Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.639
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 185(20): 3753-3769.e18, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36179668

RESUMEN

Interactions between angiogenesis and neurogenesis regulate embryonic brain development. However, a comprehensive understanding of the stages of vascular cell maturation is lacking, especially in the prenatal human brain. Using fluorescence-activated cell sorting, single-cell transcriptomics, and histological and ultrastructural analyses, we show that an ensemble of endothelial and mural cell subtypes tile the brain vasculature during the second trimester. These vascular cells follow distinct developmental trajectories and utilize diverse signaling mechanisms, including collagen, laminin, and midkine, to facilitate cell-cell communication and maturation. Interestingly, our results reveal that tip cells, a subtype of endothelial cells, are highly enriched near the ventricular zone, the site of active neurogenesis. Consistent with these observations, prenatal vascular cells transplanted into cortical organoids exhibit restricted lineage potential that favors tip cells, promotes neurogenesis, and reduces cellular stress. Together, our results uncover important mechanisms into vascular maturation during this critical period of human brain development.


Asunto(s)
Células Endoteliales , Neovascularización Fisiológica , Encéfalo , Colágeno , Humanos , Laminina , Midkina , Neovascularización Patológica/patología , Neovascularización Fisiológica/fisiología , Pericitos
2.
Cell ; 184(3): 810-826.e23, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33406409

RESUMEN

Development of the human intestine is not well understood. Here, we link single-cell RNA sequencing and spatial transcriptomics to characterize intestinal morphogenesis through time. We identify 101 cell states including epithelial and mesenchymal progenitor populations and programs linked to key morphogenetic milestones. We describe principles of crypt-villus axis formation; neural, vascular, mesenchymal morphogenesis, and immune population of the developing gut. We identify the differentiation hierarchies of developing fibroblast and myofibroblast subtypes and describe diverse functions for these including as vascular niche cells. We pinpoint the origins of Peyer's patches and gut-associated lymphoid tissue (GALT) and describe location-specific immune programs. We use our resource to present an unbiased analysis of morphogen gradients that direct sequential waves of cellular differentiation and define cells and locations linked to rare developmental intestinal disorders. We compile a publicly available online resource, spatio-temporal analysis resource of fetal intestinal development (STAR-FINDer), to facilitate further work.


Asunto(s)
Intestinos/citología , Intestinos/crecimiento & desarrollo , Análisis de la Célula Individual , Células Endoteliales/citología , Sistema Nervioso Entérico/citología , Feto/embriología , Fibroblastos/citología , Humanos , Inmunidad , Enfermedades Intestinales/congénito , Enfermedades Intestinales/patología , Mucosa Intestinal/crecimiento & desarrollo , Intestinos/irrigación sanguínea , Ligandos , Mesodermo/citología , Neovascularización Fisiológica , Pericitos/citología , Células Madre/citología , Factores de Tiempo , Factores de Transcripción/metabolismo
3.
Cell ; 173(1): 153-165.e22, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29502968

RESUMEN

CNS injury often severs axons. Scar tissue that forms locally at the lesion site is thought to block axonal regeneration, resulting in permanent functional deficits. We report that inhibiting the generation of progeny by a subclass of pericytes led to decreased fibrosis and extracellular matrix deposition after spinal cord injury in mice. Regeneration of raphespinal and corticospinal tract axons was enhanced and sensorimotor function recovery improved following spinal cord injury in animals with attenuated pericyte-derived scarring. Using optogenetic stimulation, we demonstrate that regenerated corticospinal tract axons integrated into the local spinal cord circuitry below the lesion site. The number of regenerated axons correlated with improved sensorimotor function recovery. In conclusion, attenuation of pericyte-derived fibrosis represents a promising therapeutic approach to facilitate recovery following CNS injury.


Asunto(s)
Cicatriz/patología , Traumatismos de la Médula Espinal/patología , Animales , Axones/fisiología , Axones/efectos de la radiación , Modelos Animales de Enfermedad , Potenciales Evocados/efectos de la radiación , Matriz Extracelular/metabolismo , Fibrosis , Luz , Ratones , Ratones Transgénicos , Pericitos/citología , Pericitos/metabolismo , Estimulación Luminosa , Tractos Piramidales/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Recuperación de la Función , Regeneración , Corteza Sensoriomotora/fisiología , Traumatismos de la Médula Espinal/fisiopatología
4.
Cell ; 175(2): 372-386.e17, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30270042

RESUMEN

Intestinal mesenchymal cells play essential roles in epithelial homeostasis, matrix remodeling, immunity, and inflammation. But the extent of heterogeneity within the colonic mesenchyme in these processes remains unknown. Using unbiased single-cell profiling of over 16,500 colonic mesenchymal cells, we reveal four subsets of fibroblasts expressing divergent transcriptional regulators and functional pathways, in addition to pericytes and myofibroblasts. We identified a niche population located in proximity to epithelial crypts expressing SOX6, F3 (CD142), and WNT genes essential for colonic epithelial stem cell function. In colitis, we observed dysregulation of this niche and emergence of an activated mesenchymal population. This subset expressed TNF superfamily member 14 (TNFSF14), fibroblastic reticular cell-associated genes, IL-33, and Lysyl oxidases. Further, it induced factors that impaired epithelial proliferation and maturation and contributed to oxidative stress and disease severity in vivo. Our work defines how the colonic mesenchyme remodels to fuel inflammation and barrier dysfunction in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/fisiopatología , Mesodermo/fisiología , Animales , Proliferación Celular , Colitis/genética , Colitis/fisiopatología , Colon/fisiología , Células Epiteliales/metabolismo , Fibroblastos/fisiología , Heterogeneidad Genética , Homeostasis , Humanos , Inflamación , Mucosa Intestinal/inmunología , Mucosa Intestinal/fisiología , Intestinos/inmunología , Intestinos/fisiología , Células Madre Mesenquimatosas/fisiología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Miofibroblastos , Pericitos , Células RAW 264.7 , Factores de Transcripción SOXD/fisiología , Análisis de la Célula Individual/métodos , Tromboplastina/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Vía de Señalización Wnt/fisiología
5.
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
6.
Cell ; 163(5): 1064-1078, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26590417

RESUMEN

Structural and functional brain connectivity, synaptic activity, and information processing require highly coordinated signal transduction between different cell types within the neurovascular unit and intact blood-brain barrier (BBB) functions. Here, we examine the mechanisms regulating the formation and maintenance of the BBB and functions of BBB-associated cell types. Furthermore, we discuss the growing evidence associating BBB breakdown with the pathogenesis of inherited monogenic neurological disorders and complex multifactorial diseases, including Alzheimer's disease.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Enfermedades del Sistema Nervioso/fisiopatología , Animales , Astrocitos/metabolismo , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/fisiología , Células Endoteliales/metabolismo , Humanos , Proteínas de Transporte de Membrana/metabolismo , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/patología , Pericitos/citología
7.
Nature ; 632(8024): 429-436, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38987599

RESUMEN

Tumours can obtain nutrients and oxygen required to progress and metastasize through the blood supply1. Inducing angiogenesis involves the sprouting of established vessel beds and their maturation into an organized network2,3. Here we generate a comprehensive atlas of tumour vasculature at single-cell resolution, encompassing approximately 200,000 cells from 372 donors representing 31 cancer types. Trajectory inference suggested that tumour angiogenesis was initiated from venous endothelial cells and extended towards arterial endothelial cells. As neovascularization elongates (through angiogenic stages SI, SII and SIII), APLN+ tip cells at the SI stage (APLN+ TipSI) advanced to TipSIII cells with increased Notch signalling. Meanwhile, stalk cells, following tip cells, transitioned from high chemokine expression to elevated TEK (also known as Tie2) expression. Moreover, APLN+ TipSI cells not only were associated with disease progression and poor prognosis but also hold promise for predicting response to anti-VEGF therapy. Lymphatic endothelial cells demonstrated two distinct differentiation lineages: one responsible for lymphangiogenesis and the other involved in antigen presentation. In pericytes, endoplasmic reticulum stress was associated with the proangiogenic BASP1+ matrix-producing pericytes. Furthermore, intercellular communication analysis showed that neovascular endothelial cells could shape an immunosuppressive microenvironment conducive to angiogenesis. This study depicts the complexity of tumour vasculature and has potential clinical significance for anti-angiogenic therapy.


Asunto(s)
Células Endoteliales , Neoplasias , Neovascularización Patológica , Análisis de la Célula Individual , Humanos , Presentación de Antígeno , Comunicación Celular , Diferenciación Celular , Linaje de la Célula , Progresión de la Enfermedad , Estrés del Retículo Endoplásmico , Células Endoteliales/citología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Linfangiogénesis , Neoplasias/irrigación sanguínea , Neoplasias/clasificación , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/patología , Pericitos/patología , Pericitos/citología , Pericitos/metabolismo , Pronóstico , Receptores Notch/metabolismo , Transducción de Señal , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Pez Cebra
8.
Nature ; 622(7983): 611-618, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37699522

RESUMEN

Clostridioides difficile infection (CDI) is a major cause of healthcare-associated gastrointestinal infections1,2. The exaggerated colonic inflammation caused by C. difficile toxins such as toxin B (TcdB) damages tissues and promotes C. difficile colonization3-6, but how TcdB causes inflammation is unclear. Here we report that TcdB induces neurogenic inflammation by targeting gut-innervating afferent neurons and pericytes through receptors, including the Frizzled receptors (FZD1, FZD2 and FZD7) in neurons and chondroitin sulfate proteoglycan 4 (CSPG4) in pericytes. TcdB stimulates the secretion of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) from neurons and pro-inflammatory cytokines from pericytes. Targeted delivery of the TcdB enzymatic domain, through fusion with a detoxified diphtheria toxin, into peptidergic sensory neurons that express exogeneous diphtheria toxin receptor (an approach we term toxogenetics) is sufficient to induce neurogenic inflammation and recapitulates major colonic histopathology associated with CDI. Conversely, mice lacking SP, CGRP or the SP receptor (neurokinin 1 receptor) show reduced pathology in both models of caecal TcdB injection and CDI. Blocking SP or CGRP signalling reduces tissue damage and C. difficile burden in mice infected with a standard C. difficile strain or with hypervirulent strains expressing the TcdB2 variant. Thus, targeting neurogenic inflammation provides a host-oriented therapeutic approach for treating CDI.


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Inflamación Neurogénica , Neuronas Aferentes , Pericitos , Animales , Ratones , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/farmacología , Péptido Relacionado con Gen de Calcitonina/antagonistas & inhibidores , Péptido Relacionado con Gen de Calcitonina/metabolismo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Inflamación Neurogénica/inducido químicamente , Inflamación Neurogénica/microbiología , Inflamación Neurogénica/patología , Pericitos/efectos de los fármacos , Pericitos/microbiología , Pericitos/patología , Receptores de Neuroquinina-1/metabolismo , Sustancia P/antagonistas & inhibidores , Sustancia P/metabolismo , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/microbiología , Neuronas Aferentes/patología , Mediadores de Inflamación/metabolismo , Ciego/efectos de los fármacos , Ciego/metabolismo , Transducción de Señal/efectos de los fármacos
9.
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
10.
Immunity ; 50(3): 645-654.e6, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30770250

RESUMEN

The epidermal growth factor receptor ligand Amphiregulin has a well-documented role in the restoration of tissue homeostasis after injury; however, the mechanism by which Amphiregulin contributes to wound repair remains unknown. Here we show that Amphiregulin functioned by releasing bioactive transforming growth factor beta (TGF-ß) from latent complexes via integrin-αV activation. Using acute injury models in two different tissues, we found that by inducing TGF-ß activation on mesenchymal stromal cells (pericytes), Amphiregulin induced their differentiation into myofibroblasts, thereby selectively contributing to the restoration of vascular barrier function within injured tissue. Furthermore, we identified macrophages as a critical source of Amphiregulin, revealing a direct effector mechanism by which these cells contribute to tissue restoration after acute injury. Combined, these observations expose a so far under-appreciated mechanism of how cells of the immune system selectively control the differentiation of tissue progenitor cells during tissue repair and inflammation.


Asunto(s)
Anfirregulina/metabolismo , Macrófagos/metabolismo , Pericitos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/fisiología , Femenino , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/metabolismo
11.
Cell ; 153(1): 139-52, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23540695

RESUMEN

Glioblastomas (GBMs) are highly vascular and lethal brain tumors that display cellular hierarchies containing self-renewing tumorigenic glioma stem cells (GSCs). Because GSCs often reside in perivascular niches and may undergo mesenchymal differentiation, we interrogated GSC potential to generate vascular pericytes. Here, we show that GSCs give rise to pericytes to support vessel function and tumor growth. In vivo cell lineage tracing with constitutive and lineage-specific fluorescent reporters demonstrated that GSCs generate the majority of vascular pericytes. Selective elimination of GSC-derived pericytes disrupts the neovasculature and potently inhibits tumor growth. Analysis of human GBM specimens showed that most pericytes are derived from neoplastic cells. GSCs are recruited toward endothelial cells via the SDF-1/CXCR4 axis and are induced to become pericytes predominantly by transforming growth factor ß. Thus, GSCs contribute to vascular pericytes that may actively remodel perivascular niches. Therapeutic targeting of GSC-derived pericytes may effectively block tumor progression and improve antiangiogenic therapy.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Madre Neoplásicas/patología , Pericitos/patología , Animales , Encéfalo/patología , Neoplasias Encefálicas/irrigación sanguínea , Diferenciación Celular , Células Endoteliales/patología , Glioblastoma/irrigación sanguínea , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Factor de Crecimiento Transformador beta/metabolismo , Trasplante Heterólogo
12.
Nature ; 603(7903): 885-892, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35165441

RESUMEN

The human brain vasculature is of great medical importance: its dysfunction causes disability and death1, and the specialized structure it forms-the blood-brain barrier-impedes the treatment of nearly all brain disorders2,3. Yet so far, we have no molecular map of the human brain vasculature. Here we develop vessel isolation and nuclei extraction for sequencing (VINE-seq) to profile the major vascular and perivascular cell types of the human brain through 143,793 single-nucleus transcriptomes from 25 hippocampus and cortex samples of 9 individuals with Alzheimer's disease and 8 individuals with no cognitive impairment. We identify brain-region- and species-enriched genes and pathways. We reveal molecular principles of human arteriovenous organization, recapitulating a gradual endothelial and punctuated mural cell continuum. We discover two subtypes of human pericytes, marked by solute transport and extracellular matrix (ECM) organization; and define perivascular versus meningeal fibroblast specialization. In Alzheimer's disease, we observe selective vulnerability of ECM-maintaining pericytes and gene expression patterns that implicate dysregulated blood flow. With an expanded survey of brain cell types, we find that 30 of the top 45 genes that have been linked to Alzheimer's disease risk by genome-wide association studies (GWASs) are expressed in the human brain vasculature, and we confirm this by immunostaining. Vascular GWAS genes map to endothelial protein transport, adaptive immune and ECM pathways. Many are microglia-specific in mice, suggesting a partial evolutionary transfer of Alzheimer's disease risk. Our work uncovers the molecular basis of the human brain vasculature, which will inform our understanding of overall brain health, disease and therapy.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Susceptibilidad a Enfermedades , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/irrigación sanguínea , Encéfalo/citología , Encéfalo/metabolismo , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Estudio de Asociación del Genoma Completo , Hipocampo/irrigación sanguínea , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Ratones , Microglía/metabolismo , Pericitos/metabolismo , Transcriptoma
13.
Annu Rev Cell Dev Biol ; 30: 677-704, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25150008

RESUMEN

Two opposing descriptions of so-called mesenchymal stem cells (MSCs) exist at this time. One sees MSCs as the postnatal, self-renewing, and multipotent stem cells for the skeleton. This cell coincides with a specific type of bone marrow perivascular cell. In skeletal physiology, this skeletal stem cell is pivotal to the growth and lifelong turnover of bone and to its native regeneration capacity. In hematopoietic physiology, its role as a key player in maintaining hematopoietic stem cells in their niche and in regulating the hematopoietic microenvironment is emerging. In the alternative description, MSCs are ubiquitous in connective tissues and are defined by in vitro characteristics and by their use in therapy, which rests on their ability to modulate the function of host tissues rather than on stem cell properties. Here, I discuss how the two views developed, conceptually and experimentally, and attempt to clarify the confusion arising from their collision.


Asunto(s)
Células Madre Mesenquimatosas/citología , Animales , Células de la Médula Ósea/clasificación , Células de la Médula Ósea/citología , Huesos/citología , Antígeno CD146/análisis , Separación Celular/métodos , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Células Clonales/citología , Tejido Conectivo/inmunología , Humanos , Inmunomodulación , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/clasificación , Ratones , Modelos Biológicos , Pericitos/citología , Células Madre Pluripotentes/citología , Quimera por Radiación , Nicho de Células Madre , Células del Estroma/clasificación , Células del Estroma/citología , Trasplante Heterotópico
14.
Development ; 151(18)2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39166965

RESUMEN

The microvascular system consists of two cell types: endothelial and mural (pericytes and vascular smooth muscle cells; VSMCs) cells. Communication between endothelial and mural cells plays a pivotal role in the maintenance of vascular homeostasis; however, in vivo molecular and cellular mechanisms underlying mural cell development remain unclear. In this study, we found that macrophages played a crucial role in TGFß-dependent pericyte-to-VSMC differentiation during retinal vasculature development. In mice with constitutively active Foxo1 overexpression, substantial accumulation of TGFß1-producing macrophages and pericytes around the angiogenic front region was observed. Additionally, the TGFß-SMAD pathway was activated in pericytes adjacent to macrophages, resulting in excess ectopic α-smooth muscle actin-positive VSMCs. Furthermore, we identified endothelial SEMA3C as an attractant for macrophages. In vivo neutralization of SEMA3C rescued macrophage accumulation and ectopic VSMC phenotypes in the mice, as well as drug-induced macrophage depletion. Therefore, macrophages play an important physiological role in VSMC development via the FOXO1-SEMA3C pathway.


Asunto(s)
Proteína Forkhead Box O1 , Macrófagos , Músculo Liso Vascular , Miocitos del Músculo Liso , Semaforinas , Animales , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Ratones , Semaforinas/metabolismo , Semaforinas/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/citología , Pericitos/metabolismo , Pericitos/citología , Diferenciación Celular , Transducción de Señal , Vasos Retinianos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Factor de Crecimiento Transformador beta1/metabolismo , Ratones Endogámicos C57BL
15.
PLoS Biol ; 22(4): e3002590, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683849

RESUMEN

Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.


Asunto(s)
Encéfalo , Pericitos , Factores de Transcripción , Proteínas de Pez Cebra , Animales , Encéfalo/metabolismo , Encéfalo/embriología , Diferenciación Celular , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Mesodermo/metabolismo , Mesodermo/citología , Cresta Neural/metabolismo , Cresta Neural/citología , Pericitos/metabolismo , Pericitos/citología , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
16.
Cell ; 151(2): 384-99, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23063127

RESUMEN

Vascular endothelial growth factor and its receptors, FLK1/KDR and FLT1, are key regulators of angiogenesis. Unlike FLK1/KDR, the role of FLT1 has remained elusive. FLT1 is produced as soluble (sFLT1) and full-length isoforms. Here, we show that pericytes from multiple tissues produce sFLT1. To define the biologic role of sFLT1, we chose the glomerular microvasculature as a model system. Deletion of Flt1 from specialized glomerular pericytes, known as podocytes, causes reorganization of their cytoskeleton with massive proteinuria and kidney failure, characteristic features of nephrotic syndrome in humans. The kinase-deficient allele of Flt1 rescues this phenotype, demonstrating dispensability of the full-length isoform. Using cell imaging, proteomics, and lipidomics, we show that sFLT1 binds to the glycosphingolipid GM3 in lipid rafts on the surface of podocytes, promoting adhesion and rapid actin reorganization. sFLT1 also regulates pericyte function in vessels outside of the kidney. Our findings demonstrate an autocrine function for sFLT1 to control pericyte behavior.


Asunto(s)
Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Podocitos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Gangliósidos/metabolismo , Humanos , Técnicas In Vitro , Metabolismo de los Lípidos , Lípidos/química , Ratones , Ratones Transgénicos , Pericitos/metabolismo , Proteinuria/metabolismo , Transducción de Señal , Sindecanos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
17.
Nature ; 589(7841): 281-286, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33176333

RESUMEN

Kidney fibrosis is the hallmark of chronic kidney disease progression; however, at present no antifibrotic therapies exist1-3. The origin, functional heterogeneity and regulation of scar-forming cells that occur during human kidney fibrosis remain poorly understood1,2,4. Here, using single-cell RNA sequencing, we profiled the transcriptomes of cells from the proximal and non-proximal tubules of healthy and fibrotic human kidneys to map the entire human kidney. This analysis enabled us to map all matrix-producing cells at high resolution, and to identify distinct subpopulations of pericytes and fibroblasts as the main cellular sources of scar-forming myofibroblasts during human kidney fibrosis. We used genetic fate-tracing, time-course single-cell RNA sequencing and ATAC-seq (assay for transposase-accessible chromatin using sequencing) experiments in mice, and spatial transcriptomics in human kidney fibrosis, to shed light on the cellular origins and differentiation of human kidney myofibroblasts and their precursors at high resolution. Finally, we used this strategy to detect potential therapeutic targets, and identified NKD2 as a myofibroblast-specific target in human kidney fibrosis.


Asunto(s)
Linaje de la Célula , Fibrosis/patología , Túbulos Renales/patología , Miofibroblastos/patología , Insuficiencia Renal Crónica/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Estudios de Casos y Controles , Diferenciación Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Masculino , Mesodermo/citología , Mesodermo/patología , Ratones , Miofibroblastos/metabolismo , Pericitos/citología , Pericitos/patología , RNA-Seq , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Análisis de la Célula Individual , Transcriptoma
18.
Annu Rev Physiol ; 85: 137-164, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36763972

RESUMEN

Pericytes, attached to the surface of capillaries, play an important role in regulating local blood flow. Using optogenetic tools and genetically encoded reporters in conjunction with confocal and multiphoton imaging techniques, the 3D structure, anatomical organization, and physiology of pericytes have recently been the subject of detailed examination. This work has revealed novel functions of pericytes and morphological features such as tunneling nanotubes in brain and tunneling microtubes in heart. Here, we discuss the state of our current understanding of the roles of pericytes in blood flow control in brain and heart, where functions may differ due to the distinct spatiotemporal metabolic requirements of these tissues. We also outline the novel concept of electro-metabolic signaling, a universal mechanistic framework that links tissue metabolic state with blood flow regulation by pericytes and vascular smooth muscle cells, with capillary KATP and Kir2.1 channels as primary sensors. Finally, we present major unresolved questions and outline how they can be addressed.


Asunto(s)
Nanotubos , Pericitos , Humanos , Encéfalo , Corazón , Capilares
19.
Hum Mol Genet ; 33(14): 1215-1228, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38652261

RESUMEN

Immunotherapy has revolutionized the treatment of tumors, but there are still a large number of patients who do not benefit from immunotherapy. Pericytes play an important role in remodeling the immune microenvironment. However, how pericytes affect the prognosis and treatment resistance of tumors is still unknown. This study jointly analyzed single-cell RNA sequencing (scRNA-seq) data and bulk RNA sequencing data of multiple cancers to reveal pericyte function in the colorectal cancer microenvironment. Analyzing over 800 000 cells, it was found that colorectal cancer had more pericyte enrichment in tumor tissues than other cancers. We then combined the TCGA database with multiple public datasets and enrolled more than 1000 samples, finding that pericyte may be closely related to poor prognosis due to the higher epithelial-mesenchymal transition (EMT) and hypoxic characteristics. At the same time, patients with more pericytes have higher immune checkpoint molecule expressions and lower immune cell infiltration. Finally, the contributions of pericyte in poor treatment response have been demonstrated in multiple immunotherapy datasets (n = 453). All of these observations suggest that pericyte can be used as a potential biomarker to predict patient disease progression and immunotherapy response.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Colorrectales , Inmunoterapia , Pericitos , Análisis de la Célula Individual , Microambiente Tumoral , Humanos , Pericitos/inmunología , Pericitos/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Análisis de la Célula Individual/métodos , Pronóstico , Inmunoterapia/métodos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Biomarcadores de Tumor/genética , Análisis de Secuencia de ARN/métodos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica
20.
EMBO J ; 41(9): e111189, 2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35437788

RESUMEN

A functional blood-brain barrier relies on a tightly controlled interplay between endothelial cells, pericytes, and astrocytes, which together form the neurovascular unit. Recent work by Lee et al (2022) discovers endothelial cell-derived lactate as a crucial metabolic fuel for brain pericytes, revealing a new way of CNS vascular communication that links nutrient metabolism to blood-brain barrier function.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales , Astrocitos , Encéfalo , Ecosistema , Pericitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA