo
4.
Nature ; 601(7893): 446-451, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937935

RESUMEN

Exosomes and other small extracellular vesicles (sEVs) provide a unique mode of cell-to-cell communication in which microRNAs (miRNAs) produced and released from one cell are taken up by cells at a distance where they can enact changes in gene expression1-3. However, the mechanism by which miRNAs are sorted into exosomes/sEVs or retained in cells remains largely unknown. Here we demonstrate that miRNAs possess sorting sequences that determine their secretion in sEVs (EXOmotifs) or cellular retention (CELLmotifs) and that different cell types, including white and brown adipocytes, endothelium, liver and muscle, make preferential use of specific sorting sequences, thus defining the sEV miRNA profile of that cell type. Insertion or deletion of these CELLmotifs or EXOmotifs in a miRNA increases or decreases retention in the cell of production or secretion into exosomes/sEVs. Two RNA-binding proteins, Alyref and Fus, are involved in the export of miRNAs carrying one of the strongest EXOmotifs, CGGGAG. Increased miRNA delivery mediated by EXOmotifs leads to enhanced inhibition of target genes in distant cells. Thus, this miRNA code not only provides important insights that link circulating exosomal miRNAs to tissues of origin, but also provides an approach for improved targeting in RNA-mediated therapies.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Adipocitos/citología , Comunicación Celular , Endotelio/citología , Exosomas/genética , Exosomas/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Hígado/citología , MicroARNs/genética , MicroARNs/metabolismo , Músculos/citología
5.
Nat Immunol ; 15(10): 982-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25173345

RESUMEN

Lymphocytes are recruited from blood by high-endothelial venules (HEVs). We performed transcriptomic analyses and identified molecular signatures that distinguish HEVs from capillary endothelium and that define tissue-specific HEV specialization. Capillaries expressed gene programs for vascular development. HEV-expressed genes showed enrichment for genes encoding molecules involved in immunological defense and lymphocyte migration. We identify capillary and HEV markers and candidate mechanisms for regulated recruitment of lymphocytes, including a lymph node HEV-selective transmembrane mucin; transcriptional control of functionally specialized carbohydrate ligands for lymphocyte L-selectin; HEV expression of molecules for transendothelial migration; and metabolic programs for lipid mediators of lymphocyte motility and chemotaxis. We also elucidate a carbohydrate-recognition pathway that targets B cells to intestinal lymphoid tissues, defining CD22 as a lectin-homing receptor for mucosal HEVs.


Asunto(s)
Capilares/metabolismo , Endotelio/metabolismo , Perfilación de la Expresión Génica , Linfocitos/metabolismo , Tejido Linfoide/irrigación sanguínea , Vénulas/metabolismo , Animales , Movimiento Celular/genética , Células Endoteliales/metabolismo , Endotelio/citología , Femenino , Citometría de Flujo , Ontología de Genes , Ganglios Linfáticos/irrigación sanguínea , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos
6.
Nature ; 583(7815): 296-302, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32612232

RESUMEN

The mammalian immune system implements a remarkably effective set of mechanisms for fighting pathogens1. Its main components are haematopoietic immune cells, including myeloid cells that control innate immunity, and lymphoid cells that constitute adaptive immunity2. However, immune functions are not unique to haematopoietic cells, and many other cell types display basic mechanisms of pathogen defence3-5. To advance our understanding of immunology outside the haematopoietic system, here we systematically investigate the regulation of immune genes in the three major types of structural cells: epithelium, endothelium and fibroblasts. We characterize these cell types across twelve organs in mice, using cellular phenotyping, transcriptome sequencing, chromatin accessibility profiling and epigenome mapping. This comprehensive dataset revealed complex immune gene activity and regulation in structural cells. The observed patterns were highly organ-specific and seem to modulate the extensive interactions between structural cells and haematopoietic immune cells. Moreover, we identified an epigenetically encoded immune potential in structural cells under tissue homeostasis, which was triggered in response to systemic viral infection. This study highlights the prevalence and organ-specific complexity of immune gene activity in non-haematopoietic structural cells, and it provides a high-resolution, multi-omics atlas of the epigenetic and transcriptional networks that regulate structural cells in the mouse.


Asunto(s)
Endotelio/inmunología , Células Epiteliales/inmunología , Fibroblastos/inmunología , Regulación de la Expresión Génica/inmunología , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Especificidad de Órganos/inmunología , Inmunidad Adaptativa , Animales , Cromatina/genética , Cromatina/metabolismo , Endotelio/citología , Epigénesis Genética/inmunología , Epigenoma/genética , Células Epiteliales/citología , Femenino , Fibroblastos/citología , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Sistema Inmunológico/virología , Inmunidad Innata , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Especificidad de Órganos/genética , Transcripción Genética/inmunología , Transcriptoma/genética
7.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35129199

RESUMEN

Skeletal elements frequently associate with vasculature and somatosensory nerves, which regulate bone development and homeostasis. However, the deep, internal location of bones in many vertebrates has limited in vivo exploration of the neurovascular-bone relationship. Here, we use the zebrafish caudal fin, an optically accessible organ formed of repeating bony ray skeletal units, to determine the cellular relationship between nerves, bones and endothelium. In adult zebrafish, we establish the presence of somatosensory axons running through the inside of the bony fin rays, juxtaposed with osteoblasts on the inner hemiray surface. During development we show that the caudal fin progresses through sequential stages of endothelial plexus formation, bony ray addition, ray innervation and endothelial remodeling. Surprisingly, the initial stages of fin morphogenesis proceed normally in animals lacking either fin endothelium or somatosensory nerves. Instead, we find that sp7+ osteoblasts are required for endothelial remodeling and somatosensory axon innervation in the developing fin. Overall, this study demonstrates that the proximal neurovascular-bone relationship in the adult caudal fin is established during fin organogenesis and suggests that ray-associated osteoblasts pattern axons and endothelium.


Asunto(s)
Aletas de Animales/fisiología , Axones/metabolismo , Endotelio/metabolismo , Organogénesis/fisiología , Pez Cebra/crecimiento & desarrollo , Aletas de Animales/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Endotelio/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Larva/crecimiento & desarrollo , Larva/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción Sp7/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
Immunity ; 44(4): 901-12, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27096319

RESUMEN

Sickness behavior and cognitive dysfunction occur frequently by unknown mechanisms in virus-infected individuals with malignancies treated with type I interferons (IFNs) and in patients with autoimmune disorders. We found that during sickness behavior, single-stranded RNA viruses, double-stranded RNA ligands, and IFNs shared pathways involving engagement of melanoma differentiation-associated protein 5 (MDA5), retinoic acid-inducible gene 1 (RIG-I), and mitochondrial antiviral signaling protein (MAVS), and subsequently induced IFN responses specifically in brain endothelia and epithelia of mice. Behavioral alterations were specifically dependent on brain endothelial and epithelial IFN receptor chain 1 (IFNAR). Using gene profiling, we identified that the endothelia-derived chemokine ligand CXCL10 mediated behavioral changes through impairment of synaptic plasticity. These results identified brain endothelial and epithelial cells as natural gatekeepers for virus-induced sickness behavior, demonstrated tissue specific IFNAR engagement, and established the CXCL10-CXCR3 axis as target for the treatment of behavioral changes during virus infection and type I IFN therapy.


Asunto(s)
Encéfalo/citología , Quimiocina CXCL10/inmunología , Trastornos del Conocimiento/genética , Células Endoteliales/inmunología , Células Epiteliales/inmunología , Conducta de Enfermedad/fisiología , Receptor de Interferón alfa y beta/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Encéfalo/inmunología , Comunicación Celular/inmunología , Células Cultivadas , Trastornos del Conocimiento/psicología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Endotelio/citología , Endotelio/inmunología , Epitelio/inmunología , Interferón Tipo I/uso terapéutico , Helicasa Inducida por Interferón IFIH1 , Masculino , Ratones , ARN Bicatenario/genética , Receptor de Interferón alfa y beta/inmunología , Receptores CXCR3/inmunología , Transducción de Señal/inmunología , Virosis/inmunología
9.
Nature ; 566(7745): 490-495, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30787436

RESUMEN

Across the animal kingdom, gastrulation represents a key developmental event during which embryonic pluripotent cells diversify into lineage-specific precursors that will generate the adult organism. Here we report the transcriptional profiles of 116,312 single cells from mouse embryos collected at nine sequential time points ranging from 6.5 to 8.5 days post-fertilization. We construct a molecular map of cellular differentiation from pluripotency towards all major embryonic lineages, and explore the complex events involved in the convergence of visceral and primitive streak-derived endoderm. Furthermore, we use single-cell profiling to show that Tal1-/- chimeric embryos display defects in early mesoderm diversification, and we thus demonstrate how combining temporal and transcriptional information can illuminate gene function. Together, this comprehensive delineation of mammalian cell differentiation trajectories in vivo represents a baseline for understanding the effects of gene mutations during development, as well as a roadmap for the optimization of in vitro differentiation protocols for regenerative medicine.


Asunto(s)
Diferenciación Celular/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Gastrulación , Organogénesis , Análisis de la Célula Individual , Animales , Linaje de la Célula/genética , Quimera/embriología , Quimera/genética , Quimera/metabolismo , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Endotelio/citología , Endotelio/embriología , Endotelio/metabolismo , Femenino , Gastrulación/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Hematopoyesis/genética , Masculino , Mesodermo/citología , Mesodermo/embriología , Ratones , Mutación/genética , Células Mieloides/citología , Organogénesis/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Línea Primitiva/embriología , Proteína 1 de la Leucemia Linfocítica T Aguda/deficiencia , Proteína 1 de la Leucemia Linfocítica T Aguda/genética
10.
Nature ; 545(7655): 432-438, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28514439

RESUMEN

A variety of tissue lineages can be differentiated from pluripotent stem cells by mimicking embryonic development through stepwise exposure to morphogens, or by conversion of one differentiated cell type into another by enforced expression of master transcription factors. Here, to yield functional human haematopoietic stem cells, we perform morphogen-directed differentiation of human pluripotent stem cells into haemogenic endothelium followed by screening of 26 candidate haematopoietic stem-cell-specifying transcription factors for their capacity to promote multi-lineage haematopoietic engraftment in mouse hosts. We recover seven transcription factors (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1 and SPI1) that are sufficient to convert haemogenic endothelium into haematopoietic stem and progenitor cells that engraft myeloid, B and T cells in primary and secondary mouse recipients. Our combined approach of morphogen-driven differentiation and transcription-factor-mediated cell fate conversion produces haematopoietic stem and progenitor cells from pluripotent stem cells and holds promise for modelling haematopoietic disease in humanized mice and for therapeutic strategies in genetic blood disorders.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Animales , Reprogramación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Endotelio/citología , Femenino , Trasplante de Células Madre Hematopoyéticas , Proteínas Homeobox A10 , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Regulador Transcripcional ERG/metabolismo
11.
Nature ; 546(7659): 533-538, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28614297

RESUMEN

Conventional two-dimensional differentiation from pluripotency fails to recapitulate cell interactions occurring during organogenesis. Three-dimensional organoids generate complex organ-like tissues; however, it is unclear how heterotypic interactions affect lineage identity. Here we use single-cell RNA sequencing to reconstruct hepatocyte-like lineage progression from pluripotency in two-dimensional culture. We then derive three-dimensional liver bud organoids by reconstituting hepatic, stromal, and endothelial interactions, and deconstruct heterogeneity during liver bud development. We find that liver bud hepatoblasts diverge from the two-dimensional lineage, and express epithelial migration signatures characteristic of organ budding. We benchmark three-dimensional liver buds against fetal and adult human liver single-cell RNA sequencing data, and find a striking correspondence between the three-dimensional liver bud and fetal liver cells. We use a receptor-ligand pairing analysis and a high-throughput inhibitor assay to interrogate signalling in liver buds, and show that vascular endothelial growth factor (VEGF) crosstalk potentiates endothelial network formation and hepatoblast differentiation. Our molecular dissection reveals interlineage communication regulating organoid development, and illuminates previously inaccessible aspects of human liver development.


Asunto(s)
Comunicación Celular , Diferenciación Celular , Linaje de la Célula , Hígado/citología , Hígado/embriología , Organogénesis , Técnicas de Cultivo de Tejidos/métodos , Anciano , Hipoxia de la Célula , Movimiento Celular , Endotelio/citología , Células Epiteliales/citología , Matriz Extracelular/metabolismo , Femenino , Feto/citología , Hepatocitos/citología , Humanos , Masculino , Persona de Mediana Edad , Organoides/citología , Células Madre Pluripotentes/citología , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
12.
Nature ; 545(7655): 439-445, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28514438

RESUMEN

Developmental pathways that orchestrate the fleeting transition of endothelial cells into haematopoietic stem cells remain undefined. Here we demonstrate a tractable approach for fully reprogramming adult mouse endothelial cells to haematopoietic stem cells (rEC-HSCs) through transient expression of the transcription-factor-encoding genes Fosb, Gfi1, Runx1, and Spi1 (collectively denoted hereafter as FGRS) and vascular-niche-derived angiocrine factors. The induction phase (days 0-8) of conversion is initiated by expression of FGRS in mature endothelial cells, which results in endogenous Runx1 expression. During the specification phase (days 8-20), RUNX1+ FGRS-transduced endothelial cells commit to a haematopoietic fate, yielding rEC-HSCs that no longer require FGRS expression. The vascular niche drives a robust self-renewal and expansion phase of rEC-HSCs (days 20-28). rEC-HSCs have a transcriptome and long-term self-renewal capacity similar to those of adult haematopoietic stem cells, and can be used for clonal engraftment and serial primary and secondary multi-lineage reconstitution, including antigen-dependent adaptive immune function. Inhibition of TGFß and CXCR7 or activation of BMP and CXCR4 signalling enhanced generation of rEC-HSCs. Pluripotency-independent conversion of endothelial cells into autologous authentic engraftable haematopoietic stem cells could aid treatment of haematological disorders.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Endotelio/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Inmunidad Adaptativa , Envejecimiento/genética , Animales , Línea Celular , Linaje de la Célula , Autorrenovación de las Células , Células Clonales/citología , Células Clonales/trasplante , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
13.
Blood ; 136(7): 845-856, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32392346

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Endotelio/embriología , Hemangioblastos/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Diferenciación Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Embrión de Mamíferos , Endotelio/citología , Endotelio/metabolismo , Femenino , Dosificación de Gen/fisiología , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos/citología , Hematopoyesis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , RNA-Seq/métodos
14.
J Immunol ; 204(4): 1012-1021, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31924649

RESUMEN

Cell polarization is a key step for leukocytes adhesion and transmigration during leukocytes' inflammatory infiltration. Polarized localization of plasma membrane (PM) phosphatidylinositol-4-phosphate (PtdIns4P) directs the polarization of RPH3A, which contains a PtdIns4P binding site. Consequently, RPH3A mediates the RAB21 and PIP5K1C90 polarization, which is important for neutrophil adhesion to endothelia during inflammation. However, the mechanism by which RPH3A is recruited only to PM PtdIns4P rather than Golgi PtdIns4P remains unclear. By using ADP-ribosylation factor 6 (ARF6) small interfering RNA, ARF6 dominant-negative mutant ARF6(T27N), and ARF6 activation inhibitor SecinH3, we demonstrate that ARF6 plays an important role in the polarization of RPH3A, RAB21, and PIP5K1C90 in murine neutrophils. PM ARF6 is polarized and colocalized with RPH3A, RAB21, PIP5K1C90, and PM PtdIns4P in mouse and human neutrophils upon integrin stimulation. Additionally, ARF6 binds to RPH3A and enhances the interaction between the PM PtdIns4P and RPH3A. Consistent with functional roles of polarization of RPH3A, Rab21, and PIP5K1C90, ARF6 is also required for neutrophil adhesion on the inflamed endothelial layer. Our study reveals a previously unknown role of ARF6 in neutrophil polarization as being the coincidence-detection code with PM PtdIns4P. Cooperation of ARF6 and PM PtdIns4P direct RPH3A polarization, which is important for neutrophil firm adhesion to endothelia.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endotelio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Activación Neutrófila , Neutrófilos/inmunología , Proteínas de Transporte Vesicular/metabolismo , Factor 6 de Ribosilación del ADP , Animales , Adhesión Celular/inmunología , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular/inmunología , Células Endoteliales , Endotelio/citología , Endotelio/inmunología , Femenino , Voluntarios Sanos , Humanos , Ratones , Neutrófilos/citología , Neutrófilos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Cultivo Primario de Células , Rabfilina-3A
15.
Mol Biol Rep ; 49(9): 8495-8505, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35802277

RESUMEN

BACKGROUND: The present study aimed to investigate the mechanisms through which long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) affected the endothelial differentiation of mouse derived adipose-derived stem cells (ADSCs). MATERIALS AND METHODS: ADSCs were isolated and identified by specific surface marker detection. The effects of lncRNA MEG3 on endothelial differentiation of ADSCs were also detected via quantitative PCR, western blotting, immunofluorescence and Matrigel angiogenesis assays. In addition, using target gene prediction tools and luciferase reporter assays, the downstream target gene was demonstrated. RESULTS: LncRNA MEG3 targeted and reduced the expression levels of microRNA-145-5p (miR-145-5p), which upregulated the expression levels of Krüppel like factor 4 (KLF4), promoting endothelial differentiation of ADSCs. CONCLUSION: LncRNA MEG3 induced endothelial differentiation of ADSCs by targeting miR-145-5p/KLF4, which may provide novel insights to illustrate the mechanism of endothelial differentiation of ADSCs.


Asunto(s)
Endotelio , Factor 4 Similar a Kruppel , MicroARNs , ARN Largo no Codificante , Células Madre , Tejido Adiposo/citología , Animales , Diferenciación Celular/genética , Endotelio/citología , Factor 4 Similar a Kruppel/genética , Ratones , MicroARNs/genética , ARN Largo no Codificante/genética , Células Madre/metabolismo
16.
Nature ; 538(7625): 392-396, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27732581

RESUMEN

Macrophages are required for normal embryogenesis, tissue homeostasis and immunity against microorganisms and tumours. Adult tissue-resident macrophages largely originate from long-lived, self-renewing embryonic precursors and not from haematopoietic stem-cell activity in the bone marrow. Although fate-mapping studies have uncovered a great amount of detail on the origin and kinetics of fetal macrophage development in the yolk sac and liver, the molecules that govern the tissue-specific migration of these cells remain completely unknown. Here we show that an endothelium-specific molecule, plasmalemma vesicle-associated protein (PLVAP), regulates the seeding of fetal monocyte-derived macrophages to tissues in mice. We found that PLVAP-deficient mice have completely normal levels of both yolk-sac- and bone-marrow-derived macrophages, but that fetal liver monocyte-derived macrophage populations were practically missing from tissues. Adult PLVAP-deficient mice show major alterations in macrophage-dependent iron recycling and mammary branching morphogenesis. PLVAP forms diaphragms in the fenestrae of liver sinusoidal endothelium during embryogenesis, interacts with chemoattractants and adhesion molecules and regulates the egress of fetal liver monocytes to the systemic vasculature. Thus, PLVAP selectively controls the exit of macrophage precursors from the fetal liver and, to our knowledge, is the first molecule identified in any organ as regulating the migratory events during embryonic macrophage ontogeny.


Asunto(s)
Proteínas Portadoras/metabolismo , Linaje de la Célula , Movimiento Celular , Endotelio/citología , Feto/citología , Hígado/citología , Hígado/metabolismo , Macrófagos/citología , Proteínas de la Membrana/metabolismo , Animales , Vasos Sanguíneos/citología , Células de la Médula Ósea/citología , Proteínas Portadoras/genética , Endotelio/metabolismo , Femenino , Feto/metabolismo , Heparina/metabolismo , Homeostasis , Hierro/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Monocitos/citología , Morfogénesis , Neuropilina-1/metabolismo , Especificidad de Órganos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Saco Vitelino/citología
17.
Cell Mol Life Sci ; 78(23): 7107-7132, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34613423

RESUMEN

Over the past few years, extensive efforts have been made to generate in-vitro pancreatic micro-tissue, for disease modeling or cell replacement approaches in pancreatic related diseases such as diabetes mellitus. To obtain these goals, a closer look at the diverse cells participating in pancreatic development is necessary. Five major non-epithelial pancreatic (pN-Epi) cell populations namely, pancreatic endothelium, mesothelium, neural crests, pericytes, and stellate cells exist in pancreas throughout its development, and they are hypothesized to be endogenous inducers of the development. In this review, we discuss different pN-Epi cells migrating to and existing within the pancreas and their diverse effects on pancreatic epithelium during organ development mediated via associated signaling pathways, soluble factors or mechanical cell-cell interactions. In-vivo and in-vitro experiments, with a focus on N-Epi cells' impact on pancreas endocrine development, have also been considered. Pluripotent stem cell technology and multicellular three-dimensional organoids as new approaches to generate pancreatic micro-tissues have also been discussed. Main challenges for reaching a detailed understanding of the role of pN-Epi cells in pancreas development in utilizing for in-vitro recapitulation have been summarized. Finally, various novel and innovative large-scale bioengineering approaches which may help to recapitulate cell-cell interactions and are crucial for generation of large-scale in-vitro multicellular pancreatic micro-tissues, are discussed.


Asunto(s)
Comunicación Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Diabetes Mellitus/terapia , Páncreas/crecimiento & desarrollo , Ingeniería de Tejidos/métodos , Diferenciación Celular/fisiología , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/metabolismo , Humanos , Organogénesis/fisiología , Organoides/citología , Páncreas/citología , Enfermedades Pancreáticas/terapia , Células Madre Pluripotentes/citología
18.
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
19.
J Biol Chem ; 295(9): 2804-2821, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31964714

RESUMEN

Animal cells express heparan sulfate proteoglycans that perform many important cellular functions by way of heparan sulfate-protein interactions. The identification of membrane heparan sulfate-binding proteins is challenging because of their low abundance and the need for extensive enrichment. Here, we report a proteomics workflow for the identification and characterization of membrane-anchored and extracellular proteins that bind heparan sulfate. The technique is based on limited proteolysis of live cells in the absence of denaturation and fixation, heparin-affinity chromatography, and high-resolution LC-MS/MS, and we designate it LPHAMS. Application of LPHAMS to U937 monocytic and primary murine and human endothelial cells identified 55 plasma membrane, extracellular matrix, and soluble secreted proteins, including many previously unidentified heparin-binding proteins. The method also facilitated the mapping of the heparin-binding domains, making it possible to predict the location of the heparin-binding site. To validate the discovery feature of LPHAMS, we characterized one of the newly-discovered heparin-binding proteins, C-type lectin 14a (CLEC14A), a member of the C-type lectin family that modulates angiogenesis. We found that the C-type lectin domain of CLEC14A binds one-to-one to heparin with nanomolar affinity, and using molecular modeling and mutagenesis, we mapped its heparin-binding site. CLEC14A physically interacted with other glycosaminoglycans, including endothelial heparan sulfate and chondroitin sulfate E, but not with neutral or sialylated oligosaccharides. The LPHAMS technique should be applicable to other cells and glycans and provides a way to expand the repertoire of glycan-binding proteins for further study.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Endotelio/química , Heparitina Sulfato/metabolismo , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Proteómica/métodos , Animales , Sitios de Unión , Células Cultivadas , Endotelio/citología , Humanos , Ratones , Unión Proteica , Células U937
20.
J Biol Chem ; 295(51): 17632-17645, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33454003

RESUMEN

Thoracic great vessels such as the aorta and subclavian arteries are formed through dynamic remodeling of embryonic pharyngeal arch arteries (PAAs). Previous work has shown that loss of a basic helix-loop-helix transcription factor Hey1 in mice causes abnormal fourth PAA development and lethal great vessel anomalies resembling congenital malformations in humans. However, how Hey1 mediates vascular formation remains unclear. In this study, we revealed that Hey1 in vascular endothelial cells, but not in smooth muscle cells, played essential roles for PAA development and great vessel morphogenesis in mouse embryos. Tek-Cre-mediated Hey1 deletion in endothelial cells affected endothelial tube formation and smooth muscle differentiation in embryonic fourth PAAs and resulted in interruption of the aortic arch and other great vessel malformations. Cell specificity and signal responsiveness of Hey1 expression were controlled through multiple cis-regulatory regions. We found two distal genomic regions that had enhancer activity in endothelial cells and in the pharyngeal epithelium and somites, respectively. The novel endothelial enhancer was conserved across species and was specific to large-caliber arteries. Its transcriptional activity was regulated by Notch signaling in vitro and in vivo, but not by ALK1 signaling and other transcription factors implicated in endothelial cell specificity. The distal endothelial enhancer was not essential for basal Hey1 expression in mouse embryos but may likely serve for Notch-dependent transcriptional control in endothelial cells together with the proximal regulatory region. These findings help in understanding the significance and regulation of endothelial Hey1 as a mediator of multiple signaling pathways in embryonic vascular formation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Endotelio/metabolismo , Receptores Notch/metabolismo , Animales , Arterias/crecimiento & desarrollo , Arterias/metabolismo , Región Branquial/irrigación sanguínea , Región Branquial/crecimiento & desarrollo , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Diferenciación Celular , Embrión de Mamíferos/metabolismo , Endotelio/citología , Femenino , Humanos , Ratones , Ratones Noqueados , Morfogénesis , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Transducción de Señal , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA