Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38951020

RESUMEN

Blood vessels are critical to deliver oxygen and nutrients to tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier (BBB), which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and protects the neural tissue from toxins and pathogens, and alterations of this barrier are important components of the pathogenesis and progression of various neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the brain endothelial cells (ECs) that form the walls of the blood vessels. These properties are regulated by interactions between different vascular, perivascular, immune, and neural cells. Understanding how these cell populations interact to regulate barrier properties is essential for understanding how the brain functions in both health and disease contexts.

2.
Dev Cell ; 58(17): 1534-1547.e6, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37437574

RESUMEN

The blood-brain barrier (BBB) is a unique set of properties of the brain vasculature which severely restrict its permeability to proteins and small molecules. Classic chick-quail chimera studies have shown that these properties are not intrinsic to the brain vasculature but rather are induced by surrounding neural tissue. Here, we identify Spock1 as a candidate neuronal signal for regulating BBB permeability in zebrafish and mice. Mosaic genetic analysis shows that neuronally expressed Spock1 is cell non-autonomously required for a functional BBB. Leakage in spock1 mutants is associated with altered extracellular matrix (ECM), increased endothelial transcytosis, and altered pericyte-endothelial interactions. Furthermore, a single dose of recombinant SPOCK1 partially restores BBB function in spock1 mutants by quenching gelatinase activity and restoring vascular expression of BBB genes including mcamb. These analyses support a model in which neuronally secreted Spock1 initiates BBB properties by altering the ECM, thereby regulating pericyte-endothelial interactions and downstream vascular gene expression.


Asunto(s)
Barrera Hematoencefálica , Proteoglicanos , Pez Cebra , Animales , Ratones , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo , Endotelio/metabolismo , Proteoglicanos/metabolismo
3.
Stroke ; 54(6): e251-e271, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37009740

RESUMEN

BACKGROUND: Preservation of brain health has emerged as a leading public health priority for the aging world population. Advances in neurovascular biology have revealed an intricate relationship among brain cells, meninges, and the hematic and lymphatic vasculature (the neurovasculome) that is highly relevant to the maintenance of cognitive function. In this scientific statement, a multidisciplinary team of experts examines these advances, assesses their relevance to brain health and disease, identifies knowledge gaps, and provides future directions. METHODS: Authors with relevant expertise were selected in accordance with the American Heart Association conflict-of-interest management policy. They were assigned topics pertaining to their areas of expertise, reviewed the literature, and summarized the available data. RESULTS: The neurovasculome, composed of extracranial, intracranial, and meningeal vessels, as well as lymphatics and associated cells, subserves critical homeostatic functions vital for brain health. These include delivering O2 and nutrients through blood flow and regulating immune trafficking, as well as clearing pathogenic proteins through perivascular spaces and dural lymphatics. Single-cell omics technologies have unveiled an unprecedented molecular heterogeneity in the cellular components of the neurovasculome and have identified novel reciprocal interactions with brain cells. The evidence suggests a previously unappreciated diversity of the pathogenic mechanisms by which disruption of the neurovasculome contributes to cognitive dysfunction in neurovascular and neurodegenerative diseases, providing new opportunities for the prevention, recognition, and treatment of these conditions. CONCLUSIONS: These advances shed new light on the symbiotic relationship between the brain and its vessels and promise to provide new diagnostic and therapeutic approaches for brain disorders associated with cognitive dysfunction.


Asunto(s)
Disfunción Cognitiva , Accidente Cerebrovascular , Estados Unidos , Humanos , American Heart Association , Accidente Cerebrovascular/terapia , Encéfalo , Cognición
4.
Nat Cardiovasc Res ; 1(4): 389-400, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35571675

RESUMEN

Endothelial cells have a crucial role in nervous system function, and mounting evidence points to endothelial impairment as a major contributor to a wide range of neurological diseases. However, tools to genetically interrogate these cells in vivo remain limited. Here, we describe AAV-BI30, a capsid that specifically and efficiently transduces endothelial cells throughout the central nervous system. At relatively low systemic doses, this vector transduces the majority of arterial, capillary, and venous endothelial cells in the brain, retina, and spinal cord vasculature of adult C57BL/6 mice. Furthermore, we show that AAV-BI30 robustly transduces endothelial cells in multiple mouse strains and rats in vivo and human brain microvascular endothelial cells in vitro. Finally, we demonstrate AAV-BI30's capacity to achieve efficient and endothelial-specific Cre-mediated gene manipulation in the central nervous system. This combination of attributes makes AAV-BI30 uniquely well-suited to address outstanding research questions in neurovascular biology and aid the development of therapeutics to remediate endothelial dysfunction in disease.

5.
Neuron ; 110(10): 1641-1655.e6, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35294899

RESUMEN

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.


Asunto(s)
Células Endoteliales , Pericitos , Animales , Barrera Hematoencefálica/metabolismo , Sistema Nervioso Central/metabolismo , Células Endoteliales/metabolismo , Integrina alfa5/metabolismo , Integrinas/metabolismo , Ratones , Pericitos/fisiología , Vitronectina/metabolismo
6.
Nature ; 596(7872): 444-448, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34349262

RESUMEN

MFSD2A is a sodium-dependent lysophosphatidylcholine symporter that is responsible for the uptake of docosahexaenoic acid into the brain1,2, which is crucial for the development and performance of the brain3. Mutations that affect MFSD2A cause microcephaly syndromes4,5. The ability of MFSD2A to transport lipid is also a key mechanism that underlies its function as an inhibitor of transcytosis to regulate the blood-brain barrier6,7. Thus, MFSD2A represents an attractive target for modulating the permeability of the blood-brain barrier for drug delivery. Here we report the cryo-electron microscopy structure of mouse MFSD2A. Our structure defines the architecture of this important transporter, reveals its unique extracellular domain and uncovers its substrate-binding cavity. The structure-together with our functional studies and molecular dynamics simulations-identifies a conserved sodium-binding site, reveals a potential lipid entry pathway and helps to rationalize MFSD2A mutations that underlie microcephaly syndromes. These results shed light on the critical lipid transport function of MFSD2A and provide a framework to aid in the design of specific modulators for therapeutic purposes.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Metabolismo de los Lípidos , Simportadores/química , Simportadores/metabolismo , Animales , Sitios de Unión , Transporte Biológico , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación , Dominios Proteicos , Sodio/metabolismo , Simportadores/genética , Simportadores/ultraestructura
7.
ACS Chem Biol ; 16(1): 106-115, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33315366

RESUMEN

The cell membrane of brain endothelial cells is enriched in omega-3 phospholipid species. Numerous omega-3 phospholipid species were recently proposed to be important for maintaining the low rate of transcytosis and, thus, could be important for regulating one of the mechanisms of the blood brain barrier (BBB). However, the spatial distribution of these phospholipid species within the brain was previously unknown. Here, we combined advanced mass spectrometry imaging techniques to generate a map of these phospholipids in the brain at near single cell resolution. Furthermore, we explored the effects of omega-3 dietary deprivation on both docosahexaenoic acid (DHA)-containing phospholipids and the global brain phospholipid profile. We demonstrate the unique spatial distribution of individual DHA-containing phospholipids, which may be important for the regiospecific properties of the BBB. Finally, 24 diet discriminative phospholipids were identified and showed an increase in saturated phospholipid species and ceramide containing phospholipid species under omega-3 dietary deficiency.


Asunto(s)
Ácidos Grasos Omega-3/metabolismo , Fosfolípidos/farmacología , Transcitosis/efectos de los fármacos , Animales , Barrera Hematoencefálica , Femenino , Masculino , Ratones , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
8.
Nat Rev Neurosci ; 21(8): 416-432, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32636528

RESUMEN

To continuously process neural activity underlying sensation, movement and cognition, the CNS requires a homeostatic microenvironment that is not only enriched in nutrients to meet its high metabolic demands but that is also devoid of toxins that might harm the sensitive neural tissues. This highly regulated microenvironment is made possible by two unique features of CNS vasculature absent in the peripheral organs. First, the blood-blood barrier, which partitions the circulating blood from the CNS, acts as a gatekeeper to facilitate the selective trafficking of substances between the blood and the parenchyma. Second, neurovascular coupling ensures that, following local neural activation, regional blood flow is increased to quickly supply more nutrients and remove metabolic waste. Here, we review how neural and vascular activity act on one another with regard to these two properties.


Asunto(s)
Barrera Hematoencefálica/fisiología , Encéfalo/irrigación sanguínea , Encéfalo/fisiología , Neuronas/fisiología , Acoplamiento Neurovascular/fisiología , Animales , Humanos , Modelos Neurológicos
9.
Elife ; 92020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32613945

RESUMEN

The mouse cerebral cortex contains neurons that express choline acetyltransferase (ChAT) and are a potential local source of acetylcholine. However, the neurotransmitters released by cortical ChAT+ neurons and their synaptic connectivity are unknown. We show that the nearly all cortical ChAT+ neurons in mice are specialized VIP+ interneurons that release GABA strongly onto other inhibitory interneurons and acetylcholine sparsely onto layer 1 interneurons and other VIP+/ChAT+ interneurons. This differential transmission of ACh and GABA based on the postsynaptic target neuron is reflected in VIP+/ChAT+ interneuron pre-synaptic terminals, as quantitative molecular analysis shows that only a subset of these are specialized to release acetylcholine. In addition, we identify a separate, sparse population of non-VIP ChAT+ neurons in the medial prefrontal cortex with a distinct developmental origin that robustly release acetylcholine in layer 1. These results demonstrate both cortex-region heterogeneity in cortical ChAT+ interneurons and target-specific co-release of acetylcholine and GABA.


Asunto(s)
Acetilcolina/metabolismo , Encéfalo/metabolismo , Colina O-Acetiltransferasa/metabolismo , Neuronas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Corteza Cerebral/metabolismo , Heterocigoto , Interneuronas/metabolismo , Ratones , Corteza Prefrontal/metabolismo , Terminales Presinápticos/metabolismo
10.
Nature ; 579(7797): 106-110, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32076269

RESUMEN

Proper brain function depends on neurovascular coupling: neural activity rapidly increases local blood flow to meet moment-to-moment changes in regional brain energy demand1. Neurovascular coupling is the basis for functional brain imaging2, and impaired neurovascular coupling is implicated in neurodegeneration1. The underlying molecular and cellular mechanisms of neurovascular coupling remain poorly understood. The conventional view is that neurons or astrocytes release vasodilatory factors that act directly on smooth muscle cells (SMCs) to induce arterial dilation and increase local blood flow1. Here, using two-photon microscopy to image neural activity and vascular dynamics simultaneously in the barrel cortex of awake mice under whisker stimulation, we found that arteriolar endothelial cells (aECs) have an active role in mediating neurovascular coupling. We found that aECs, unlike other vascular segments of endothelial cells in the central nervous system, have abundant caveolae. Acute genetic perturbations that eliminated caveolae in aECs, but not in neighbouring SMCs, impaired neurovascular coupling. Notably, caveolae function in aECs is independent of the endothelial NO synthase (eNOS)-mediated NO pathway. Ablation of both caveolae and eNOS completely abolished neurovascular coupling, whereas the single mutants exhibited partial impairment, revealing that the caveolae-mediated pathway in aECs is a major contributor to neurovascular coupling. Our findings indicate that vasodilation is largely mediated by endothelial cells that actively relay signals from the central nervous system to SMCs via a caveolae-dependent pathway.


Asunto(s)
Arteriolas/citología , Arteriolas/metabolismo , Caveolas/metabolismo , Sistema Nervioso Central/citología , Acoplamiento Neurovascular , Animales , Corteza Cerebral/citología , Células Endoteliales/metabolismo , Femenino , Masculino , Ratones , Microscopía de Fluorescencia por Excitación Multifotónica , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/metabolismo , Vasodilatación , Vibrisas/fisiología
11.
Elife ; 82019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31429822

RESUMEN

As an optically transparent model organism with an endothelial blood-brain barrier (BBB), zebrafish offer a powerful tool to study the vertebrate BBB. However, the precise developmental profile of functional zebrafish BBB acquisition and the subcellular and molecular mechanisms governing the zebrafish BBB remain poorly characterized. Here, we capture the dynamics of developmental BBB leakage using live imaging, revealing a combination of steady accumulation in the parenchyma and sporadic bursts of tracer leakage. Electron microscopy studies further reveal high levels of transcytosis in brain endothelium early in development that are suppressed later. The timing of this suppression of transcytosis coincides with the establishment of BBB function. Finally, we demonstrate a key mammalian BBB regulator Mfsd2a, which inhibits transcytosis, plays a conserved role in zebrafish, as mfsd2aa mutants display increased BBB permeability due to increased transcytosis. Our findings indicate a conserved developmental program of barrier acquisition between zebrafish and mice.


Asunto(s)
Barrera Hematoencefálica/embriología , Barrera Hematoencefálica/fisiología , Células Endoteliales/fisiología , Transcitosis , Pez Cebra , Animales , Microscopía Intravital , Microscopía Electrónica
12.
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
13.
Curr Opin Neurobiol ; 57: 32-38, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30708291

RESUMEN

The blood-brain barrier (BBB) is a functional interface separating the brain from the circulatory system and is essential for homeostasis of the central nervous system (CNS). The BBB regulates molecular flux to maintain an optimal environment for neuronal function and protects the brain from toxins and pathogens. Endothelial cells forming the walls of CNS blood vessels constitute the BBB. CNS endothelial cells exhibit two features that underlie the restrictive properties of the BBB: specialized tight junctions that prevent paracellular passage between the blood and the brain, and unusually low levels of vesicle trafficking that limit transcellular transport or transcytosis. While the prevailing view in the field was that specialized tight junctions contributed to CNS barrier properties, recent findings have revealed the importance of maintaining low rates of transcytosis at the BBB. It is now clear that suppression of transcytosis at the BBB is an active process and CNS-specific genetic programs inhibit this pathway to maintain a functional barrier.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales , Transporte Biológico , Encéfalo , Transcitosis
14.
Ann Neurol ; 84(3): 409-423, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30014540

RESUMEN

OBJECTIVE: Cortical spreading depolarizations (CSDs) are intense and ubiquitous depolarization waves relevant for the pathophysiology of migraine and brain injury. CSDs disrupt the blood-brain barrier (BBB), but the mechanisms are unknown. METHODS: A total of six CSDs were evoked over 1 hour by topical application of 300 mM of KCl or optogenetically with 470 nm (blue) LED over the right hemisphere in anesthetized mice (C57BL/6 J wild type, Thy1-ChR2-YFP line 18, and cav-1-/- ). BBB disruption was assessed by Evans blue (2% EB, 3 ml/kg, intra-arterial) or dextran (200 mg/kg, fluorescein, 70,000 MW, intra-arterial) extravasation in parietotemporal cortex at 3 to 24 hours after CSD. Endothelial cell ultrastructure was examined using transmission electron microscopy 0 to 24 hours after the same CSD protocol in order to assess vesicular trafficking, endothelial tight junctions, and pericyte integrity. Mice were treated with vehicle, isoform nonselective rho-associated kinase (ROCK) inhibitor fasudil (10 mg/kg, intraperitoneally 30 minutes before CSD), or ROCK-2 selective inhibitor KD025 (200 mg/kg, per oral twice-daily for 5 doses before CSD). RESULTS: We show that CSD-induced BBB opening to water and large molecules is mediated by increased endothelial transcytosis starting between 3 and 6 hours and lasting approximately 24 hours. Endothelial tight junctions, pericytes, and basement membrane remain preserved after CSDs. Moreover, we show that CSD-induced BBB disruption is exclusively caveolin-1-dependent and requires rho-kinase 2 activity. Importantly, hyperoxia failed to prevent CSD-induced BBB breakdown, suggesting that the latter is independent of tissue hypoxia. INTERPRETATION: Our data elucidate the mechanisms by which CSDs lead to transient BBB disruption, with diagnostic and therapeutic implications for migraine and brain injury.


Asunto(s)
Caveolina 1/metabolismo , Endotelio/metabolismo , Pericitos/metabolismo , Transcitosis/fisiología , Animales , Barrera Hematoencefálica/metabolismo , Depresión de Propagación Cortical/genética , Depresión de Propagación Cortical/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Migrañosos/metabolismo , Uniones Estrechas/metabolismo
15.
Genes Dev ; 32(7-8): 466-478, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29692355

RESUMEN

The blood-brain barrier (BBB) restricts free access of molecules between the blood and the brain and is essential for regulating the neural microenvironment. Here, we describe how the BBB was initially characterized and how the current field evaluates barrier properties. We next detail the cellular nature of the BBB and discuss both the conservation and variation of BBB function across taxa. Finally, we examine our current understanding of mouse and zebrafish model systems, as we expect that comparison of the BBB across organisms will provide insight into the human BBB under normal physiological conditions and in neurological diseases.


Asunto(s)
Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Animales , Astrocitos/fisiología , Encéfalo/fisiología , Permeabilidad de la Membrana Celular , Células Endoteliales/metabolismo , Evolución Molecular , Humanos , Ratones , Pericitos/fisiología , Pez Cebra/metabolismo
16.
Pattern Recognit ; 63: 710-718, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28566796

RESUMEN

To simultaneously overcome the challenges imposed by the nature of optical imaging characterized by a range of artifacts including space-varying signal to noise ratio (SNR), scattered light, and non-uniform illumination, we developed a novel method that segments the 3-D vasculature directly from original fluorescence microscopy images eliminating the need for employing pre- and post-processing steps such as noise removal and segmentation refinement as used with the majority of segmentation techniques. Our method comprises two initialization and constrained recovery and enhancement stages. The initialization approach is fully automated using features derived from bi-scale statistical measures and produces seed points robust to non-uniform illumination, low SNR, and local structural variations. This algorithm achieves the goal of segmentation via design of an iterative approach that extracts the structure through voting of feature vectors formed by distance, local intensity gradient, and median measures. Qualitative and quantitative analysis of the experimental results obtained from synthetic and real data prove the effcacy of this method in comparison to the state-of-the-art enhancing-segmenting methods. The algorithmic simplicity, freedom from having a priori probabilistic information about the noise, and structural definition gives this algorithm a wide potential range of applications where i.e. structural complexity significantly complicates the segmentation problem.

17.
Neuron ; 94(3): 581-594.e5, 2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28416077

RESUMEN

The blood-brain barrier (BBB) provides a constant homeostatic brain environment that is essential for proper neural function. An unusually low rate of vesicular transport (transcytosis) has been identified as one of the two unique properties of CNS endothelial cells, relative to peripheral endothelial cells, that maintain the restrictive quality of the BBB. However, it is not known how this low rate of transcytosis is achieved. Here we provide a mechanism whereby the regulation of CNS endothelial cell lipid composition specifically inhibits the caveolae-mediated transcytotic route readily used in the periphery. An unbiased lipidomic analysis reveals significant differences in endothelial cell lipid signatures from the CNS and periphery, which underlie a suppression of caveolae vesicle formation and trafficking in brain endothelial cells. Furthermore, lipids transported by Mfsd2a establish a unique lipid environment that inhibits caveolae vesicle formation in CNS endothelial cells to suppress transcytosis and ensure BBB integrity.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Caveolas/metabolismo , Metabolismo de los Lípidos/genética , Proteínas de Transporte de Membrana/genética , Transcitosis/genética , Animales , Barrera Hematoencefálica/ultraestructura , Western Blotting , Caveolas/ultraestructura , Células Endoteliales , Células HEK293 , Humanos , Inmunohistoquímica , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Permeabilidad , Simportadores
18.
Neuron ; 93(6): 1325-1333.e3, 2017 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-28334606

RESUMEN

Blood-central nervous system (CNS) barriers partition neural tissues from the blood, providing a homeostatic environment for proper neural function. The endothelial cells that form blood-CNS barriers have specialized tight junctions and low rates of transcytosis to limit the flux of substances between blood and CNS. However, the relative contributions of these properties to CNS barrier permeability are unknown. Here, by studying functional blood-retinal barrier (BRB) formation in mice, we found that immature vessel leakage occurs entirely through transcytosis, as specialized tight junctions are functional as early as vessel entry into the CNS. A functional barrier forms only when transcytosis is gradually suppressed during development. Mutant mice with elevated or reduced levels of transcytosis have delayed or precocious sealing of the BRB, respectively. Therefore, the temporal regulation of transcytosis governs the development of a functional BRB, and suppression of transcytosis is a principal contributor for functional barrier formation.


Asunto(s)
Barrera Hematorretinal/crecimiento & desarrollo , Transcitosis/fisiología , Animales , Barrera Hematorretinal/ultraestructura , Caveolina 1/genética , Caveolina 1/fisiología , Células Endoteliales/fisiología , Femenino , Masculino , Proteínas de Transporte de Membrana/biosíntesis , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/fisiología , Ratones , Ratones Noqueados , Simportadores , Uniones Estrechas/genética , Uniones Estrechas/fisiología , Transcitosis/genética
19.
Elife ; 52016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26910011

RESUMEN

Vascular network density determines the amount of oxygen and nutrients delivered to host tissues, but how the vast diversity of densities is generated is unknown. Reiterations of endothelial-tip-cell selection, sprout extension and anastomosis are the basis for vascular network generation, a process governed by the VEGF/Notch feedback loop. Here, we find that temporal regulation of this feedback loop, a previously unexplored dimension, is the key mechanism to determine vascular density. Iterating between computational modeling and in vivo live imaging, we demonstrate that the rate of tip-cell selection determines the length of linear sprout extension at the expense of branching, dictating network density. We provide the first example of a host tissue-derived signal (Semaphorin3E-Plexin-D1) that accelerates tip cell selection rate, yielding a dense network. We propose that temporal regulation of this critical, iterative aspect of network formation could be a general mechanism, and additional temporal regulators may exist to sculpt vascular topology.


Asunto(s)
Proliferación Celular , Células Endoteliales/fisiología , Neovascularización Fisiológica , Receptores Notch/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Simulación por Computador , Ratones Endogámicos C57BL , Ratones Noqueados , Imagen Óptica
20.
Trends Neurosci ; 38(10): 598-608, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26442694

RESUMEN

The blood-brain barrier (BBB) maintains the optimal microenvironment in the central nervous system (CNS) for proper brain function. The BBB comprises specialized CNS endothelial cells with fundamental molecular properties essential for the function and integrity of the BBB. The restrictive nature of the BBB hinders the delivery of therapeutics for many neurological disorders. In addition, recent evidence shows that BBB dysfunction can precede or hasten the progression of several neurological diseases. Despite the physiological significance of the BBB in health and disease, major discoveries of the molecular regulators of BBB formation and function have occurred only recently. This review highlights recent findings describing the molecular determinants and core cellular pathways that confer BBB properties on CNS endothelial cells.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...