Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.594
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(11): 2345-2360.e16, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37167971

RESUMEN

A functional network of blood vessels is essential for organ growth and homeostasis, yet how the vasculature matures and maintains homeostasis remains elusive in live mice. By longitudinally tracking the same neonatal endothelial cells (ECs) over days to weeks, we found that capillary plexus expansion is driven by vessel regression to optimize network perfusion. Neonatal ECs rearrange positions to evenly distribute throughout the developing plexus and become positionally stable in adulthood. Upon local ablation, adult ECs survive through a plasmalemmal self-repair response, while neonatal ECs are predisposed to die. Furthermore, adult ECs reactivate migration to assist vessel repair. Global ablation reveals coordinated maintenance of the adult vascular architecture that allows for eventual network recovery. Lastly, neonatal remodeling and adult maintenance of the skin vascular plexus are orchestrated by temporally restricted, neonatal VEGFR2 signaling. Our work sheds light on fundamental mechanisms that underlie both vascular maturation and adult homeostasis in vivo.


Asunto(s)
Células Endoteliales , Neovascularización Fisiológica , Animales , Ratones , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Piel , Membrana Celular
2.
Cell ; 173(3): 762-775.e16, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29677517

RESUMEN

Mechanotransduction plays a crucial role in vascular biology. One example of this is the local regulation of vascular resistance via flow-mediated dilation (FMD). Impairment of this process is a hallmark of endothelial dysfunction and a precursor to a wide array of vascular diseases, such as hypertension and atherosclerosis. Yet the molecules responsible for sensing flow (shear stress) within endothelial cells remain largely unknown. We designed a 384-well screening system that applies shear stress on cultured cells. We identified a mechanosensitive cell line that exhibits shear stress-activated calcium transients, screened a focused RNAi library, and identified GPR68 as necessary and sufficient for shear stress responses. GPR68 is expressed in endothelial cells of small-diameter (resistance) arteries. Importantly, Gpr68-deficient mice display markedly impaired acute FMD and chronic flow-mediated outward remodeling in mesenteric arterioles. Therefore, GPR68 is an essential flow sensor in arteriolar endothelium and is a critical signaling component in cardiovascular pathophysiology.


Asunto(s)
Mecanotransducción Celular , Interferencia de ARN , Receptores Acoplados a Proteínas G/fisiología , Animales , Materiales Biocompatibles , Calcio/metabolismo , Línea Celular Tumoral , Células Endoteliales/fisiología , Endotelio Vascular/citología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Concentración de Iones de Hidrógeno , Arterias Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/genética , Resistencia al Corte , Estrés Mecánico , Resistencia Vascular
3.
Annu Rev Cell Dev Biol ; 35: 591-613, 2019 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-31299172

RESUMEN

The vertebrate vasculature displays high organotypic specialization, with the structure and function of blood vessels catering to the specific needs of each tissue. A unique feature of the central nervous system (CNS) vasculature is the blood-brain barrier (BBB). The BBB regulates substance influx and efflux to maintain a homeostatic environment for proper brain function. Here, we review the development and cell biology of the BBB, focusing on the cellular and molecular regulation of barrier formation and the maintenance of the BBB through adulthood. We summarize unique features of CNS endothelial cells and highlight recent progress in and general principles of barrier regulation. Finally, we illustrate why a mechanistic understanding of the development and maintenance of the BBB could provide novel therapeutic opportunities for CNS drug delivery.


Asunto(s)
Transporte Biológico/fisiología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/crecimiento & desarrollo , Sistema Nervioso Central/citología , Células Endoteliales/citología , Animales , Astrocitos/citología , Membrana Basal/citología , Membrana Basal/metabolismo , Transporte Biológico/genética , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/fisiología , Sistema Nervioso Central/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Homeostasis , Humanos , Leucocitos , Acoplamiento Neurovascular/fisiología , Pericitos/citología , Uniones Estrechas , Transcitosis/fisiología , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología
4.
Cell ; 167(1): 275-284.e6, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27662093

RESUMEN

The VEGF-A isoforms play a crucial role in vascular development, and the VEGF signaling pathway is a clinically validated therapeutic target for several pathological conditions. Alternative mRNA splicing leads to the generation of multiple VEGF-A isoforms, including VEGF165. A recent study reported the presence of another isoform, VEGF-Ax, arising from programmed readthrough translation. Compared to VEGF165, VEGF-Ax has a 22-amino-acid extension in the COOH terminus and has been reported to function as a negative regulator of VEGF signaling in endothelial cells, with potent anti-angiogenic effects. Here, we show that, contrary to the earlier report, VEGF-Ax stimulates endothelial cell mitogenesis, angiogenesis, as well as vascular permeability. Accordingly, VEGF-Ax induces phosphorylation of key tyrosine residues in VEGFR-2. Notably, VEGF-Ax was less potent than VEGF165, consistent with its impaired binding to the VEGF co-receptor neuropilin-1.


Asunto(s)
Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular , Empalme Alternativo , Secuencia de Aminoácidos , Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Permeabilidad Capilar/genética , Permeabilidad Capilar/fisiología , Quimiotaxis/efectos de los fármacos , Clonación Molecular , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Cobayas , Células HEK293 , Humanos , Ratones , Mitógenos/farmacología , Mitosis/efectos de los fármacos , Mitosis/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Neuropilina-1/metabolismo , Biosíntesis de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Tirosina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Immunity ; 54(9): 1989-2004.e9, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34363750

RESUMEN

The migration of neutrophils from the blood circulation to sites of infection or injury is a key immune response and requires the breaching of endothelial cells (ECs) that line the inner aspect of blood vessels. Unregulated neutrophil transendothelial cell migration (TEM) is pathogenic, but the molecular basis of its physiological termination remains unknown. Here, we demonstrated that ECs of venules in inflamed tissues exhibited a robust autophagic response that was aligned temporally with the peak of neutrophil trafficking and was strictly localized to EC contacts. Genetic ablation of EC autophagy led to excessive neutrophil TEM and uncontrolled leukocyte migration in murine inflammatory models, while pharmacological induction of autophagy suppressed neutrophil infiltration into tissues. Mechanistically, autophagy regulated the remodeling of EC junctions and expression of key EC adhesion molecules, facilitating their intracellular trafficking and degradation. Collectively, we have identified autophagy as a modulator of EC leukocyte trafficking machinery aimed at terminating physiological inflammation.


Asunto(s)
Autofagia/fisiología , Células Endoteliales/fisiología , Infiltración Neutrófila/fisiología , Migración Transendotelial y Transepitelial/fisiología , Animales , Quimiotaxis de Leucocito/fisiología , Células Endoteliales/patología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Uniones Intercelulares/fisiología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología
6.
Nat Rev Mol Cell Biol ; 18(8): 477-494, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28537573

RESUMEN

Blood and lymphatic vessels pervade almost all body tissues and have numerous essential roles in physiology and disease. The inner lining of these networks is formed by a single layer of endothelial cells, which is specialized according to the needs of the tissue that it supplies. Whereas the general mechanisms of blood and lymphatic vessel development are being defined with increasing molecular precision, studies of the processes of endothelial specialization remain mostly descriptive. Recent insights from genetic animal models illuminate how endothelial cells interact with each other and with their tissue environment, providing paradigms for vessel type- and organ-specific endothelial differentiation. Delineating these governing principles will be crucial for understanding how tissues develop and maintain, and how their function becomes abnormal in disease.


Asunto(s)
Células Endoteliales/citología , Animales , Vasos Sanguíneos/citología , Diferenciación Celular/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Vasos Linfáticos/citología
7.
EMBO J ; 43(8): 1519-1544, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38528180

RESUMEN

Pericytes and endothelial cells (ECs) constitute the fundamental components of blood vessels. While the role of ECs in tumor angiogenesis and the tumor microenvironment is well appreciated, pericyte function in tumors remains underexplored. In this study, we used pericyte-specific deletion of the nitric oxide (NO) receptor, soluble guanylate cyclase (sGC), to investigate via single-cell RNA sequencing how pericytes influence the vascular niche and the tumor microenvironment. Our findings demonstrate that pericyte sGC deletion disrupts EC-pericyte interactions, impairing Notch-mediated intercellular communication and triggering extensive transcriptomic reprogramming in both pericytes and ECs. These changes further extended their influence to neighboring cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) through paracrine signaling, collectively suppressing tumor growth. Inhibition of pericyte sGC has minimal impact on quiescent vessels but significantly increases the vulnerability of angiogenic tumor vessels to conventional anti-angiogenic therapy. In conclusion, our findings elucidate the role of pericytes in shaping the tumor vascular niche and tumor microenvironment and support pericyte sGC targeting as a promising strategy for improving anti-angiogenic therapy for cancer treatment.


Asunto(s)
Neoplasias , Pericitos , Humanos , Pericitos/patología , Pericitos/fisiología , Guanilil Ciclasa Soluble , Células Endoteliales/fisiología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neoplasias/genética , Neoplasias/patología , Guanilato Ciclasa , Microambiente Tumoral
8.
Immunity ; 51(4): 638-654.e9, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31561945

RESUMEN

Macrophages are strongly adapted to their tissue of residence. Yet, little is known about the cell-cell interactions that imprint the tissue-specific identities of macrophages in their respective niches. Using conditional depletion of liver Kupffer cells, we traced the developmental stages of monocytes differentiating into Kupffer cells and mapped the cellular interactions imprinting the Kupffer cell identity. Kupffer cell loss induced tumor necrosis factor (TNF)- and interleukin-1 (IL-1) receptor-dependent activation of stellate cells and endothelial cells, resulting in the transient production of chemokines and adhesion molecules orchestrating monocyte engraftment. Engrafted circulating monocytes transmigrated into the perisinusoidal space and acquired the liver-associated transcription factors inhibitor of DNA 3 (ID3) and liver X receptor-α (LXR-α). Coordinated interactions with hepatocytes induced ID3 expression, whereas endothelial cells and stellate cells induced LXR-α via a synergistic NOTCH-BMP pathway. This study shows that the Kupffer cell niche is composed of stellate cells, hepatocytes, and endothelial cells that together imprint the liver-specific macrophage identity.


Asunto(s)
Células Endoteliales/fisiología , Células Estrelladas Hepáticas/fisiología , Hepatocitos/fisiología , Macrófagos del Hígado/fisiología , Hígado/citología , Macrófagos/fisiología , Monocitos/fisiología , Animales , Comunicación Celular , Diferenciación Celular , Células Cultivadas , Microambiente Celular , Femenino , Regulación de la Expresión Génica , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Notch/metabolismo
9.
Physiol Rev ; 99(3): 1467-1525, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31140373

RESUMEN

A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.


Asunto(s)
Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Transducción de Señal/fisiología , Animales , Barrera Alveolocapilar/fisiología , Barrera Hematoencefálica/fisiología , Endotelio Vascular/citología , Humanos
10.
Immunity ; 47(1): 80-92.e4, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28709801

RESUMEN

Lymph nodes (LNs) are strategically situated throughout the body at junctures of the blood vascular and lymphatic systems to direct immune responses against antigens draining from peripheral tissues. The current paradigm describes LN development as a programmed process that is governed through the interaction between mesenchymal lymphoid tissue organizer (LTo) cells and hematopoietic lymphoid tissue inducer (LTi) cells. Using cell-type-specific ablation of key molecules involved in lymphoid organogenesis, we found that initiation of LN development is dependent on LTi-cell-mediated activation of lymphatic endothelial cells (LECs) and that engagement of mesenchymal stromal cells is a succeeding event. LEC activation was mediated mainly by signaling through receptor activator of NF-κB (RANK) and the non-canonical NF-κB pathway and was steered by sphingosine-1-phosphate-receptor-dependent retention of LTi cells in the LN anlage. Finally, the finding that pharmacologically enforced interaction between LTi cells and LECs promotes ectopic LN formation underscores the central LTo function of LECs.


Asunto(s)
Células Endoteliales/fisiología , Ganglios Linfáticos/fisiología , Células Madre Mesenquimatosas/fisiología , Organogénesis , Animales , Diferenciación Celular , Células Cultivadas , Coristoma , Embrión de Mamíferos , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal
11.
Cell ; 144(3): 402-13, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21295700

RESUMEN

The functions of caveolae, the characteristic plasma membrane invaginations, remain debated. Their abundance in cells experiencing mechanical stress led us to investigate their role in membrane-mediated mechanical response. Acute mechanical stress induced by osmotic swelling or by uniaxial stretching results in a rapid disappearance of caveolae, in a reduced caveolin/Cavin1 interaction, and in an increase of free caveolins at the plasma membrane. Tether-pulling force measurements in cells and in plasma membrane spheres demonstrate that caveola flattening and disassembly is the primary actin- and ATP-independent cell response that buffers membrane tension surges during mechanical stress. Conversely, stress release leads to complete caveola reassembly in an actin- and ATP-dependent process. The absence of a functional caveola reservoir in myotubes from muscular dystrophic patients enhanced membrane fragility under mechanical stress. Our findings support a new role for caveolae as a physiological membrane reservoir that quickly accommodates sudden and acute mechanical stresses.


Asunto(s)
Caveolas/fisiología , Células Endoteliales/citología , Células Musculares/fisiología , Actinas/fisiología , Adenosina Trifosfato/fisiología , Animales , Caveolas/ultraestructura , Línea Celular , Células Endoteliales/fisiología , Humanos , Ratones , Células Musculares/citología , Estrés Mecánico
12.
Cell ; 146(6): 873-87, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21925313

RESUMEN

Blood vessels form extensive networks that nurture all tissues in the body. Abnormal vessel growth and function are hallmarks of cancer and ischemic and inflammatory diseases, and they contribute to disease progression. Therapeutic approaches to block vascular supply have reached the clinic, but limited efficacy and resistance pose unresolved challenges. Recent insights establish how endothelial cells communicate with each other and with their environment to form a branched vascular network. The emerging principles of vascular growth provide exciting new perspectives, the translation of which might overcome the current limitations of pro- and antiangiogenic medicine.


Asunto(s)
Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Humanos , Inflamación/tratamiento farmacológico
13.
Immunol Rev ; 311(1): 39-49, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35909222

RESUMEN

The blood-brain barrier (BBB) is a selectively permeable barrier separating the periphery from the central nervous system (CNS). The BBB restricts the flow of most material into and out of the CNS, including many drugs that could be used as potent therapies. BBB permeability is modulated by several cells that are collectively called the neurovascular unit (NVU). The NVU consists of specialized CNS endothelial cells (ECs), pericytes, astrocytes, microglia, and neurons. CNS ECs maintain a complex "seal" via tight junctions, forming the BBB; breakdown of these tight junctions leads to BBB disruption. Pericytes control the vascular flow within capillaries and help maintain the basal lamina. Astrocytes control much of the flow of material that has moved beyond the CNS EC layer and can form a secondary barrier under inflammatory conditions. Microglia survey the border of the NVU for noxious material. Neuronal activity also plays a role in the maintenance of the BBB. Since astrocytes, pericytes, microglia, and neurons are all able to modulate the permeability of the BBB, understating the complex contributions of each member of the NVU will potentially uncover novel and effective methods for delivery of neurotherapies to the CNS.


Asunto(s)
Células Endoteliales , Pericitos , Astrocitos/metabolismo , Barrera Hematoencefálica/fisiología , Sistema Nervioso Central , Células Endoteliales/fisiología , Humanos , Pericitos/metabolismo
14.
Physiol Rev ; 98(4): 2025-2061, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30067156

RESUMEN

The vasculature is a remarkably interesting, complex, and interconnected organ. It provides a conduit for oxygen and nutrients, filtration of waste products, and rapid communication between organs. Much remains to be learned about the specialized vascular beds that fulfill these diverse, yet vital functions. This review was prompted by the discovery that Notch signaling in mouse endothelial cells is crucial for the development of specialized vascular beds found in the heart, kidneys, liver, intestines, and bone. We will address the intriguing questions raised by the role of Notch signaling and that of its regulator, the metalloprotease ADAM10, in the development of specialized vascular beds. We will cover fundamentals of ADAM10/Notch signaling, the concept of Notch-dependent cell fate decisions, and how these might govern the development of organ-specific vascular beds through angiogenesis or vasculogenesis. We will also consider common features of the affected vessels, including the presence of fenestra or sinusoids and their occurrence in portal systems with two consecutive capillary beds. We hope to stimulate further discussion and study of the role of ADAM10/Notch signaling in the development of specialized vascular structures, which might help uncover new targets for the repair of vascular beds damaged in conditions like coronary artery disease and glomerulonephritis.


Asunto(s)
Proteína ADAM10/metabolismo , Capilares/metabolismo , Capilares/fisiología , Células Endoteliales/metabolismo , Receptores Notch/metabolismo , Animales , Diferenciación Celular/fisiología , Células Endoteliales/fisiología , Humanos , Transducción de Señal/fisiología
15.
EMBO J ; 40(1): e105242, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33215738

RESUMEN

Age-associated alterations of the hormone-secreting endocrine system cause organ dysfunction and disease states. However, the cell biology of endocrine tissue ageing remains poorly understood. Here, we perform comparative 3D imaging to understand age-related perturbations of the endothelial cell (EC) compartment in endocrine glands. Datasets of a wide range of markers highlight a decline in capillary and artery numbers, but not of perivascular cells in pancreas, testis and thyroid gland, with age in mice and humans. Further, angiogenesis and ß-cell expansion in the pancreas are coupled by a distinct age-dependent subset of ECs. While this EC subpopulation supports pancreatic ß cells, it declines during ageing concomitant with increased expression of the gap junction protein Gja1. EC-specific ablation of Gja1 restores ß-cell expansion in the aged pancreas. These results provide a proof of concept for understanding age-related vascular changes and imply that therapeutic targeting of blood vessels may restore aged endocrine tissue function. This comprehensive data atlas offers over > 1,000 multicolour volumes for exploration and research in endocrinology, ageing, matrix and vascular biology.


Asunto(s)
Envejecimiento/fisiología , Sistema Endocrino/fisiología , Células Endoteliales/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Vasos Sanguíneos , Glándulas Endocrinas/fisiología , Femenino , Humanos , Imagenología Tridimensional/métodos , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/patología , Páncreas/fisiología , Testículo/fisiología , Glándula Tiroides/fisiología , Adulto Joven
16.
Development ; 149(18)2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36134690

RESUMEN

Heart regeneration requires multiple cell types to enable cardiomyocyte (CM) proliferation. How these cells interact to create growth niches is unclear. Here, we profile proliferation kinetics of cardiac endothelial cells (CECs) and CMs in the neonatal mouse heart and find that they are spatiotemporally coupled. We show that coupled myovascular expansion during cardiac growth or regeneration is dependent upon VEGF-VEGFR2 signaling, as genetic deletion of Vegfr2 from CECs or inhibition of VEGFA abrogates both CEC and CM proliferation. Repair of cryoinjury displays poor spatial coupling of CEC and CM proliferation. Boosting CEC density after cryoinjury with virus encoding Vegfa enhances regeneration. Using Mendelian randomization, we demonstrate that circulating VEGFA levels are positively linked with human myocardial mass, suggesting that Vegfa can stimulate human cardiac growth. Our work demonstrates the importance of coupled CEC and CM expansion and reveals a myovascular niche that may be therapeutically targeted for heart regeneration.


Asunto(s)
Células Endoteliales , Factor A de Crecimiento Endotelial Vascular , Animales , Proliferación Celular , Células Endoteliales/fisiología , Corazón/fisiología , Humanos , Recién Nacido , Ratones , Miocitos Cardíacos/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Nat Rev Neurosci ; 21(2): 93-102, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31913356

RESUMEN

A rapidly ageing population and a limited therapeutic toolbox urgently necessitate new approaches to treat neurodegenerative diseases. Brain ageing, the key risk factor for neurodegeneration, involves complex cellular and molecular processes that eventually result in cognitive decline. Although cell-intrinsic defects in neurons and glia may partially explain this decline, cell-extrinsic changes in the systemic environment, mediated by blood, have recently been shown to contribute to brain dysfunction with age. Here, we review the current understanding of how systemic factors mediate brain ageing, how these factors are regulated and how we can translate these findings into therapies for neurodegenerative diseases.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/fisiología , Homeostasis , Enfermedades Neurodegenerativas/fisiopatología , Envejecimiento/inmunología , Animales , Encéfalo/inmunología , Células Endoteliales/inmunología , Células Endoteliales/fisiología , Ejercicio Físico/fisiología , Humanos , Microbiota/inmunología , Microbiota/fisiología , Enfermedades Neurodegenerativas/inmunología , Neuroglía/inmunología , Neuroglía/fisiología , Neuronas/inmunología , Neuronas/fisiología
18.
Immunity ; 45(4): 877-888, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760341

RESUMEN

Lymph node (LN) expansion during an immune response relies on the transient remodeling of its vasculature. Although the mechanisms driving LN endothelial cell division are beginning to be understood, a comprehensive view of LN endothelial cell dynamics at the single-cell level is lacking. Here, we used multicolored fluorescent fate-mapping models to track the behavior of blood endothelial cells during LN expansion upon inflammation and subsequent return to homeostasis. We found that expansion of the LN vasculature relied on the sequential assembly of endothelial cell proliferative units. This segmented growth was sustained by the clonal proliferation of high endothelial venule (HEV) cells, which act as local progenitors to create capillaries and HEV neo-vessels at the periphery of the LN. Return to homeostasis was accompanied by the stochastic death of pre-existing and neo-synthesized LN endothelial cells. Thus, our fate-mapping studies unravel-at a single-cell level-the complex dynamics of vascular-tree remodeling during LN expansion and contraction.


Asunto(s)
Proliferación Celular/fisiología , Células Endoteliales/inmunología , Células Endoteliales/fisiología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/fisiología , Animales , Capilares/inmunología , Capilares/fisiología , Células Cultivadas , Homeostasis/inmunología , Homeostasis/fisiología , Inflamación/inmunología , Inflamación/patología , Ratones
19.
Cell ; 140(4): 460-76, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178740

RESUMEN

The growth of lymphatic vessels (lymphangiogenesis) is actively involved in a number of pathological processes including tissue inflammation and tumor dissemination but is insufficient in patients suffering from lymphedema, a debilitating condition characterized by chronic tissue edema and impaired immunity. The recent explosion of knowledge on the molecular mechanisms governing lymphangiogenesis provides new possibilities to treat these diseases.


Asunto(s)
Linfangiogénesis , Animales , Células Endoteliales/fisiología , Humanos , Inflamación/fisiopatología , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/fisiología , Vasos Linfáticos/fisiopatología , Neoplasias/fisiopatología
20.
Proc Natl Acad Sci U S A ; 119(18): e2118927119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35482920

RESUMEN

Every blood vessel is lined by a single layer of highly specialized, yet adaptable and multifunctional endothelial cells. These cells, the endothelium, control vascular contractility, hemostasis, and inflammation and regulate the exchange of oxygen, nutrients, and waste products between circulating blood and tissue. To control each function, the endothelium processes endlessly arriving requests from multiple sources using separate clusters of cells specialized to detect specific stimuli. A well-developed but poorly understood communication system operates between cells to integrate multiple lines of information and coordinate endothelial responses. Here, the nature of the communication network has been addressed using single-cell Ca2+ imaging across thousands of endothelial cells in intact blood vessels. Cell activities were cross-correlated and compared to a stochastic model to determine network connections. Highly correlated Ca2+ activities occurred in scattered cell clusters, and network communication links between them exhibited unexpectedly short path lengths. The number of connections between cells (degree distribution) followed a power-law relationship revealing a scale-free network topology. The path length and degree distribution revealed an endothelial network with a "small-world" configuration. The small-world configuration confers particularly dynamic endothelial properties including high signal-propagation speed, stability, and a high degree of synchronizability. Local activation of small clusters of cells revealed that the short path lengths and rapid signal transmission were achieved by shortcuts via connecting extensions to nonlocal cells. These findings reveal that the endothelial network design is effective for local and global efficiency in the interaction of the cells and rapid and robust communication between endothelial cells in order to efficiently control cardiovascular activity.


Asunto(s)
Células Endoteliales , Transducción de Señal , Células Endoteliales/fisiología , Endotelio , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA