Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 554(7693): 475-480, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29443965

RESUMO

Cerebrovascular disease is the third most common cause of death in developed countries, but our understanding of the cells that compose the cerebral vasculature is limited. Here, using vascular single-cell transcriptomics, we provide molecular definitions for the principal types of blood vascular and vessel-associated cells in the adult mouse brain. We uncover the transcriptional basis of the gradual phenotypic change (zonation) along the arteriovenous axis and reveal unexpected cell type differences: a seamless continuum for endothelial cells versus a punctuated continuum for mural cells. We also provide insight into pericyte organotypicity and define a population of perivascular fibroblast-like cells that are present on all vessel types except capillaries. Our work illustrates the power of single-cell transcriptomics to decode the higher organizational principles of a tissue and may provide the initial chapter in a molecular encyclopaedia of the mammalian vasculature.


Assuntos
Vasos Sanguíneos/citologia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Células Endoteliais/classificação , Animais , Artérias/citologia , Arteríolas/citologia , Capilares/citologia , Feminino , Fibroblastos/classificação , Masculino , Camundongos , Miócitos de Músculo Liso/classificação , Especificidade de Órgãos , Pericitos/classificação , Análise de Célula Única , Transcriptoma , Veias/citologia
2.
Nature ; 560(7716): E3, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29925939

RESUMO

In Fig. 1b of this Article, 'Csf1r' was misspelt 'Csfr1'. In addition, in Extended Data Fig. 11b, owing to an error during figure formatting, the genes listed in the first column shifted down three rows below the first gene on the list, causing a mismatch between the gene names and their characteristics. These errors have been corrected online, and the original Extended Data Fig. 11b is provided as Supplementary Information to the accompanying Amendment.

3.
Circ Res ; 128(4): e46-e62, 2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33375813

RESUMO

RATIONALE: Pericytes are capillary mural cells playing a role in stabilizing newly formed blood vessels during development and tissue repair. Loss of pericytes has been described in several brain disorders, and genetically induced pericyte deficiency in the brain leads to increased macromolecular leakage across the blood-brain barrier (BBB). However, the molecular details of the endothelial response to pericyte deficiency remain elusive. OBJECTIVE: To map the transcriptional changes in brain endothelial cells resulting from lack of pericyte contact at single-cell level and to correlate them with regional heterogeneities in BBB function and vascular phenotype. METHODS AND RESULTS: We reveal transcriptional, morphological, and functional consequences of pericyte absence for brain endothelial cells using a combination of methodologies, including single-cell RNA sequencing, tracer analyses, and immunofluorescent detection of protein expression in pericyte-deficient adult Pdgfbret/ret mice. We find that endothelial cells without pericyte contact retain a general BBB-specific gene expression profile, however, they acquire a venous-shifted molecular pattern and become transformed regarding the expression of numerous growth factors and regulatory proteins. Adult Pdgfbret/ret brains display ongoing angiogenic sprouting without concomitant cell proliferation providing unique insights into the endothelial tip cell transcriptome. We also reveal heterogeneous modes of pericyte-deficient BBB impairment, where hotspot leakage sites display arteriolar-shifted identity and pinpoint putative BBB regulators. By testing the causal involvement of some of these using reverse genetics, we uncover a reinforcing role for angiopoietin 2 at the BBB. CONCLUSIONS: By elucidating the complexity of endothelial response to pericyte deficiency at cellular resolution, our study provides insight into the importance of brain pericytes for endothelial arterio-venous zonation, angiogenic quiescence, and a limited set of BBB functions. The BBB-reinforcing role of ANGPT2 (angiopoietin 2) is paradoxical given its wider role as TIE2 (TEK receptor tyrosine kinase) receptor antagonist and may suggest a unique and context-dependent function of ANGPT2 in the brain.


Assuntos
Barreira Hematoencefálica/metabolismo , Pericitos/citologia , Animais , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/patologia , Proliferação de Células , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Linfocinas/deficiência , Linfocinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Pericitos/metabolismo , Pericitos/patologia , Fator de Crescimento Derivado de Plaquetas/deficiência , Fator de Crescimento Derivado de Plaquetas/genética , Análise de Célula Única , Transcriptoma
4.
Nature ; 532(7599): 380-4, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-27074508

RESUMO

Blood vessels define local microenvironments in the skeletal system, play crucial roles in osteogenesis and provide niches for haematopoietic stem cells. The properties of niche-forming vessels and their changes in the ageing organism remain incompletely understood. Here we show that Notch signalling in endothelial cells leads to the expansion of haematopoietic stem cell niches in bone, which involves increases in CD31-positive capillaries and platelet-derived growth factor receptor-ß (PDGFRß)-positive perivascular cells, arteriole formation and elevated levels of cellular stem cell factor. Although endothelial hypoxia-inducible factor signalling promotes some of these changes, it fails to enhance vascular niche function because of a lack of arterialization and expansion of PDGFRß-positive cells. In ageing mice, niche-forming vessels in the skeletal system are strongly reduced but can be restored by activation of endothelial Notch signalling. These findings indicate that vascular niches for haematopoietic stem cells are part of complex, age-dependent microenvironments involving multiple cell populations and vessel subtypes.


Assuntos
Envelhecimento/fisiologia , Arteríolas/fisiologia , Osso e Ossos/irrigação sanguínea , Capilares/fisiologia , Células-Tronco Hematopoéticas/citologia , Nicho de Células-Tronco , Animais , Arteríolas/citologia , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Capilares/citologia , Contagem de Células , Células Endoteliais/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Osteogênese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Fator de Células-Tronco/metabolismo
5.
Development ; 144(19): 3590-3601, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28851707

RESUMO

Tissue fluid drains through blind-ended lymphatic capillaries, via smooth muscle cell (SMC)-covered collecting vessels into venous circulation. Both defective SMC recruitment to collecting vessels and ectopic recruitment to lymphatic capillaries are thought to contribute to vessel failure, leading to lymphedema. However, mechanisms controlling lymphatic SMC recruitment and its role in vessel maturation are unknown. Here, we demonstrate that platelet-derived growth factor B (PDGFB) regulates lymphatic SMC recruitment in multiple vascular beds. PDGFB is selectively expressed by lymphatic endothelial cells (LECs) of collecting vessels. LEC-specific deletion of Pdgfb prevented SMC recruitment causing dilation and failure of pulsatile contraction of collecting vessels. However, vessel remodelling and identity were unaffected. Unexpectedly, Pdgfb overexpression in LECs did not induce SMC recruitment to capillaries. This was explained by the demonstrated requirement of PDGFB extracellular matrix (ECM) retention for lymphatic SMC recruitment, and the low presence of PDGFB-binding ECM components around lymphatic capillaries. These results demonstrate the requirement of LEC-autonomous PDGFB expression and retention for SMC recruitment to lymphatic vessels, and suggest an ECM-controlled checkpoint that prevents SMC investment of capillaries, which is a common feature in lymphedematous skin.


Assuntos
Células Endoteliais/metabolismo , Vasos Linfáticos/anatomia & histologia , Vasos Linfáticos/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Animais , Animais Recém-Nascidos , Capilares/metabolismo , Comunicação Celular , Derme/metabolismo , Matriz Extracelular/metabolismo , Feminino , Membro Posterior/metabolismo , Masculino , Mesentério/metabolismo , Morfogênese , Tamanho do Órgão
8.
Arterioscler Thromb Vasc Biol ; 35(2): 409-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25477343

RESUMO

OBJECTIVE: Vascular smooth muscle cells (VSMC) are important for contraction, blood flow distribution, and regulation of blood vessel diameter, but to what extent they contribute to the integrity of blood vessels and blood-brain barrier function is less well understood. In this report, we explored the impact of the loss of VSMC in the Notch3(-/-) mouse on blood vessel integrity in the central nervous system. APPROACH AND RESULTS: Notch3(-/-) mice showed focal disruptions of the blood-brain barrier demonstrated by extravasation of tracers accompanied by fibrin deposition in the retinal vasculature. This blood-brain barrier leakage was accompanied by a regionalized and patchy loss of VSMC, with VSMC gaps predominantly in arterial resistance vessels of larger caliber. The loss of VSMC appeared to be caused by progressive degeneration of VSMC resulting in a gradual loss of VSMC marker expression and a progressive acquisition of an aberrant VSMC phenotype closer to the gaps, followed by enhanced apoptosis and cellular disintegration in the gaps. Arterial VSMC were the only mural cell type that was morphologically affected, despite Notch3 also being expressed in pericytes. Transcriptome analysis of isolated brain microvessels revealed gene expression changes in Notch3(-/-) mice consistent with loss of arterial VSMC and presumably secondary transcriptional changes were observed in endothelial genes, which may explain the compromised vascular integrity. CONCLUSIONS: We demonstrate that Notch3 is important for survival of VSMC, and reveal a critical role for Notch3 and VSMC in blood vessel integrity and blood-brain barrier function in the mammalian vasculature.


Assuntos
Barreira Hematoencefálica/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores Notch/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Apoptose , Biomarcadores/metabolismo , Vasos Sanguíneos/metabolismo , Barreira Hematoencefálica/patologia , Permeabilidade Capilar , Células Endoteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvasos/metabolismo , Microvasos/patologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Pericitos/metabolismo , Fenótipo , Receptor Notch3 , Receptores Notch/deficiência , Receptores Notch/genética , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Transdução de Sinais , Transcrição Gênica
9.
Nature ; 468(7323): 557-61, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20944627

RESUMO

The blood-brain barrier (BBB) consists of specific physical barriers, enzymes and transporters, which together maintain the necessary extracellular environment of the central nervous system (CNS). The main physical barrier is found in the CNS endothelial cell, and depends on continuous complexes of tight junctions combined with reduced vesicular transport. Other possible constituents of the BBB include extracellular matrix, astrocytes and pericytes, but the relative contribution of these different components to the BBB remains largely unknown. Here we demonstrate a direct role of pericytes at the BBB in vivo. Using a set of adult viable pericyte-deficient mouse mutants we show that pericyte deficiency increases the permeability of the BBB to water and a range of low-molecular-mass and high-molecular-mass tracers. The increased permeability occurs by endothelial transcytosis, a process that is rapidly arrested by the drug imatinib. Furthermore, we show that pericytes function at the BBB in at least two ways: by regulating BBB-specific gene expression patterns in endothelial cells, and by inducing polarization of astrocyte end-feet surrounding CNS blood vessels. Our results indicate a novel and critical role for pericytes in the integration of endothelial and astrocyte functions at the neurovascular unit, and in the regulation of the BBB.


Assuntos
Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Pericitos/metabolismo , Animais , Astrócitos/metabolismo , Benzamidas , Sistema Nervoso Central/irrigação sanguínea , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Mesilato de Imatinib , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Transcitose/efeitos dos fármacos
10.
Cell Rep ; 43(3): 113911, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38446668

RESUMO

Claudin-5 (CLDN5) is an endothelial tight junction protein essential for blood-brain barrier (BBB) formation. Abnormal CLDN5 expression is common in brain disease, and knockdown of Cldn5 at the BBB has been proposed to facilitate drug delivery to the brain. To study the consequences of CLDN5 loss in the mature brain, we induced mosaic endothelial-specific Cldn5 gene ablation in adult mice (Cldn5iECKO). These mice displayed increased BBB permeability to tracers up to 10 kDa in size from 6 days post induction (dpi) and ensuing lethality from 10 dpi. Single-cell RNA sequencing at 11 dpi revealed profound transcriptomic differences in brain endothelial cells regardless of their Cldn5 status in mosaic mice, suggesting major non-cell-autonomous responses. Reactive microglia and astrocytes suggested rapid cellular responses to BBB leakage. Our study demonstrates a critical role for CLDN5 in the adult BBB and provides molecular insight into the consequences and risks associated with CLDN5 inhibition.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Animais , Camundongos , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Claudina-5/genética , Claudina-5/metabolismo , Células Endoteliais/metabolismo
11.
J Exp Med ; 221(2)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38117255

RESUMO

In dorsal root ganglia (DRG), macrophages reside close to sensory neurons and have largely been explored in the context of pain, nerve injury, and repair. However, we discovered that most DRG macrophages interact with and monitor the vasculature by sampling macromolecules from the blood. Characterization of the DRG vasculature revealed a specialized endothelial bed that transformed in molecular, structural, and permeability properties along the arteriovenous axis and was covered by macrophage-interacting pericytes and fibroblasts. Macrophage phagocytosis spatially aligned with peak endothelial permeability, a process regulated by enhanced caveolar transcytosis in endothelial cells. Profiling the DRG immune landscape revealed two subsets of perivascular macrophages with distinct transcriptome, turnover, and function. CD163+ macrophages self-maintained locally, specifically participated in vasculature monitoring, displayed distinct responses during peripheral inflammation, and were conserved in mouse and man. Our work provides a molecular explanation for the permeability of the blood-DRG barrier and identifies an unappreciated role of macrophages as integral components of the DRG-neurovascular unit.


Assuntos
Células Endoteliais , Gânglios Espinais , Humanos , Macrófagos , Pericitos , Permeabilidade
12.
Sci Rep ; 13(1): 15022, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37699967

RESUMO

The heart depends on a functional vasculature for oxygenation and transport of nutrients, and it is of interest to learn how primary impairment of the vasculature can indirectly affect cardiac function and heart morphology. Notch3-deficiency causes vascular smooth muscle cell (VSMC) loss in the vasculature but the consequences for the heart remain largely elusive. Here, we demonstrate that Notch3-/- mice have enlarged hearts with left ventricular hypertrophy and mild fibrosis. Cardiomyocytes were hypertrophic but not hyperproliferative, and the expression of several cardiomyocyte markers, including Tnt2, Myh6, Myh7 and Actn2, was altered. Furthermore, expression of genes regulating the metabolic status of the heart was affected: both Pdk4 and Cd36 were downregulated, indicating a metabolic switch from fatty acid oxidation to glucose consumption. Notch3-/- mice furthermore showed lower liver lipid content. Notch3 was expressed in heart VSMC and pericytes but not in cardiomyocytes, suggesting that a perturbation of Notch signalling in VSMC and pericytes indirectly impairs the cardiomyocytes. In keeping with this, Pdgfbret/ret mice, characterized by reduced numbers of VSMC and pericytes, showed left ventricular and cardiomyocyte hypertrophy. In conclusion, we demonstrate that reduced Notch3 or PDGFB signalling in vascular mural cells leads to cardiomyocyte dysfunction.


Assuntos
Cardiomegalia , Hipertrofia Ventricular Esquerda , Animais , Camundongos , Becaplermina , Metabolismo dos Lipídeos , Miócitos Cardíacos , Proteínas Proto-Oncogênicas c-sis
13.
Cell Rep Methods ; 3(3): 100431, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-37056377

RESUMO

Studying disease-related changes in the brain vasculature is warranted due to its crucial role in supplying oxygen and nutrients and removing waste and due to the anticipated vascular dysfunction in brain diseases. To this end, we have developed a protocol for fast and simple isolation of brain vascular fragments without the use of transgenic reporters. We used it to isolate and analyze 22,515 cells by single-cell RNA sequencing. The cells distributed into 23 distinct clusters corresponding to all known vascular and perivascular cell types in the brain. Western blot analysis also suggested that the protocol is suitable for proteomic analysis. We further adapted it for the establishment of primary cell cultures. The protocol generated highly reproducible results. In conclusion, we have developed a simple and robust brain vascular isolation protocol suitable for different experimental modalities, such as single-cell analyses, western blotting, and primary cell culture.


Assuntos
Sistema Cardiovascular , Proteômica , Camundongos , Animais , Encéfalo/irrigação sanguínea , Células Cultivadas
14.
Neuron ; 111(23): 3745-3764.e7, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37776854

RESUMO

Leptomeninges, consisting of the pia mater and arachnoid, form a connective tissue investment and barrier enclosure of the brain. The exact nature of leptomeningeal cells has long been debated. In this study, we identify five molecularly distinct fibroblast-like transcriptomes in cerebral leptomeninges; link them to anatomically distinct cell types of the pia, inner arachnoid, outer arachnoid barrier, and dural border layer; and contrast them to a sixth fibroblast-like transcriptome present in the choroid plexus and median eminence. Newly identified transcriptional markers enabled molecular characterization of cell types responsible for adherence of arachnoid layers to one another and for the arachnoid barrier. These markers also proved useful in identifying the molecular features of leptomeningeal development, injury, and repair that were preserved or changed after traumatic brain injury. Together, the findings highlight the value of identifying fibroblast transcriptional subsets and their cellular locations toward advancing the understanding of leptomeningeal physiology and pathology.


Assuntos
Aracnoide-Máter , Meninges , Camundongos , Animais , Aracnoide-Máter/anatomia & histologia , Pia-Máter , Plexo Corióideo , Encéfalo
15.
Biochim Biophys Acta ; 1808(6): 1544-51, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21070744

RESUMO

Cell-penetrating peptides (CPPs) are a promising group of delivery vectors for various therapeutic agents but their application is often hampered by poor stability in the presence of serum. Different strategies to improve peptide stability have been exploited, one of them being "retro-inversion" (RI) of natural peptides. With this approach the stability of CPPs has been increased, thereby making them more efficient transporters. Several RI-CPPs were here assessed and compared to the corresponding parent peptides in different cell-lines. Surprisingly, treatment of cells with these peptides induced trypsin insensitivity and rapid severe toxicity in contrast to L-peptides. This was measured as reduced metabolic activity and condensed cell nuclei, in parity with the apoptosis inducing agent staurosporine. Furthermore, effects on mitochondrial network, focal adhesions, actin cytoskeleton and caspase-3 activation were analyzed and adverse effects were evident at 20 µM peptide concentration within 4 h while parent L-peptides had negligible effects. To our knowledge this is the first time RI peptides are reported to cause cellular toxicity, displayed by decreased metabolic activity, morphological changes and induction of apoptosis. Considering the wide range of research areas that involves the use of RI-peptides, this finding is of major importance and needs to be taken under consideration in applications of RI-peptides.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Peptídeos/farmacologia , Sequência de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Peptídeos Penetradores de Células , Relação Dose-Resposta a Droga , Fluoresceínas/química , Produtos do Gene tat/química , Produtos do Gene tat/farmacologia , Células HeLa , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Peptídeos/química
16.
Mol Ther ; 19(8): 1457-67, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21343913

RESUMO

Finding suitable nonviral delivery vehicles for nucleic acid-based therapeutics is a landmark goal in gene therapy. Cell-penetrating peptides (CPPs) are one class of delivery vectors that has been exploited for this purpose. However, since CPPs use endocytosis to enter cells, a large fraction of peptides remain trapped in endosomes. We have previously reported that stearylation of amphipathic CPPs, such as transportan 10 (TP10), dramatically increases transfection of oligonucleotides in vitro partially by promoting endosomal escape. Therefore, we aimed to evaluate whether stearyl-TP10 could be used for the delivery of plasmids as well. Our results demonstrate that stearyl-TP10 forms stable nanoparticles with plasmids that efficiently enter different cell-types in a ubiquitous manner, including primary cells, resulting in significantly higher gene expression levels than when using stearyl-Arg9 or unmodified CPPs. In fact, the transfection efficacy of stearyl-TP10 almost reached the levels of Lipofectamine 2000 (LF2000), however, without any of the observed lipofection-associated toxicities. Most importantly, stearyl-TP10/plasmid nanoparticles are nonimmunogenic, mediate efficient gene delivery in vivo, when administrated intramuscularly (i.m.) or intradermally (i.d.) without any associated toxicity in mice.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Plasmídeos/metabolismo , Transfecção/métodos , Animais , Transporte Biológico , Linhagem Celular , Cricetinae , Cricetulus , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Endossomos/metabolismo , Terapia Genética/métodos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ácidos Nucleicos/metabolismo
17.
J Cereb Blood Flow Metab ; 42(2): 264-279, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34689641

RESUMO

Platelet-derived growth factor B (PDGFB) released from endothelial cells is indispensable for pericyte recruitment during angiogenesis in embryonic and postnatal organ growth. Constitutive genetic loss-of-function of PDGFB leads to pericyte hypoplasia and the formation of a sparse, dilated and venous-shifted brain microvasculature with dysfunctional blood-brain barrier (BBB) in mice, as well as the formation of microvascular calcification in both mice and humans. Endothelial PDGFB is also expressed in the adult quiescent microvasculature, but here its importance is unknown. We show that deletion of Pdgfb in endothelial cells in 2-months-old mice causes a slowly progressing pericyte loss leading, at 12-18 months of age, to ≈50% decrease in endothelial:pericyte cell ratio, ≈60% decrease in pericyte longitudinal capillary coverage and >70% decrease in pericyte marker expression. Similar to constitutive loss of Pdgfb, this correlates with increased BBB permeability. However, in contrast to the constitutive loss of Pdgfb, adult-induced loss does not lead to vessel dilation, impaired arterio-venous zonation or the formation of microvascular calcifications. We conclude that PDFGB expression in quiescent adult microvascular brain endothelium is critical for the maintenance of pericyte coverage and normal BBB function, but that microvessel dilation, rarefaction, arterio-venous skewing and calcification reflect developmental roles of PDGFB.


Assuntos
Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar , Endotélio Vascular/metabolismo , Linfocinas/metabolismo , Pericitos/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Calcificação Vascular/metabolismo , Animais , Barreira Hematoencefálica/patologia , Endotélio Vascular/patologia , Regulação da Expressão Gênica , Linfocinas/genética , Camundongos , Camundongos Knockout , Pericitos/patologia , Fator de Crescimento Derivado de Plaquetas/genética , Calcificação Vascular/genética , Calcificação Vascular/patologia
18.
Stem Cell Reports ; 17(5): 1089-1104, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35452595

RESUMO

Humanized mouse models and mouse-adapted SARS-CoV-2 virus are increasingly used to study COVID-19 pathogenesis, so it is important to learn where the SARS-CoV-2 receptor ACE2 is expressed. Here we mapped ACE2 expression during mouse postnatal development and in adulthood. Pericytes in the CNS, heart, and pancreas express ACE2 strongly, as do perineurial and adrenal fibroblasts, whereas endothelial cells do not at any location analyzed. In a number of other organs, pericytes do not express ACE2, including in the lung where ACE2 instead is expressed in bronchial epithelium and alveolar type II cells. The onset of ACE2 expression is organ specific: in bronchial epithelium already at birth, in brain pericytes before, and in heart pericytes after postnatal day 10.5. Establishing the vascular localization of ACE2 expression is central to correctly interpret data from modeling COVID-19 in the mouse and may shed light on the cause of vascular COVID-19 complications.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Pericitos , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , COVID-19/complicações , Doenças Cardiovasculares/virologia , Células Endoteliais , Camundongos , Pericitos/metabolismo , SARS-CoV-2
19.
Nat Commun ; 13(1): 2003, 2022 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-35422069

RESUMO

Blood-brain barrier (BBB) dysfunction is associated with worse epilepsy outcomes however the underlying molecular mechanisms of BBB dysfunction remain to be elucidated. Tight junction proteins are important regulators of BBB integrity and in particular, the tight junction protein claudin-5 is the most enriched in brain endothelial cells and regulates size-selectivity at the BBB. Additionally, disruption of claudin-5 expression has been implicated in numerous disorders including schizophrenia, depression and traumatic brain injury, yet its role in epilepsy has not been fully deciphered. Here we report that claudin-5 protein levels are significantly diminished in surgically resected brain tissue from patients with treatment-resistant epilepsy. Concomitantly, dynamic contrast-enhanced MRI in these patients showed widespread BBB disruption. We show that targeted disruption of claudin-5 in the hippocampus or genetic heterozygosity of claudin-5 in mice exacerbates kainic acid-induced seizures and BBB disruption. Additionally, inducible knockdown of claudin-5 in mice leads to spontaneous recurrent seizures, severe neuroinflammation, and mortality. Finally, we identify that RepSox, a regulator of claudin-5 expression, can prevent seizure activity in experimental epilepsy. Altogether, we propose that BBB stabilizing drugs could represent a new generation of agents to prevent seizure activity in epilepsy patients.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Animais , Barreira Hematoencefálica/metabolismo , Claudina-5/genética , Claudina-5/metabolismo , Células Endoteliais/metabolismo , Humanos , Camundongos , Convulsões/metabolismo , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/metabolismo
20.
Brain Pathol ; 30(3): 446-464, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31561281

RESUMO

Primary familial brain calcification (PFBC) is an age-dependent and rare neurodegenerative disorder characterized by microvascular calcium phosphate deposits in the deep brain regions. Known genetic causes of PFBC include loss-of-function mutations in genes involved in either of three processes-platelet-derived growth factor (PDGF) signaling, phosphate homeostasis or protein glycosylation-with unclear molecular links. To provide insight into the pathogenesis of PFBC, we analyzed murine models of PFBC for the first two of these processes in Pdgfbret/ret and Slc20a2-/- mice with regard to the structure, molecular composition, development and distribution of perivascular calcified nodules. Analyses by transmission electron microscopy and immunofluorescence revealed that calcified nodules in both of these models have a multilayered ultrastructure and occur in direct contact with reactive astrocytes and microglia. However, whereas nodules in Pdgfbret/ret mice were large, solitary and smooth surfaced, the nodules in Slc20a2-/- mice were multi-lobulated and occurred in clusters. The regional distribution of nodules also differed between the two models. Proteomic analysis and immunofluorescence stainings revealed a common molecular composition of the nodules in the two models, involving proteins implicated in bone homeostasis, but also proteins not previously linked to tissue mineralization. While the brain vasculature of Pdgfbret/ret mice has been reported to display reduced pericyte coverage and abnormal permeability, we found that Slc20a2-/- mice have a normal pericyte coverage and no overtly increased permeability. Thus, lack of pericytes and increase in permeability of the blood-brain barrier are likely not the causal triggers for PFBC pathogenesis. Instead, gene expression and spatial correlations suggest that astrocytes are intimately linked to the calcification process in PFBC.


Assuntos
Astrócitos/metabolismo , Encefalopatias/metabolismo , Calcinose/metabolismo , Matriz Extracelular/metabolismo , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo , Animais , Astrócitos/patologia , Encefalopatias/genética , Encefalopatias/patologia , Calcinose/genética , Calcinose/patologia , Modelos Animais de Doenças , Progressão da Doença , Matriz Extracelular/patologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Microglia/patologia , Mutação , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA