Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 151(3): 559-75, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23084400

RESUMEN

ETS transcription factors ETV2, FLI1, and ERG1 specify pluripotent stem cells into induced vascular endothelial cells (iVECs). However, iVECs are unstable and drift toward nonvascular cells. We show that human midgestation c-Kit(-) lineage-committed amniotic cells (ACs) can be reprogrammed into vascular endothelial cells (rAC-VECs) without transitioning through a pluripotent state. Transient ETV2 expression in ACs generates immature rAC-VECs, whereas coexpression with FLI1/ERG1 endows rAC-VECs with a vascular repertoire and morphology matching mature endothelial cells (ECs). Brief TGFß-inhibition functionalizes VEGFR2 signaling, augmenting specification of ACs into rAC-VECs. Genome-wide transcriptional analyses showed that rAC-VECs are similar to adult ECs in which vascular-specific genes are expressed and nonvascular genes are silenced. Functionally, rAC-VECs form stable vasculature in Matrigel plugs and regenerating livers. Therefore, short-term ETV2 expression and TGFß inhibition with constitutive ERG1/FLI1 coexpression reprogram mature ACs into durable rAC-VECs with clinical-scale expansion potential. Banking of HLA-typed rAC-VECs establishes a vascular inventory for treatment of diverse disorders.


Asunto(s)
Líquido Amniótico/citología , Diferenciación Celular , Células Endoteliales/citología , Proteínas Proto-Oncogénicas c-ets/metabolismo , Proteínas Oncogénicas de Retroviridae/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos
2.
Cell ; 147(3): 539-53, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036563

RESUMEN

To identify pathways involved in adult lung regeneration, we employ a unilateral pneumonectomy (PNX) model that promotes regenerative alveolarization in the remaining intact lung. We show that PNX stimulates pulmonary capillary endothelial cells (PCECs) to produce angiocrine growth factors that induce proliferation of epithelial progenitor cells supporting alveologenesis. Endothelial cells trigger expansion of cocultured epithelial cells, forming three-dimensional angiospheres reminiscent of alveolar-capillary sacs. After PNX, endothelial-specific inducible genetic ablation of Vegfr2 and Fgfr1 in mice inhibits production of MMP14, impairing alveolarization. MMP14 promotes expansion of epithelial progenitor cells by unmasking cryptic EGF-like ectodomains that activate the EGF receptor (EGFR). Consistent with this, neutralization of MMP14 impairs EGFR-mediated alveolar regeneration, whereas administration of EGF or intravascular transplantation of MMP14(+) PCECs into pneumonectomized Vegfr2/Fgfr1-deficient mice restores alveologenesis and lung inspiratory volume and compliance function. VEGFR2 and FGFR1 activation in PCECs therefore increases MMP14-dependent bioavailability of EGFR ligands to initiate and sustain alveologenesis.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Pulmón/citología , Pulmón/fisiología , Alveolos Pulmonares/citología , Animales , Células Endoteliales/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Neovascularización Fisiológica , Neumonectomía , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Regeneración , Células Madre/metabolismo , Técnicas de Cultivo de Tejidos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Nature ; 585(7825): 426-432, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908310

RESUMEN

Endothelial cells adopt tissue-specific characteristics to instruct organ development and regeneration1,2. This adaptability is lost in cultured adult endothelial cells, which do not vascularize tissues in an organotypic manner. Here, we show that transient reactivation of the embryonic-restricted ETS variant transcription factor 2 (ETV2)3 in mature human endothelial cells cultured in a serum-free three-dimensional matrix composed of a mixture of laminin, entactin and type-IV collagen (LEC matrix) 'resets' these endothelial cells to adaptable, vasculogenic cells, which form perfusable and plastic vascular plexi. Through chromatin remodelling, ETV2 induces tubulogenic pathways, including the activation of RAP1, which promotes the formation of durable lumens4,5. In three-dimensional matrices-which do not have the constraints of bioprinted scaffolds-the 'reset' vascular endothelial cells (R-VECs) self-assemble into stable, multilayered and branching vascular networks within scalable microfluidic chambers, which are capable of transporting human blood. In vivo, R-VECs implanted subcutaneously in mice self-organize into durable pericyte-coated vessels that functionally anastomose to the host circulation and exhibit long-lasting patterning, with no evidence of malformations or angiomas. R-VECs directly interact with cells within three-dimensional co-cultured organoids, removing the need for the restrictive synthetic semipermeable membranes that are required for organ-on-chip systems, therefore providing a physiological platform for vascularization, which we call 'Organ-On-VascularNet'. R-VECs enable perfusion of glucose-responsive insulin-secreting human pancreatic islets, vascularize decellularized rat intestines and arborize healthy or cancerous human colon organoids. Using single-cell RNA sequencing and epigenetic profiling, we demonstrate that R-VECs establish an adaptive vascular niche that differentially adjusts and conforms to organoids and tumoroids in a tissue-specific manner. Our Organ-On-VascularNet model will permit metabolic, immunological and physiochemical studies and screens to decipher the crosstalk between organotypic endothelial cells and parenchymal cells for identification of determinants of endothelial cell heterogeneity, and could lead to advances in therapeutic organ repair and tumour targeting.


Asunto(s)
Vasos Sanguíneos/citología , Carcinogénesis , Células Endoteliales/citología , Hemodinámica , Neoplasias/irrigación sanguínea , Organogénesis , Organoides/irrigación sanguínea , Vasos Sanguíneos/crecimiento & desarrollo , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Cromatina/metabolismo , Epigénesis Genética , Epigenómica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Islotes Pancreáticos/irrigación sanguínea , Modelos Biológicos , Especificidad de Órganos , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción , Transcriptoma
4.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-35008993

RESUMEN

Cell migration is a complex, tightly regulated multistep process in which cytoskeletal reorganization and focal adhesion redistribution play a central role. Core to both individual and collective migration is the persistent random walk, which is characterized by random force generation and resistance to directional change. We first discuss a model that describes the stochastic movement of ECs and characterizes EC persistence in wound healing. To that end, we pharmacologically disrupted cytoskeletal dynamics, cytochalasin D for actin and nocodazole for tubulin, to understand its contributions to cell morphology, stiffness, and motility. As such, the use of Atomic Force Microscopy (AFM) enabled us to probe the topography and stiffness of ECs, while time lapse microscopy provided observations in wound healing models. Our results suggest that actin and tubulin dynamics contribute to EC shape, compressive moduli, and directional organization in collective migration. Insights from the model and time lapse experiment suggest that EC speed and persistence are directionally organized in wound healing. Pharmacological disruptions suggest that actin and tubulin dynamics play a role in collective migration. Current insights from both the model and experiment represent an important step in understanding the biomechanics of EC migration as a therapeutic target.


Asunto(s)
Rastreo Celular , Citoesqueleto/metabolismo , Células Endoteliales/fisiología , Algoritmos , Animales , Biomarcadores , Movimiento Celular , Rastreo Celular/métodos , Técnica del Anticuerpo Fluorescente , Humanos , Microscopía de Fuerza Atómica , Modelos Biológicos , Imagen Molecular/métodos , Cicatrización de Heridas
5.
Microvasc Res ; 138: 104205, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34146583

RESUMEN

The rapid engraftment of vascular networks is critical for functional incorporation of tissue explants. However, existing methods for inducing angiogenesis utilize approaches that yield vasculature with poor temporal stability or inadequate mechanical integrity, which reduce their robustness in vivo. The transcription factor Ets variant 2 (Etv2) specifies embryonic hematopoietic and vascular endothelial cell (EC) development, and is transiently reactivated during postnatal vascular regeneration and tumor angiogenesis. This study investigates the role for Etv2 upregulation in forming stable vascular beds both in vitro and in vivo. Control and Etv2+ prototypical fetal-derived human umbilical vein ECs (HUVECs) and adult ECs were angiogenically grown into vascular beds. These vessel beds were characterized using fractal dimension and lacunarity, to quantify their branching complexity and space-filling homogeneity, respectively. Atomic force microscopy (AFM) was used to explore whether greater complexity and homogeneity lead to more mechanically stable vessels. Additionally, markers of EC integrity were used to probe for mechanistic clues. Etv2+ HUVECs exhibit greater branching, vessel density, and structural homogeneity, and decreased stiffness in vitro and in vivo, indicating a greater propensity for stable vessel formation. When co-cultured with colon tumor organoid tissue, Etv2+ HUVECs had decreased fractal dimension and lacunarity compared to Etv2+ HUVECs cultured alone, indicating that vessel density and homogeneity of vessel spacing increased due to the presence of Etv2. This study sets forth the novel concept that fractal dimension, lacunarity, and AFM are as informative as conventional angiogenic measurements, including vessel branching and density, to assess vascular perfusion and stability.


Asunto(s)
Forma de la Célula , Neoplasias del Colon/irrigación sanguínea , Fractales , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Procesamiento de Imagen Asistido por Computador , Microscopía de Fuerza Atómica , Neovascularización Fisiológica , Factores de Transcripción/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Neovascularización Patológica , Protocadherinas/metabolismo , Técnicas de Cultivo de Tejidos , Factores de Transcripción/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Nature ; 505(7481): 97-102, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24256728

RESUMEN

Chemical or traumatic damage to the liver is frequently associated with aberrant healing (fibrosis) that overrides liver regeneration. The mechanism by which hepatic niche cells differentially modulate regeneration and fibrosis during liver repair remains to be defined. Hepatic vascular niche predominantly represented by liver sinusoidal endothelial cells deploys paracrine trophogens, known as angiocrine factors, to stimulate regeneration. Nevertheless, it is not known how pro-regenerative angiocrine signals from liver sinusoidal endothelial cells is subverted to promote fibrosis. Here, by combining an inducible endothelial-cell-specific mouse gene deletion strategy and complementary models of acute and chronic liver injury, we show that divergent angiocrine signals from liver sinusoidal endothelial cells stimulate regeneration after immediate injury and provoke fibrosis after chronic insult. The pro-fibrotic transition of vascular niche results from differential expression of stromal-derived factor-1 receptors, CXCR7 and CXCR4 (refs 18, 19, 20, 21), in liver sinusoidal endothelial cells. After acute injury, CXCR7 upregulation in liver sinusoidal endothelial cells acts with CXCR4 to induce transcription factor Id1, deploying pro-regenerative angiocrine factors and triggering regeneration. Inducible deletion of Cxcr7 in sinusoidal endothelial cells (Cxcr7(iΔEC/iΔEC)) from the adult mouse liver impaired liver regeneration by diminishing Id1-mediated production of angiocrine factors. By contrast, after chronic injury inflicted by iterative hepatotoxin (carbon tetrachloride) injection and bile duct ligation, constitutive FGFR1 signalling in liver sinusoidal endothelial cells counterbalanced CXCR7-dependent pro-regenerative response and augmented CXCR4 expression. This predominance of CXCR4 over CXCR7 expression shifted angiocrine response of liver sinusoidal endothelial cells, stimulating proliferation of desmin(+) hepatic stellate-like cells and enforcing a pro-fibrotic vascular niche. Endothelial-cell-specific ablation of either Fgfr1 (Fgfr1(iΔEC/iΔEC)) or Cxcr4 (Cxcr4(iΔEC/iΔEC)) in mice restored the pro-regenerative pathway and prevented FGFR1-mediated maladaptive subversion of angiocrine factors. Similarly, selective CXCR7 activation in liver sinusoidal endothelial cells abrogated fibrogenesis. Thus, we demonstrate that in response to liver injury, differential recruitment of pro-regenerative CXCR7-Id1 versus pro-fibrotic FGFR1-CXCR4 angiocrine pathways in vascular niche balances regeneration and fibrosis. These results provide a therapeutic roadmap to achieve hepatic regeneration without provoking fibrosis.


Asunto(s)
Cirrosis Hepática/patología , Regeneración Hepática/fisiología , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Transducción de Señal , Enfermedad Aguda , Animales , Conductos Biliares/cirugía , Tetracloruro de Carbono , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/patología , Quimiocina CXCL12/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ligadura , Ratones , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
7.
Nature ; 562(7725): 42-43, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30275551
8.
Microcirculation ; 25(5): e12455, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29665185

RESUMEN

OBJECTIVE: Endothelial cells form vascular beds in all organs and are exposed to a range of mechanical forces that regulate cellular phenotype. We sought to determine the role of endothelial luminal surface stiffness in tissue-specific mechanotransduction of laminar shear stress in microvascular mouse cells and the role of arachidonic acid in mediating this response. METHODS: Microvascular mouse endothelial cells were subjected to laminar shear stress at 4 dynes/cm2 for 12 hours in parallel plate flow chambers that enabled real-time optical microscopy and atomic force microscopy measurements of cell stiffness. RESULTS: Lung endothelial cells aligned parallel to flow, while cardiac endothelial cells did not. This rapid alignment was accompanied by increased cell stiffness. The addition of arachidonic acid to cardiac endothelial cells increased alignment and stiffness in response to shear stress. Inhibition of arachidonic acid in lung endothelial cells and embryonic stem cell-derived endothelial cells prevented cellular alignment and decreased cell stiffness. CONCLUSIONS: Our findings suggest that increased endothelial luminal surface stiffness in microvascular cells may facilitate mechanotransduction and alignment in response to laminar shear stress. Furthermore, the arachidonic acid pathway may mediate this tissue-specific process. An improved understanding of this response will aid in the treatment of organ-specific vascular disease.


Asunto(s)
Células Endoteliales/fisiología , Mecanotransducción Celular , Estrés Mecánico , Animales , Ácido Araquidónico/farmacología , Fenómenos Biomecánicos , Células Cultivadas , Pulmón/citología , Ratones , Microcirculación , Miocardio/citología , Propiedades de Superficie
9.
Nature ; 468(7321): 310-5, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21068842

RESUMEN

During embryogenesis, endothelial cells induce organogenesis before the development of circulation. These findings suggest that endothelial cells not only form passive conduits to deliver nutrients and oxygen, but also establish an instructive vascular niche, which through elaboration of paracrine trophogens stimulates organ regeneration, in a manner similar to endothelial-cell-derived angiocrine factors that support haematopoiesis. However, the precise mechanism by which tissue-specific subsets of endothelial cells promote organogenesis in adults is unknown. Here we demonstrate that liver sinusoidal endothelial cells (LSECs) constitute a unique population of phenotypically and functionally defined VEGFR3(+)CD34(-)VEGFR2(+)VE-cadherin(+)FactorVIII(+)CD45(-) endothelial cells, which through the release of angiocrine trophogens initiate and sustain liver regeneration induced by 70% partial hepatectomy. After partial hepatectomy, residual liver vasculature remains intact without experiencing hypoxia or structural damage, which allows study of physiological liver regeneration. Using this model, we show that inducible genetic ablation of vascular endothelial growth factor (VEGF)-A receptor-2 (VEGFR2) in the LSECs impairs the initial burst of hepatocyte proliferation (days 1-3 after partial hepatectomy) and subsequent reconstitution of the hepatovascular mass (days 4-8 after partial hepatectomy) by inhibiting upregulation of the endothelial-cell-specific transcription factor Id1. Accordingly, Id1-deficient mice also manifest defects throughout liver regeneration, owing to diminished expression of LSEC-derived angiocrine factors, including hepatocyte growth factor (HGF) and Wnt2. Notably, in in vitro co-cultures, VEGFR2-Id1 activation in LSECs stimulates hepatocyte proliferation. Indeed, intrasplenic transplantation of Id1(+/+) or Id1(-/-) LSECs transduced with Wnt2 and HGF (Id1(-/-)Wnt2(+)HGF(+) LSECs) re-establishes an inductive vascular niche in the liver sinusoids of the Id1(-/-) mice, initiating and restoring hepatovascular regeneration. Therefore, in the early phases of physiological liver regeneration, VEGFR2-Id1-mediated inductive angiogenesis in LSECs through release of angiocrine factors Wnt2 and HGF provokes hepatic proliferation. Subsequently, VEGFR2-Id1-dependent proliferative angiogenesis reconstitutes liver mass. Therapeutic co-transplantation of inductive VEGFR2(+)Id1(+)Wnt2(+)HGF(+) LSECs with hepatocytes provides an effective strategy to achieve durable liver regeneration.


Asunto(s)
Endotelio/metabolismo , Regeneración Hepática/fisiología , Hígado/irrigación sanguínea , Hígado/citología , Neovascularización Fisiológica/fisiología , Transducción de Señal , Animales , Proliferación Celular , Técnicas de Cocultivo , Endotelio/citología , Hepatectomía , Factor de Crecimiento de Hepatocito/metabolismo , Hepatocitos/citología , Proteína 1 Inhibidora de la Diferenciación/deficiencia , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Ratones , Fenotipo , Regulación hacia Arriba , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína wnt2/metabolismo
10.
Stem Cells ; 32(1): 177-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23963623

RESUMEN

The ability to generate and maintain stable in vitro cultures of mouse endothelial cells (ECs) has great potential for genetic dissection of the numerous pathologies involving vascular dysfunction as well as therapeutic applications. However, previous efforts at achieving sustained cultures of primary stable murine vascular cells have fallen short, and the cellular requirements for EC maintenance in vitro remain undefined. In this study, we have generated vascular ECs from mouse embryonic stem (ES) cells and show that active Akt is essential to their survival and propagation as homogeneous monolayers in vitro. These cells harbor the phenotypical, biochemical, and functional characteristics of ECs and expand throughout long-term cultures, while maintaining their angiogenic capacity. Moreover, Akt-transduced embryonic ECs form functional perfused vessels in vivo that anastomose with host blood vessels. We provide evidence for a novel function of Akt in stabilizing EC identity, whereby the activated form of the protein protects mouse ES cell-derived ECs from TGFß-mediated transdifferentiation by downregulating SMAD3. These findings identify a role for Akt in regulating the developmental potential of ES cell-derived ECs and demonstrate that active Akt maintains endothelial identity in embryonic ECs by interfering with active TGFß-mediated processes that would ordinarily usher these cells to alternate fates.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Células Endoteliales/citología , Ratones , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
11.
Circ Res ; 107(5): 667-76, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20634485

RESUMEN

RATIONALE: Stromal cell-derived factor (SDF)-1/CXCR4 axis has an instrumental role during cardiac development and has been shown to be a potential therapeutic target for optimizing ventricular remodeling after acute myocardial infarction (AMI) and in ischemic cardiomyopathy. Although a therapeutic target, the specific role of cardiac myocyte CXCR4 (CM-CXCR4) expression following cardiogenesis and survival of cardiac myocyte and left ventricular remodeling after AMI is unknown. OBJECTIVE: We hypothesized that cardiac myocyte derived CXCR4 is critical for cardiac development, but it may have no role in adulthood secondary to the short transient expression of SDF-1 and the delayed expression of CM-CXCR4 following AMI. To address this issue, we developed congenital and conditional CM-CXCR4(-/-) mouse models. METHODS AND RESULTS: Two strains of CM-CXCR4(flox/flox) mice were generated by crossing CXCR4(flox/flox) mice with MCM-Cre(+/-) mouse and MLC2v-Cre(+/-) mouse on the C57BL/6J background, yielding CXCR4(flox/flox) MCM-Cre(+/-) and CXCR4(flox/flox)MLC2v-Cre(+/-) mice. Studies demonstrated recombination in both models congenitally in the MLC2v-Cre(+/-) mice and following tamoxifen administration in the MCM-Cre(+/-) mice. Surprisingly the CXCR4(flox/flox)MLC2v-Cre(+/-) are viable, had normal cardiac function, and had no evidence of ventricular septal defect. CXCR4(flox/flox)MCM(+/-) treated with tamoxifen 2 weeks before AMI demonstrated 90% decrease in cardiac CXCR4 expression 48 hours after AMI. Twenty-one days post AMI, echocardiography revealed no statistically significant difference in the wall thickness, left ventricular dimensions or ejection fraction (40.9+/-7.5 versus 34.4+/-2.6%) in CXCR4(flox/flox) mice versus CM-CXCR4(-/-) mice regardless of strategy of Cre expression. No differences in vascular density (2369+/-131 versus 2471+/-126 vessels/mm(2); CXCR4(flox/flox) versus CM-CXCR4(-/-) mouse), infarct size, collagen content, or noninfarct zone cardiac myocyte size were observed 21 days after AMI. CONCLUSIONS: We conclude that cardiac myocyte-derived CXCR4 is not essential for cardiac development and, potentially because of the mismatch in timings of peaks of SDF-1 and CXCR4, has no major role in ventricular remodeling after AMI.


Asunto(s)
Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Receptores CXCR4/metabolismo , Remodelación Ventricular , Animales , Miosinas Cardíacas/genética , Movimiento Celular , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Integrasas/genética , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Cadenas Ligeras de Miosina/genética , ARN Mensajero/metabolismo , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Factores de Tiempo , Transfección , Función Ventricular Izquierda
12.
Biomolecules ; 12(12)2022 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-36551265

RESUMEN

Endothelial cells in vivo are subjected to a wide array of mechanical stimuli, such as cyclic stretch. Notably, a 10% stretch is associated with an atheroprotective endothelial phenotype, while a 20% stretch is associated with an atheroprone endothelial phenotype. Here, a systems biology-based approach is used to present a comprehensive overview of the functional responses and molecular regulatory networks that characterize the transition from an atheroprotective to an atheroprone phenotype in response to cyclic stretch. Using primary human umbilical vein endothelial cells (HUVECs), we determined the role of the equibiaxial cyclic stretch in vitro, with changes to the radius of the magnitudes of 10% and 20%, which are representative of physiological and pathological strain, respectively. Following the transcriptome analysis of next-generation sequencing data, we identified four key endothelial responses to pathological cyclic stretch: cell cycle regulation, inflammatory response, fatty acid metabolism, and mTOR signaling, driven by a regulatory network of eight transcription factors. Our study highlights the dynamic regulation of several key stretch-sensitive endothelial functions relevant to the induction of an atheroprone versus an atheroprotective phenotype and lays the foundation for further investigation into the mechanisms governing vascular pathology. This study has significant implications for the development of treatment modalities for vascular disease.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana , Mecanotransducción Celular , Estrés Mecánico , Humanos , Células Cultivadas , Biología de Sistemas , Factores de Transcripción/metabolismo
13.
Cell Stem Cell ; 29(4): 593-609.e7, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35364013

RESUMEN

The liver vascular network is patterned by sinusoidal and hepatocyte co-zonation. How intra-liver vessels acquire their hierarchical specialized functions is unknown. We study heterogeneity of hepatic vascular cells during mouse development through functional and single-cell RNA-sequencing. The acquisition of sinusoidal endothelial cell identity is initiated during early development and completed postnatally, originating from a pool of undifferentiated vascular progenitors at E12. The peri-natal induction of the transcription factor c-Maf is a critical switch for the sinusoidal identity determination. Endothelium-restricted deletion of c-Maf disrupts liver sinusoidal development, aberrantly expands postnatal liver hematopoiesis, promotes excessive postnatal sinusoidal proliferation, and aggravates liver pro-fibrotic sensitivity to chemical insult. Enforced c-Maf overexpression in generic human endothelial cells switches on a liver sinusoidal transcriptional program that maintains hepatocyte function. c-Maf represents an inducible intra-organotypic and niche-responsive molecular determinant of hepatic sinusoidal cell identity and lays the foundation for the strategies for vasculature-driven liver repair.


Asunto(s)
Capilares , Células Endoteliales , Animales , Endotelio , Hígado/patología , Cirrosis Hepática/patología , Regeneración Hepática , Ratones , Proteínas Proto-Oncogénicas c-maf
14.
Wound Repair Regen ; 19(3): 420-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21518091

RESUMEN

Topical treatment of superficial wounds has many advantages including decreased cost and increased ease of application compared with systemic treatments. Many of the advantages, however, are lost when it is necessary for repeated doses of topical medications to be given over an extended period of time. Therefore, a drug-delivery vehicle that delivers biologically appropriate doses in a sustained fashion would prove valuable. In this study, an alginate hydrogel scaffold impregnated with the angiogenic chemokine stromal-derived factor-1 was used to provide targeted, though short-term, delivery directly into the wound bed. Wounds were created on the dorsum of mice, and either a stromal-derived factor-1-impregnated or a saline-impregnated scaffold was applied. Wounds were explanted after 1, 3, 7 days, wound area was measured, and histology and immunohistochemistry for endothelial markers were performed. The remaining wound area in stromal-derived factor-1-treated wounds vs. controls was not significant 1 day after wounding (96.7 ± 0.1 vs. 97.5 ± 1.1%, p=0.317), but was significant after 3 days postwounding (46.7 ± 0.1 vs. 82.3 ± 2.4%, p=0.046) and 7 days postwounding (2.3 ± 1.3 vs. 32.0 ± 4.0%, p=0.049). Immunohistochemistry revealed a greater degree of endothelial cell invasion into the wound bed infiltration compared with controls. The results of this study suggest significant clinical promise for our hydrogel-delivery vehicle in the treatment of wounds.


Asunto(s)
Ingeniería de Tejidos/métodos , Cicatrización de Heridas/fisiología , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Técnicas de Cultivo de Célula , Quimiocina CXCL12 , Sistemas de Liberación de Medicamentos , Hidrogel de Polietilenoglicol-Dimetacrilato , Masculino , Ratones , Ratones Endogámicos C57BL , Andamios del Tejido , Factor de von Willebrand/metabolismo
15.
Int J Comput Assist Radiol Surg ; 16(10): 1761-1773, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34424457

RESUMEN

PURPOSE: Robotic systems have the potential to overcome inherent limitations of humans and offer substantial advantages to patients including reduction in surgery time. Our group has undertaken the challenge of developing autonomous wound closure system. One of the initial steps is to allow accurate assessment of wound skin topology and wound edge location. We present a vision-laser scanner to generate 3D point cloud for 3D reconstruction of wound's edge and topology. METHODS: When the laser range sensor measures Z coordinate, two encoders installed on the actuators of the gantry robot provide the precision values of X, Y coordinates simultaneously. The 3D point cloud of the wound skin is generated by recordings of X, Y and Z during scanning is performed over wound skin surface. To reduce the scanning time, we exploit a supplementary laser LED to project a regular laser spot on the wound skin surface, which can provide an additional measurement point by incorporating artificial neural network estimation approach. In the meantime, the point cloud of the wound edge can be extracted by detecting if the laser spot is located on the wound edge in the image from 2D camera. RESULTS: The mean absolute error (MAE) and standard deviation (σ) of wound edge are measured in MeshLab environment. The MAE (σ) in X (tangent), Y (tangent), and Z (normal) are 0.32 (0.22) mm, 0.37 (0.34) mm, and 0.61 (0.29) mm, respectively. The experimental results demonstrate that the vision-laser scanner attains high accuracy in determining wound edge location along the tangent of the wound skin. CONCLUSION: A vision-laser scanner is developed for 3D reconstruction of wound's edge and topology. The experimental tests on the different wound models revealed the effectiveness of the vision-laser scanner. The proposed scanner can generate 3D point cloud of the wound skin and its edge simultaneously, and thus significantly improve the accuracy of wound closure in clinical applications.


Asunto(s)
Imagenología Tridimensional , Rayos Láser , Humanos , Luz , Redes Neurales de la Computación
16.
Nat Commun ; 10(1): 5705, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31836710

RESUMEN

Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders.


Asunto(s)
Capilares/crecimiento & desarrollo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glomérulos Renales/irrigación sanguínea , Animales , Capilares/citología , Capilares/metabolismo , Células Cultivadas , Embrión de Mamíferos , Endotelio Vascular/citología , Endotelio Vascular/crecimiento & desarrollo , Factor de Transcripción GATA5/genética , Factor de Transcripción GATA5/metabolismo , Perfilación de la Expresión Génica , Humanos , Glomérulos Renales/crecimiento & desarrollo , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo , Cultivo Primario de Células , RNA-Seq , Análisis de la Célula Individual , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
17.
J Clin Invest ; 127(11): 4163-4178, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29035282

RESUMEN

Age-related changes in the hematopoietic compartment are primarily attributed to cell-intrinsic alterations in hematopoietic stem cells (HSCs); however, the contribution of the aged microenvironment has not been adequately evaluated. Understanding the role of the bone marrow (BM) microenvironment in supporting HSC function may prove to be beneficial in treating age-related functional hematopoietic decline. Here, we determined that aging of endothelial cells (ECs), a critical component of the BM microenvironment, was sufficient to drive hematopoietic aging phenotypes in young HSCs. We used an ex vivo hematopoietic stem and progenitor cell/EC (HSPC/EC) coculture system as well as in vivo EC infusions following myelosuppressive injury in mice to demonstrate that aged ECs impair the repopulating activity of young HSCs and impart a myeloid bias. Conversely, young ECs restored the repopulating capacity of aged HSCs but were unable to reverse the intrinsic myeloid bias. Infusion of young, HSC-supportive BM ECs enhanced hematopoietic recovery following myelosuppressive injury and restored endogenous HSC function in aged mice. Coinfusion of young ECs augmented aged HSC engraftment and enhanced overall survival in lethally irradiated mice by mitigating damage to the BM vascular microenvironment. These data lay the groundwork for the exploration of EC therapies that can serve as adjuvant modalities to enhance HSC engraftment and accelerate hematopoietic recovery in the elderly population following myelosuppressive regimens.


Asunto(s)
Células Endoteliales/fisiología , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Envejecimiento , Animales , Médula Ósea/irrigación sanguínea , Trasplante de Médula Ósea , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/trasplante , Ratones Endogámicos C57BL , Microvasos/patología , Traumatismos Experimentales por Radiación/prevención & control , Tolerancia a Radiación
18.
J Clin Invest ; 127(12): 4242-4256, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29058691

RESUMEN

Angiocrine factors, such as Notch ligands, supplied by the specialized endothelial cells (ECs) within the bone marrow and splenic vascular niche play an essential role in modulating the physiology of adult hematopoietic stem and progenitor cells (HSPCs). However, the relative contribution of various Notch ligands, specifically jagged-2, to the homeostasis of HSPCs is unknown. Here, we show that under steady state, jagged-2 is differentially expressed in tissue-specific vascular beds, but its expression is induced in hematopoietic vascular niches after myelosuppressive injury. We used mice with EC-specific deletion of the gene encoding jagged-2 (Jag2) to demonstrate that while EC-derived jagged-2 was dispensable for maintaining the capacity of HSPCs to repopulate under steady-state conditions, by activating Notch2 it did contribute to the recovery of HSPCs in response to myelosuppressive conditions. Engraftment and/or expansion of HSPCs was dependent on the expression of endothelial-derived jagged-2 following myeloablation. Additionally, jagged-2 expressed in bone marrow ECs regulated HSPC cell cycle and quiescence during regeneration. Endothelial-deployed jagged-2 triggered Notch2/Hey1, while tempering Notch2/Hes1 signaling in HSPCs. Collectively, these data demonstrate that EC-derived jagged-2 activates Notch2 signaling in HSPCs to promote hematopoietic recovery and has potential as a therapeutic target to accelerate balanced hematopoietic reconstitution after myelosuppression.


Asunto(s)
Células Madre Adultas/metabolismo , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Proteína Jagged-2/biosíntesis , Transducción de Señal , Aloinjertos , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Eliminación de Gen , Proteína Jagged-2/genética , Ratones , Ratones Transgénicos , Receptor Notch2/genética , Receptor Notch2/metabolismo , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/metabolismo
19.
Nat Commun ; 8: 15374, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28524846

RESUMEN

The outer blood-retina barrier is established through the coordinated terminal maturation of the retinal pigment epithelium (RPE), fenestrated choroid endothelial cells (ECs) and Bruch's membrane, a highly organized basement membrane that lies between both cell types. Here we study the contribution of choroid ECs to this process by comparing their gene expression profile before (P5) and after (P30) the critical postnatal period when mice acquire mature visual function. Transcriptome analyses show that expression of extracellular matrix-related genes changes dramatically over this period. Co-culture experiments support the existence of a novel regulatory pathway: ECs secrete factors that remodel RPE basement membrane, and integrin receptors sense these changes triggering Rho GTPase signals that modulate RPE tight junctions and enhance RPE barrier function. We anticipate our results will spawn a search for additional roles of choroid ECs in RPE physiology and disease.


Asunto(s)
Membrana Basal/metabolismo , Lámina Basal de la Coroides/metabolismo , Matriz Extracelular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Uniones Estrechas/metabolismo , Animales , Biotinilación , Barrera Hematorretinal/metabolismo , Adhesión Celular , Supervivencia Celular , Células Cultivadas , Coroides/metabolismo , Técnicas de Cocultivo , Electrorretinografía , Femenino , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Permeabilidad , Proteína-Lisina 6-Oxidasa/metabolismo , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN
20.
Circulation ; 111(9): 1175-83, 2005 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-15753226

RESUMEN

BACKGROUND: CD133 (AC133) is a surface antigen that defines a broad population of stem cells, including myogenic and endothelial progenitors. CD133+ cells are rare in adult tissues, and the factors that support their differentiation into mature angiomyogenic cells are not known. These hurdles have hampered the use of CD133+ cells for therapeutic purposes. Because human fetal liver is a rich source of CD133+ cells, we sought to identify the growth factors that promote codifferentiation of these cells into angiogenic and myogenic cells. METHODS AND RESULTS: Human fetal liver CD133+ and CD133- cell subpopulations were cultured with 5'-azacytidine or vascular endothelial growth factor (VEGF165) and/or brain-derived nerve growth factor (BDNF). CD133+ but not CD133- cells from human fetal liver codifferentiated into spindle-shaped cells, as well as flat adherent multinucleated cells capable of spontaneous contractions in culture. The resulting spindle-shaped cells were confirmed to be endothelial cells by immunohistochemistry analysis for von Willebrand factor and by acetylated LDL uptake. Multinucleated cells were characterized as striated muscles by electron microscopy and immunohistochemistry analysis for myosin heavy chain. Presence of VEGF165 and BDNF significantly enhanced angiomyogenesis in vitro. Inoculation of cells derived from CD133+ cells, but not CD133- cells, into the ear pinna of NOD/SCID mice resulted in the formation of cardiomyocytes, as identified by immunostaining with cardiac troponin-T antibody. These cells generated electrical action potentials, detectable by ECG tracing. CONCLUSIONS: CD133 defines a population of human fetal liver cells capable of differentiating into both angiogenic and myogenic cells. Preconditioning of these CD133+ cells with VEGF165 and BDNF enhances the angiomyogenesis. CD133+ fetal liver cells ultimately may be used for therapeutic angiomyogenesis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Endoteliales/citología , Endotelio Vascular/citología , Hígado/citología , Miocitos Cardíacos/citología , Células Madre/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Antígeno AC133 , Potenciales de Acción , Animales , Antígenos CD/análisis , Azacitidina/farmacología , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Oído Externo , Células Endoteliales/química , Perfilación de la Expresión Génica , Glicoproteínas/análisis , Humanos , Lipoproteínas LDL/metabolismo , Hígado/embriología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Miocitos Cardíacos/fisiología , Péptidos/análisis , Receptores Inmunológicos/metabolismo , Receptores Depuradores , Trasplante de Células Madre , Células Madre/citología , Trasplante Heterólogo , Factor de von Willebrand/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA