Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 601
Filtrar
1.
EMBO J ; 43(8): 1519-1544, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38528180

RESUMO

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.


Assuntos
Neoplasias , Pericitos , Humanos , Pericitos/patologia , Pericitos/fisiologia , Guanilil Ciclase Solúvel , Células Endoteliais/fisiologia , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neoplasias/genética , Neoplasias/patologia , Guanilato Ciclase , Microambiente Tumoral
2.
J Cereb Blood Flow Metab ; 44(6): 881-895, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38513138

RESUMO

The blood-brain barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional changes during aging, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. In recent years, advances in microscopy and high-throughput bioinformatics have allowed a more in-depth investigation of the aging mechanisms of BBB. This review summarizes age-related alterations of the BBB structure and function from six perspectives: endothelial cells, astrocytes, pericytes, basement membrane, microglia and perivascular macrophages, and fibroblasts, ranging from the molecular level to the human multi-system level. These basic components are essential for the proper functioning of the BBB. Recent imaging methods of BBB were also reviewed. Elucidation of age-associated BBB changes may offer insights into BBB homeostasis and may provide effective therapeutic strategies to protect it during aging.


Assuntos
Envelhecimento , Barreira Hematoencefálica , Barreira Hematoencefálica/metabolismo , Humanos , Envelhecimento/fisiologia , Animais , Células Endoteliais/metabolismo , Pericitos/metabolismo , Pericitos/fisiologia , Astrócitos/metabolismo , Astrócitos/fisiologia
3.
Nat Rev Cardiol ; 21(2): 106-118, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37542118

RESUMO

Millions of cardiomyocytes die immediately after myocardial infarction, regardless of whether the culprit coronary artery undergoes prompt revascularization. Residual ischaemia in the peri-infarct border zone causes further cardiomyocyte damage, resulting in a progressive decline in contractile function. To date, no treatment has succeeded in increasing the vascularization of the infarcted heart. In the past decade, new approaches that can target the heart's highly plastic perivascular niche have been proposed. The perivascular environment is populated by mesenchymal progenitor cells, fibroblasts, myofibroblasts and pericytes, which can together mount a healing response to the ischaemic damage. In the infarcted heart, pericytes have crucial roles in angiogenesis, scar formation and stabilization, and control of the inflammatory response. Persistent ischaemia and accrual of age-related risk factors can lead to pericyte depletion and dysfunction. In this Review, we describe the phenotypic changes that characterize the response of cardiac pericytes to ischaemia and the potential of pericyte-based therapy for restoring the perivascular niche after myocardial infarction. Pericyte-related therapies that can salvage the area at risk of an ischaemic injury include exogenously administered pericytes, pericyte-derived exosomes, pericyte-engineered biomaterials, and pharmacological approaches that can stimulate the differentiation of constitutively resident pericytes towards an arteriogenic phenotype. Promising preclinical results from in vitro and in vivo studies indicate that pericytes have crucial roles in the treatment of coronary artery disease and the prevention of post-ischaemic heart failure.


Assuntos
Infarto do Miocárdio , Pericitos , Humanos , Pericitos/fisiologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos , Isquemia , Vasos Coronários
4.
Trends Cell Biol ; 34(1): 58-71, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37474376

RESUMO

Pericytes are known as the mural cells in small-caliber vessels that interact closely with the endothelium. Pericytes play a key role in vasculature formation and homeostasis, and when dysfunctional contribute to vasculature-related diseases such as diabetic retinopathy and neurodegenerative conditions. In addition, significant extravascular roles of pathological pericytes are being discovered with relevant implications for cancer and fibrosis. Pericyte research is challenged by the lack of consistent molecular markers and clear discrimination criteria versus other (mural) cells. However, advances in single-cell approaches are uncovering and clarifying mural cell identities, biological functions, and ontogeny across organs. We discuss the latest developments in pericyte pathobiology to inform future research directions and potential outcomes.


Assuntos
Endotélio Vascular , Pericitos , Humanos , Pericitos/fisiologia , Biomarcadores , Homeostase
5.
Brain ; 147(3): 1057-1074, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38153327

RESUMO

Incomplete reperfusion of the microvasculature ('no-reflow') after ischaemic stroke damages salvageable brain tissue. Previous ex vivo studies suggest pericytes are vulnerable to ischaemia and may exacerbate no-reflow, but the viability of pericytes and their association with no-reflow remains under-explored in vivo. Using longitudinal in vivo two-photon single-cell imaging over 7 days, we showed that 87% of pericytes constrict during cerebral ischaemia and remain constricted post reperfusion, and 50% of the pericyte population are acutely damaged. Moreover, we revealed ischaemic pericytes to be fundamentally implicated in capillary no-reflow by limiting and arresting blood flow within the first 24 h post stroke. Despite sustaining acute membrane damage, we observed that over half of all cortical pericytes survived ischaemia and responded to vasoactive stimuli, upregulated unique transcriptomic profiles and replicated. Finally, we demonstrated the delayed recovery of capillary diameter by ischaemic pericytes after reperfusion predicted vessel reconstriction in the subacute phase of stroke. Cumulatively, these findings demonstrate that surviving cortical pericytes remain both viable and promising therapeutic targets to counteract no-reflow after ischaemic stroke.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Humanos , Pericitos/fisiologia , Infarto Cerebral
6.
Animal Model Exp Med ; 6(4): 337-345, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37317664

RESUMO

Pericytes are the main cellular components of tiny arteries and capillaries. Studies have found that pericytes can undergo morphological contraction or relaxation under stimulation by cytokines, thus affecting the contraction and relaxation of microvessels and playing an essential role in regulating vascular microcirculation. Moreover, due to the characteristics of stem cells, pericytes can differentiate into a variety of inflammatory cell phenotypes, which then affect the immune function. Additionally, pericytes can also participate in angiogenesis and wound healing by interacting with endothelial cells in vascular microcirculation disorders. Here we review the origin, biological phenotype and function of pericytes, and discuss the potential mechanisms of pericytes in vascular microcirculation disorders, especially in pulmonary hypertension, so as to provide a sound basis and direction for the prevention and treatment of vascular microcirculation diseases.


Assuntos
Células Endoteliais , Pericitos , Pericitos/fisiologia , Microcirculação , Capilares , Biologia
7.
Pharmacol Ther ; 249: 108484, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37390969

RESUMO

Chronic and neuropathic pain are a widespread burden. Incomplete understanding of underlying pathomechanisms is one crucial factor for insufficient treatment. Recently, impairment of the blood nerve barrier (BNB) has emerged as one key aspect of pain initiation and maintenance. In this narrative review, we discuss several mechanisms and putative targets for novel treatment strategies. Cells such as pericytes, local mediators like netrin-1 and specialized proresolving mediators (SPMs), will be covered as well as circulating factors including the hormones cortisol and oestrogen and microRNAs. They are crucial in either the BNB or similar barriers and associated with pain. While clinical studies are still scarce, these findings might provide valuable insight into mechanisms and nurture development of therapeutic approaches.


Assuntos
MicroRNAs , Neuralgia , Humanos , Barreira Hematoneural/fisiologia , Pericitos/fisiologia
8.
Cell Reprogram ; 25(5): 212-223, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37366790

RESUMO

Studying human somatic cell-to-neuron conversion using primary brain-derived cells as starting cell source is hampered by limitations and variations in human biopsy material. Thus, delineating the molecular variables that allow changing the identity of somatic cells, permit adoption of neuronal phenotypes, and foster maturation of induced neurons (iNs) is challenging. Based on our previous results that pericytes derived from the adult human cerebral cortex can be directly converted into iNs (Karow et al., 2018; Karow et al., 2012), we here introduce human induced pluripotent stem cell (hiPSC)-derived pericytes (hiPSC-pericytes) as a versatile and more uniform tool to study the pericyte-to-neuron conversion process. This strategy enables us to derive scalable cell numbers and allows for engineering of the starting cell population such as introducing reporter tools before differentiation into hiPSC-pericytes and subsequent iN conversion. Harvesting the potential of this approach, we established hiPSC-derived human-human neuronal cocultures that not only allow for independent manipulation of each coculture partner but also resulted in morphologically more mature iNs. In summary, we exploit hiPSC-based methods to facilitate the analysis of human somatic cell-to-neuron conversion.


Assuntos
Células-Tronco Pluripotentes Induzidas , Adulto , Humanos , Reprogramação Celular , Pericitos/fisiologia , Neurônios , Diferenciação Celular/fisiologia
9.
Stroke ; 54(8): 2172-2177, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37381898

RESUMO

BACKGROUND: Subarachnoid hemorrhage (SAH) is characterized by an acute reduction of cerebral blood flow and subsequent cortical infarcts, but the underlying mechanisms are not well understood. Since pericytes regulate cerebral perfusion on the capillary level, we hypothesize that pericytes may reduce cerebral perfusion after SAH. METHODS: Pericytes and vessel diameters of cerebral microvessels were imaged in vivo using NG2 (neuron-glial antigen 2) reporter mice and 2-photon microscopy before and 3 hours after sham surgery or induction of SAH by perforating the middle cerebral artery with an intraluminal filament. Twenty-four hours after, SAH pericyte density was assessed by immunohistochemistry. RESULTS: SAH caused pearl-string-like constrictions of pial arterioles, slowed down blood flow velocity in pial arterioles by 50%, and reduced the volume of intraparenchymal arterioles and capillaries by up to 70% but did not affect pericyte density or induce capillary constriction by pericytes. CONCLUSIONS: Our results suggest that perfusion deficits after SAH are not induced by pericyte-mediated capillary constrictions.


Assuntos
Pericitos , Hemorragia Subaracnóidea , Camundongos , Animais , Pericitos/fisiologia , Capilares , Hemorragia Subaracnóidea/complicações , Vasoconstrição/fisiologia , Perfusão
10.
Neuromolecular Med ; 25(4): 457-470, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37166748

RESUMO

Pericytes surrounding endothelial cells in the capillaries are emerging as an attractive cell resource, which can show a large variety of functions in ischemic stroke, including preservation of the blood-brain barrier, regulation of immune function, and support for cerebral vasculature. These functions have been fully elucidated in previous studies. However, in recent years, increasing evidence has shown that pericytes play an important role in neurological recovery after ischemic stroke due to their regenerative function which can be summarized in two aspects according to current discoveries, one is that pericytes are thought to be multipotential themselves, and the other is that pericytes can promote the differentiation of oligodendrocyte progenitor cells (OPCs). Considering the neuroprotective treatment for stroke has not been much progressed in recent years, new therapies targeting pericytes may be a future direction. Here, we will review the beneficial effects of pericytes in ischemic stroke from two directions: the barrier and vascular functions and the regenerative functions of pericytes.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Humanos , Células Endoteliais , Pericitos/fisiologia , Acidente Vascular Cerebral/terapia , Barreira Hematoencefálica
11.
Cells ; 12(8)2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37190075

RESUMO

Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been paid to regenerative medicine to overcome this issue. Direct reprogramming is a promising therapeutic approach in regenerative cardiac medicine that has the potential to provide in situ cardiac regeneration. It consists of direct cell fate conversion of one cell type into another, avoiding transition through an intermediary pluripotent state. In injured cardiac tissue, this strategy directs transdifferentiation of resident non-myocyte cells (NMCs) into mature functional cardiac cells that help to restore the native tissue. Over the years, developments in reprogramming methods have suggested that regulation of several intrinsic factors in NMCs can help to achieve in situ direct cardiac reprogramming. Among NMCs, endogenous cardiac fibroblasts have been studied for their potential to be directly reprogrammed into both induced cardiomyocytes and induced cardiac progenitor cells, while pericytes can transdifferentiate towards endothelial cells and smooth muscle cells. This strategy has been indicated to improve heart function and reduce fibrosis after cardiac injury in preclinical models. This review summarizes the recent updates and progress in direct cardiac reprogramming of resident NMCs for in situ cardiac regeneration.


Assuntos
Transdiferenciação Celular , Técnicas de Reprogramação Celular , Reprogramação Celular , Fibroblastos , Cardiopatias , Coração , Pericitos , Regeneração , Coração/fisiologia , Cardiopatias/terapia , Fibroblastos/citologia , Fibroblastos/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Pericitos/citologia , Pericitos/fisiologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Animais
12.
J Physiol ; 601(12): 2263-2272, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37036208

RESUMO

The brain is an energetically demanding tissue which, to function adequately, requires constant fine tuning of its supporting blood flow, and hence energy supply. Whilst blood flow was traditionally believed to be regulated only by vascular smooth muscle cells on arteries and arterioles supplying the brain, recent work has suggested a critical role for capillary pericytes, which are also contractile. This concept has evoked some controversy, especially over the relative contributions of arterioles and capillaries to the control of cerebral blood flow. Here we outline why pericytes are in a privileged position to control cerebral blood flow. First we discuss the evidence, and fundamental equations, which describe how the small starting diameter of capillaries, compared to upstream arterioles, confers a potentially greater control by capillary pericytes than by arterioles over total cerebral vascular resistance. Then we suggest that the faster time frame over which low branch order capillary pericytes dilate in response to local energy demands provides a niche role for pericytes to regulate blood flow compared to slower responding arterioles. Finally, we discuss the role of pericytes in capillary stalling, whereby pericyte contraction appears to facilitate a transient stall of circulating blood cells, exacerbating the effect of pericytes upon cerebral blood flow.


Assuntos
Encéfalo , Pericitos , Arteríolas/fisiologia , Pericitos/fisiologia , Encéfalo/irrigação sanguínea , Capilares/fisiologia , Contração Muscular/fisiologia
13.
Adv Sci (Weinh) ; 10(20): e2206938, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37102631

RESUMO

Evidence suggests a unique association between bone aging and neurodegenerative/cerebrovascular disorders. However, the mechanisms underlying bone-brain interplay remain elusive. Here platelet-derived growth factor-BB (PDGF-BB) produced by preosteoclasts in bone is reported to promote age-associated hippocampal vascular impairment. Aberrantly elevated circulating PDGF-BB in aged mice and high-fat diet (HFD)-challenged mice correlates with capillary reduction, pericyte loss, and increased blood-brain barrier (BBB) permeability in their hippocampus. Preosteoclast-specific Pdgfb transgenic mice with markedly high plasma PDGF-BB concentration faithfully recapitulate the age-associated hippocampal BBB impairment and cognitive decline. Conversely, preosteoclast-specific Pdgfb knockout mice have attenuated hippocampal BBB impairment in aged mice or HFD-challenged mice. Persistent exposure of brain pericytes to high concentrations of PDGF-BB upregulates matrix metalloproteinase 14 (MMP14), which promotes ectodomain shedding of PDGF receptor ß (PDGFRß) from pericyte surface. MMP inhibitor treatment alleviates hippocampal pericyte loss and capillary reduction in the conditional Pdgfb transgenic mice and antagonizes BBB leakage in aged mice. The findings establish the role of bone-derived PDGF-BB in mediating hippocampal BBB disruption and identify the ligand-induced PDGFRß shedding as a feedback mechanism for age-associated PDGFRß downregulation and the consequent pericyte loss.


Assuntos
Hipocampo , Pericitos , Animais , Camundongos , Becaplermina , Camundongos Knockout , Camundongos Transgênicos , Pericitos/fisiologia , Proteínas Proto-Oncogênicas c-sis , Receptor beta de Fator de Crescimento Derivado de Plaquetas
14.
Nat Aging ; 3(2): 173-184, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-37118115

RESUMO

The microvascular inflow tract, comprising the penetrating arterioles, precapillary sphincters and first-order capillaries, is the bottleneck for brain blood flow and energy supply. Exactly how aging alters the structure and function of the microvascular inflow tract remains unclear. By in vivo four-dimensional two-photon imaging, we reveal an age-dependent decrease in vaso-responsivity accompanied by a decrease in vessel density close to the arterioles and loss of vascular mural cell processes, although the number of mural cell somas and their alpha smooth muscle actin density were preserved. The age-related reduction in vascular reactivity was mostly pronounced at precapillary sphincters, highlighting their crucial role in capillary blood flow regulation. Mathematical modeling revealed impaired pressure and flow control in aged mice during vasoconstriction. Interventions that preserve dynamics of cerebral blood vessels may ameliorate age-related decreases in blood flow and prevent brain frailty.


Assuntos
Capilares , Pericitos , Camundongos , Animais , Pericitos/fisiologia , Capilares/fisiologia , Arteríolas/fisiologia , Encéfalo/irrigação sanguínea , Hemodinâmica
15.
J Physiol ; 601(5): 889-901, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36751860

RESUMO

The brain is an energy hog, consuming available energy supplies at a rate out of all proportion to its relatively small size. This outsized demand, largely reflecting the unique computational activity of the brain, is met by an ensemble of neurovascular coupling mechanisms that link neuronal activity with local increases in blood delivery. This just-in-time replenishment strategy, made necessary by the limited energy-storage capacity of neurons, complicates the nutrient-delivery task of the cerebral vasculature, layering on a temporo-spatial requirement that invites - and challenges - mechanistic interpretation. The centre of gravity of research efforts to disentangle these mechanisms has shifted from an initial emphasis on astrocyte-arteriole-level processes to mechanisms that operate on the capillary level, a shift that has brought into sharp focus questions regarding the fine control of blood distribution to active neurons. As these investigations have drilled down into finer reaches of the microvasculature, they have revealed an arteriole-proximate subregion of CNS capillary networks that serves a regulatory function in directing blood flow into and within downstream capillaries. They have also illuminated differences in researchers' perspectives on the vascular structures and identity of mural cells in this region that impart the vasomodulatory effects that control blood distribution. In this review, we highlight the regulatory role of a variably named region of the microvasculature, referred to here as the post-arteriole transition zone, in channeling blood flow within CNS capillary networks, and underscore the contribution of dynamically contractile perivascular mural cell - generally, but not universally, recognized as pericytes - to this function.


Assuntos
Capilares , Microvasos , Arteríolas/fisiologia , Capilares/fisiologia , Pericitos/fisiologia , Encéfalo/irrigação sanguínea
16.
Dev Cell ; 58(3): 174-191.e8, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36706755

RESUMO

The blood barriers of the nervous system protect neural environments but can hinder therapeutic accessibility. The blood-brain barrier (BBB) is well characterized, consisting of endothelial cells with specialized tight junctions and low levels of transcytosis, properties conferred by contacting pericytes and astrocytes. In contrast, the blood-nerve barrier (BNB) of the peripheral nervous system is poorly defined. Here, we characterize the structure of the mammalian BNB, identify the processes that confer barrier function, and demonstrate how the barrier can be opened in response to injury. The homeostatic BNB is leakier than the BBB, which we show is due to higher levels of transcytosis. However, the barrier is reinforced by macrophages that specifically engulf leaked materials, identifying a role for resident macrophages as an important component of the BNB. Finally, we demonstrate the exploitation of these processes to effectively deliver RNA-targeting therapeutics to peripheral nerves, indicating new treatment approaches for nervous system pathologies.


Assuntos
Barreira Hematoneural , Células Endoteliais , Animais , Barreira Hematoneural/fisiologia , Células Endoteliais/fisiologia , Barreira Hematoencefálica/fisiologia , Macrófagos , Pericitos/fisiologia , Mamíferos
18.
Brain ; 145(7): 2276-2292, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35551356

RESUMO

To match the metabolic demands of the brain, mechanisms have evolved to couple neuronal activity to vasodilation, thus increasing local cerebral blood flow and delivery of oxygen and glucose to active neurons. Rather than relying on metabolic feedback signals such as the consumption of oxygen or glucose, the main signalling pathways rely on the release of vasoactive molecules by neurons and astrocytes, which act on contractile cells. Vascular smooth muscle cells and pericytes are the contractile cells associated with arterioles and capillaries, respectively, which relax and induce vasodilation. Much progress has been made in understanding the complex signalling pathways of neurovascular coupling, but issues such as the contributions of capillary pericytes and astrocyte calcium signal remain contentious. Study of neurovascular coupling mechanisms is especially important as cerebral blood flow dysregulation is a prominent feature of Alzheimer's disease. In this article we will discuss developments and controversies in the understanding of neurovascular coupling and finish by discussing current knowledge concerning neurovascular uncoupling in Alzheimer's disease.


Assuntos
Doença de Alzheimer , Acoplamento Neurovascular , Doença de Alzheimer/metabolismo , Astrócitos/metabolismo , Encéfalo , Circulação Cerebrovascular/fisiologia , Glucose/metabolismo , Humanos , Acoplamento Neurovascular/fisiologia , Oxigênio , Pericitos/fisiologia
19.
Elife ; 112022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35285797

RESUMO

Acute kidney injury is common, with ~13 million cases and 1.7 million deaths/year worldwide. A major cause is renal ischaemia, typically following cardiac surgery, renal transplant or severe haemorrhage. We examined the cause of the sustained reduction in renal blood flow ('no-reflow'), which exacerbates kidney injury even after an initial cause of compromised blood supply is removed. Adult male Sprague-Dawley rats, or NG2-dsRed male mice were used in this study. After 60 min kidney ischaemia and 30-60 min reperfusion, renal blood flow remained reduced, especially in the medulla, and kidney tubule damage was detected as Kim-1 expression. Constriction of the medullary descending vasa recta and cortical peritubular capillaries occurred near pericyte somata, and led to capillary blockages, yet glomerular arterioles and perfusion were unaffected, implying that the long-lasting decrease of renal blood flow contributing to kidney damage was generated by pericytes. Blocking Rho kinase to decrease pericyte contractility from the start of reperfusion increased the post-ischaemic diameter of the descending vasa recta capillaries at pericytes, reduced the percentage of capillaries that remained blocked, increased medullary blood flow and reduced kidney injury. Thus, post-ischaemic renal no-reflow, contributing to acute kidney injury, reflects pericytes constricting the descending vasa recta and peritubular capillaries. Pericytes are therefore an important therapeutic target for treating acute kidney injury.


Assuntos
Injúria Renal Aguda , Pericitos , Animais , Capilares , Constrição , Feminino , Humanos , Isquemia , Rim , Masculino , Camundongos , Pericitos/fisiologia , Ratos , Ratos Sprague-Dawley
20.
Neuron ; 110(10): 1641-1655.e6, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35294899

RESUMO

Endothelial cells of blood vessels of the central nervous system (CNS) constitute blood-CNS barriers. Barrier properties are not intrinsic to these cells; rather they are induced and maintained by CNS microenvironment. Notably, the abluminal surfaces of CNS capillaries are ensheathed by pericytes and astrocytes. However, extrinsic factors from these perivascular cells that regulate barrier integrity are largely unknown. Here, we establish vitronectin, an extracellular matrix protein secreted by CNS pericytes, as a regulator of blood-CNS barrier function via interactions with its integrin receptor, α5, in endothelial cells. Genetic ablation of vitronectin or mutating vitronectin to prevent integrin binding, as well as endothelial-specific deletion of integrin α5, causes barrier leakage in mice. Furthermore, vitronectin-integrin α5 signaling maintains barrier integrity by actively inhibiting transcytosis in endothelial cells. These results demonstrate that signaling from perivascular cells to endothelial cells via ligand-receptor interactions is a key mechanism to regulate barrier permeability.


Assuntos
Células Endoteliais , Pericitos , Animais , Barreira Hematoencefálica/metabolismo , Sistema Nervoso Central/metabolismo , Células Endoteliais/metabolismo , Integrina alfa5/metabolismo , Integrinas/metabolismo , Camundongos , Pericitos/fisiologia , Vitronectina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...