Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 145(13)2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29853619

RESUMO

Formation and homeostasis of the vascular system requires several coordinated cellular functions, but their precise interplay during development and their relative importance for vascular pathologies remain poorly understood. Here, we investigated the endothelial functions regulated by Cdc42 and their in vivo relevance during angiogenic sprouting and vascular morphogenesis in the postnatal mouse retina. We found that Cdc42 is required for endothelial tip cell selection, directed cell migration and filopodia formation, but dispensable for cell proliferation or apoptosis. Although the loss of Cdc42 seems generally compatible with apical-basal polarization and lumen formation in retinal blood vessels, it leads to defective endothelial axial polarization and to the formation of severe vascular malformations in capillaries and veins. Tracking of Cdc42-depleted endothelial cells in mosaic retinas suggests that these capillary-venous malformations arise as a consequence of defective cell migration, when endothelial cells that proliferate at normal rates are unable to re-distribute within the vascular network.


Assuntos
Capilares/anormalidades , Movimento Celular , Células Endoteliais/metabolismo , Veia Retiniana/anormalidades , Malformações Vasculares/embriologia , Proteína cdc42 de Ligação ao GTP/deficiência , Animais , Capilares/embriologia , Polaridade Celular/genética , Células Endoteliais/patologia , Camundongos , Camundongos Knockout , Pseudópodes/genética , Pseudópodes/metabolismo , Veia Retiniana/embriologia , Malformações Vasculares/genética , Malformações Vasculares/patologia
2.
Development ; 141(23): 4489-99, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25406396

RESUMO

Vascular development of the central nervous system and blood-brain barrier (BBB) induction are closely linked processes. The role of factors that promote endothelial sprouting and vascular leak, such as vascular endothelial growth factor A, are well described, but the factors that suppress angiogenic sprouting and their impact on the BBB are poorly understood. Here, we show that integrin αVß8 activates angiosuppressive TGFß gradients in the brain, which inhibit endothelial cell sprouting. Loss of αVß8 in the brain or downstream TGFß1-TGFBR2-ALK5-Smad3 signaling in endothelial cells increases vascular sprouting, branching and proliferation, leading to vascular dysplasia and hemorrhage. Importantly, BBB function in Itgb8 mutants is intact during early stages of vascular dysgenesis before hemorrhage. By contrast, Pdgfb(ret/ret) mice, which exhibit severe BBB disruption and vascular leak due to pericyte deficiency, have comparatively normal vascular morphogenesis and do not exhibit brain hemorrhage. Our data therefore suggest that abnormal vascular sprouting and patterning, not BBB dysfunction, underlie developmental cerebral hemorrhage.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/irrigação sanguínea , Hemorragia Cerebral/etiologia , Neovascularização Patológica/complicações , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Encéfalo/metabolismo , Contagem de Células , Células Endoteliais/fisiologia , Imuno-Histoquímica , Integrinas/metabolismo , Camundongos , Microscopia Confocal , Fator de Crescimento Transformador beta/metabolismo
3.
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
4.
Handb Exp Pharmacol ; 234: 351-368, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27832496

RESUMO

The cardiovascular system in adult organisms forms a network of interconnected endothelial cells, supported by mural cells and displaying a high degree of hierarchy: arteries emerging from the heart ramify into arterioles and then capillaries, which return to the venous systems through venules and veins. The cardiovascular system allows blood circulation, which in turn is essential for hemostasis through gas diffusion, nutrient distribution, and cell trafficking. In this chapter, we have summarized the current knowledge on how adhesion GPCRs (aGPCRs) impact heart development, followed by their role in modulating vascular angiogenesis.


Assuntos
Barreira Hematoencefálica/metabolismo , Adesão Celular , Membrana Celular/metabolismo , Coração/crescimento & desenvolvimento , Neovascularização Fisiológica , Receptores Acoplados a Proteínas G/metabolismo , Animais , Sítios de Ligação , Barreira Hematoencefálica/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Relação Estrutura-Atividade
5.
Arterioscler Thromb Vasc Biol ; 34(9): 2068-77, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24925974

RESUMO

OBJECTIVE: Using a multi-tissue, genome-wide gene expression approach, we recently identified a gene module linked to the extent of human atherosclerosis. This atherosclerosis module was enriched with inherited risk for coronary and carotid artery disease (CAD) and overlapped with genes in the transendothelial migration of leukocyte (TEML) pathway. Among the atherosclerosis module genes, the transcription cofactor Lim domain binding 2 (LDB2) was the most connected in a CAD vascular wall regulatory gene network. Here, we used human genomics and atherosclerosis-prone mice to evaluate the possible role of LDB2 in TEML and atherosclerosis. APPROACH AND RESULTS: mRNA profiles generated from blood macrophages in patients with CAD were used to infer transcription factor regulatory gene networks; Ldlr(-/-)Apob(100/100) mice were used to study the effects of Ldb2 deficiency on TEML activity and atherogenesis. LDB2 was the most connected gene in a transcription factor regulatory network inferred from TEML and atherosclerosis module genes in CAD macrophages. In Ldlr(-/-)Apob(100/100) mice, loss of Ldb2 increased atherosclerotic lesion size ≈2-fold and decreased plaque stability. The exacerbated atherosclerosis was caused by increased TEML activity, as demonstrated in air-pouch and retinal vasculature models in vivo, by ex vivo perfusion of primary leukocytes, and by leukocyte migration in vitro. In THP1 cells, migration was increased by overexpression and decreased by small interfering RNA inhibition of LDB2. A functional LDB2 variant (rs10939673) was associated with the risk and extent of CAD across several cohorts. CONCLUSIONS: As a key driver of the TEML pathway in CAD macrophages, LDB2 is a novel candidate to target CAD by inhibiting the overall activity of TEML.


Assuntos
Aterosclerose/fisiopatologia , Doenças das Artérias Carótidas/patologia , Quimiotaxia de Leucócito/fisiologia , Doença da Artéria Coronariana/patologia , Proteínas com Domínio LIM/fisiologia , Fatores de Transcrição/fisiologia , Migração Transendotelial e Transepitelial/fisiologia , Animais , Apolipoproteína B-100/genética , Doenças das Artérias Carótidas/genética , Linhagem Celular Tumoral , Quimiocina CCL2/farmacologia , Doença da Artéria Coronariana/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Proteínas com Domínio LIM/deficiência , Proteínas com Domínio LIM/genética , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , RNA Mensageiro/biossíntese , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Migração Transendotelial e Transepitelial/genética
6.
EMBO Mol Med ; 15(5): e16645, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-36912000

RESUMO

Sphingosine-1-phosphate (S1P), the circulating HDL-bound lipid mediator that acts via S1P receptors (S1PR), is required for normal vascular development. The role of this signaling axis in vascular retinopathies is unclear. Here, we show in a mouse model of oxygen-induced retinopathy (OIR) that endothelial overexpression of S1pr1 suppresses while endothelial knockout of S1pr1 worsens neovascular tuft formation. Furthermore, neovascular tufts are increased in Apom-/- mice which lack HDL-bound S1P while they are suppressed in ApomTG mice which have more circulating HDL-S1P. These results suggest that circulating HDL-S1P activation of endothelial S1PR1 suppresses neovascular pathology in OIR. Additionally, systemic administration of ApoM-Fc-bound S1P or a small-molecule Gi-biased S1PR1 agonist suppressed neovascular tuft formation. Circulating HDL-S1P activation of endothelial S1PR1 may be a key protective mechanism to guard against neovascular retinopathies that occur not only in premature infants but also in diabetic patients and aging people.


Assuntos
Neovascularização Retiniana , Camundongos , Animais , Receptores de Esfingosina-1-Fosfato , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/agonistas , Lipoproteínas HDL , Esfingosina , Lisofosfolipídeos
7.
Elife ; 112022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36197001

RESUMO

Serine palmitoyl transferase (SPT), the rate-limiting enzyme in the de novo synthesis of sphingolipids (SL), is needed for embryonic development, physiological homeostasis, and response to stress. The functions of de novo SL synthesis in vascular endothelial cells (EC), which line the entire circulatory system, are not well understood. Here, we show that the de novo SL synthesis in EC not only regulates vascular development but also maintains circulatory and peripheral organ SL levels. Mice with an endothelial-specific gene knockout of SPTLC1 (Sptlc1 ECKO), an essential subunit of the SPT complex, exhibited reduced EC proliferation and tip/stalk cell differentiation, resulting in delayed retinal vascular development. In addition, Sptlc1 ECKO mice had reduced retinal neovascularization in the oxygen-induced retinopathy model. Mechanistic studies suggest that EC SL produced from the de novo pathway are needed for lipid raft formation and efficient VEGF signaling. Post-natal deletion of the EC Sptlc1 also showed rapid reduction of several SL metabolites in plasma, red blood cells, and peripheral organs (lung and liver) but not in the retina, part of the central nervous system (CNS). In the liver, EC de novo SL synthesis was important for acetaminophen-induced rapid ceramide elevation and hepatotoxicity. These results suggest that EC-derived SL metabolites are in constant flux between the vasculature, circulatory elements, and parenchymal cells of non-CNS organs. Taken together, our data point to the central role of the endothelial SL biosynthesis in maintaining vascular development, neovascular proliferation, non-CNS tissue metabolic homeostasis, and hepatocyte response to stress.


Assuntos
Serina C-Palmitoiltransferase , Esfingolipídeos , Animais , Camundongos , Acetaminofen , Ceramidas , Células Endoteliais/metabolismo , Homeostase , Oxigênio , Serina , Serina C-Palmitoiltransferase/genética , Esfingolipídeos/metabolismo , Fator A de Crescimento do Endotélio Vascular
8.
Mutat Res ; 704(1-3): 61-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20117234

RESUMO

For decades, DNA has been considered as the main cellular target of deleterious effects of ionizing radiation (IR). Nevertheless, molecular signals initiated at cellular membranes are now identified as critical events in a large spectrum of radiation-induced cellular processes. If IR provokes DNA damage directly by energy deposit on the DNA double helix and indirectly by reactive species, origin of IR-induced molecular events initiated at the plasma membrane remains more obscure. Generation of reactive oxygen/nitrogen species (ROS/RNS) inducing proteins and lipids modifications seems to be the prevalent hypothesis. However, spatial and temporal relocalization of proteins and/or lipids represents also potential mechanisms of cell signaling generation. In the context of an oxidative stress such as IR, the best example is the translocation of the enzyme acid sphingomyelinase (ASMase) from lysosomes to the outer layer of cell membrane, which then induces sphingomyelin hydrolysis and ceramide formation. Ceramide coalescence with cholesterol forms lipids microdomains in the plasma membrane, enhancing clustering of signaling receptors (death receptors like FAS, TNF, CD40, TRAIL or G protein-coupled receptors). In this manuscript, we propose to overview the different key molecular mechanisms induced at the plasma membrane after IR in perspective with their linked molecular actors.


Assuntos
Membrana Celular/efeitos da radiação , Radiação Ionizante , Transdução de Sinais/efeitos da radiação , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Ceramidas/metabolismo , Humanos , Peroxidação de Lipídeos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
9.
Dev Cell ; 52(6): 779-793.e7, 2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-32059774

RESUMO

Transcriptional mechanisms that drive angiogenesis and organotypic vascular endothelial cell specialization are poorly understood. Here, we show that retinal endothelial sphingosine 1-phosphate receptors (S1PRs), which restrain vascular endothelial growth factor (VEGF)-induced angiogenesis, spatially restrict expression of JunB, a member of the activator protein 1 (AP-1) family of transcription factors (TFs). Mechanistically, VEGF induces JunB expression at the sprouting vascular front while S1PR-dependent vascular endothelial (VE)-cadherin assembly suppresses JunB expression in the nascent vascular network, thus creating a gradient of this TF. Endothelial-specific JunB knockout mice showed diminished expression of neurovascular guidance genes and attenuated retinal vascular network progression. In addition, endothelial S1PR signaling is required for normal expression of ß-catenin-dependent genes such as TCF/LEF1 and ZIC3 TFs, transporters, and junctional proteins. These results show that S1PR signaling restricts JunB function to the expanding vascular front, thus creating an AP-1 gradient and enabling organotypic endothelial cell specialization of the vascular network.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/metabolismo , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Células Cultivadas , Montagem e Desmontagem da Cromatina , Células Endoteliais/citologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vasos Retinianos/citologia , Vasos Retinianos/embriologia , Fator de Transcrição AP-1/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Cancer Res ; 67(4): 1803-11, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17308123

RESUMO

Because of the central role of the endothelium in tissue homeostasis, protecting the vasculature from radiation-induced death is a major concern in tissue radioprotection. Premitotic apoptosis and mitotic death are two prevalent cell death pathways induced by ionizing radiation. Endothelial cells undergo apoptosis after radiation through generation of the sphingolipid ceramide. However, if mitotic death is known as the established radiation-induced death pathway for cycling eukaryotic cells, direct involvement of mitotic death in proliferating endothelial radiosensitivity has not been clearly shown. In this study, we proved that proliferating human microvascular endothelial cells (HMEC-1) undergo two waves of death after exposure to 15 Gy radiation: an early premitotic apoptosis dependent on ceramide generation and a delayed DNA damage-induced mitotic death. The fact that sphingosine-1-phosphate (S1P), a ceramide antagonist, protects HMEC-1 only from membrane-dependent apoptosis but not from DNA damage-induced mitotic death proves the independence of the two pathways. Furthermore, adding nocodazole, a mitotic inhibitor, to S1P affected both cell death mechanisms and fully prevented radiation-induced death. If our results fit with the standard model in which S1P signaling inhibits ceramide-mediated apoptosis induced by antitumor treatments, such as radiotherapy, they exclude, for the first time, a significant role of S1P-induced molecular survival pathway against mitotic death. Discrimination between ceramide-mediated apoptosis and DNA damage-induced mitotic death may give the opportunity to define a new class of radioprotectors for normal tissues in which quiescent endothelium represents the most sensitive target, while excluding malignant tumor containing pro-proliferating angiogenic endothelial cells that are sensitive to mitotic death.


Assuntos
Apoptose/efeitos dos fármacos , Ceramidas/antagonistas & inibidores , Dano ao DNA , Células Endoteliais/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Esfingosina/análogos & derivados , Apoptose/efeitos da radiação , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Processos de Crescimento Celular , Ceramidas/biossíntese , Desipramina/farmacologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/efeitos da radiação , Humanos , Mitose/efeitos dos fármacos , Mitose/fisiologia , Nocodazol/farmacologia , Esfingosina/farmacologia
11.
Nat Commun ; 9(1): 353, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29367624

RESUMO

Establishing effective central tolerance requires the promiscuous expression of tissue-restricted antigens by medullary thymic epithelial cells. However, whether central tolerance also extends to post-translationally modified proteins is not clear. Here we show a mouse model of autoimmunity in which disease development is dependent on post-translational modification (PTM) of the tissue-restricted self-antigen collagen type II. T cells specific for the non-modified antigen undergo efficient central tolerance. By contrast, PTM-reactive T cells escape thymic selection, though the PTM variant constitutes the dominant form in the periphery. This finding implies that the PTM protein is absent in the thymus, or present at concentrations insufficient to induce negative selection of developing thymocytes and explains the lower level of tolerance induction against the PTM antigen. As the majority of self-antigens are post-translationally modified, these data raise the possibility that T cells specific for other self-antigens naturally subjected to PTM may escape central tolerance induction by a similar mechanism.


Assuntos
Artrite Experimental/imunologia , Tolerância Central/imunologia , Colágeno Tipo II/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Linfócitos T/imunologia , Animais , Autoantígenos/imunologia , Autoimunidade/imunologia , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Timócitos/imunologia , Timo/imunologia
12.
Cell Signal ; 33: 10-21, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28179144

RESUMO

The p38 MAPK signaling pathway is essential in the cellular response to stress stimuli, in particular in the endothelial cells that are major target of external stress. The importance of the bioactive sphingolipid ceramide generated by acid sphingomyelinase is also firmly established in stress-induced endothelial apoptotic cell death. Despite a suggested link between the p38 MAPK and ceramide pathways, the exact molecular events of this connection remain elusive. In the present study, by using two different activators of p38 MAPK, namely anisomycin and ionizing radiation, we depicted how ceramide generated by acid sphingomyelinase was involved in p38 MAPK-dependent apoptosis of endothelial cells. We first proved that both anisomycin and ionizing radiation conducted to apoptosis through activation of p38 MAPK in human microvascular endothelial cells HMEC-1. We then found that both treatments induced activation of acid sphingomyelinase and the generation of ceramide. This step was required for p38 MAPK activation and apoptosis. We finally showed that irradiation, as well as treatment with exogenous C16-ceramide or bacterial sphingomyelinase, induced in endothelial cells a deep reorganization of the plasma membrane with formation of large lipid platforms at the cell surface, leading to p38 MAPK activation and apoptosis in endothelial cells. Altogether, our results proved that the plasma membrane reorganization leading to ceramide production is essential for stress-induced activation of p38 MAPK and apoptosis in endothelial cells and established the link between the acid sphingomyelinase/ceramide and p38 MAPK pathways.


Assuntos
Apoptose , Membrana Celular/metabolismo , Ceramidas/metabolismo , Sistema de Sinalização das MAP Quinases , Esfingomielina Fosfodiesterase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Endoteliais/metabolismo , Ativação Enzimática , Humanos , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Estresse Fisiológico
13.
Sci Rep ; 7(1): 15453, 2017 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-29133847

RESUMO

Paladin (Pald1, mKIAA1274 or x99384) was identified in screens for vascular-specific genes and is a putative phosphatase. Paladin has also been proposed to be involved in various biological processes such as insulin signaling, innate immunity and neural crest migration. To determine the role of paladin we have now characterized the Pald1 knock-out mouse in a broad array of behavioral, physiological and biochemical tests. Here, we show that female, but not male, Pald1 heterozygous and homozygous knock-out mice display an emphysema-like histology with increased alveolar air spaces and impaired lung function with an obstructive phenotype. In contrast to many other tissues where Pald1 is restricted to the vascular compartment, Pald1 is expressed in both the epithelial and mesenchymal compartments of the postnatal lung. However, in Pald1 knock-out females, there is a specific increase in apoptosis and proliferation of endothelial cells, but not in non-endothelial cells. This results in a transient reduction of endothelial cells in the maturing lung. Our data suggests that Pald1 is required during lung vascular development and for normal function of the developing and adult lung in a sex-specific manner. To our knowledge, this is the first report of a sex-specific effect on endothelial cell apoptosis.


Assuntos
Apoptose/fisiologia , Enfisema/patologia , Células Endoteliais/patologia , Endotélio Vascular/crescimento & desenvolvimento , Fosfoproteínas Fosfatases/metabolismo , Animais , Modelos Animais de Doenças , Embrião de Mamíferos , Enfisema/genética , Feminino , Heterozigoto , Humanos , Pulmão/irrigação sanguínea , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas Fosfatases/genética , Fatores Sexuais
14.
PLoS One ; 10(9): e0137949, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26394398

RESUMO

Despite its known expression in both the vascular endothelium and the lung epithelium, until recently the physiological role of the adhesion receptor Gpr116/ADGRF5 has remained elusive. We generated a new mouse model of constitutive Gpr116 inactivation, with a large genetic deletion encompassing exon 4 to exon 21 of the Gpr116 gene. This model allowed us to confirm recent results defining Gpr116 as necessary regulator of surfactant homeostasis. The loss of Gpr116 provokes an early accumulation of surfactant in the lungs, followed by a massive infiltration of macrophages, and eventually progresses into an emphysema-like pathology. Further analysis of this knockout model revealed cerebral vascular leakage, beginning at around 1.5 months of age. Additionally, endothelial-specific deletion of Gpr116 resulted in a significant increase of the brain vascular leakage. Mice devoid of Gpr116 developed an anatomically normal and largely functional vascular network, surprisingly exhibited an attenuated pathological retinal vascular response in a model of oxygen-induced retinopathy. These data suggest that Gpr116 modulates endothelial properties, a previously unappreciated function despite the pan-vascular expression of this receptor. Our results support the key pulmonary function of Gpr116 and describe a new role in the central nervous system vasculature.


Assuntos
Células Epiteliais Alveolares/metabolismo , Endotélio Vascular/metabolismo , Surfactantes Pulmonares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Permeabilidade Capilar/genética , Feminino , Expressão Gênica , Homeostase/genética , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Modelos Biológicos , Miocárdio/metabolismo , Miocárdio/patologia , Receptores Acoplados a Proteínas G/genética , Neovascularização Retiniana/genética , Neovascularização Retiniana/metabolismo , Baço/metabolismo , Baço/patologia
15.
Nat Commun ; 5: 3195, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24463987

RESUMO

Lack of robust methods for establishment and expansion of pluripotent human embryonic stem (hES) cells still hampers development of cell therapy. Laminins (LN) are a family of highly cell-type specific basement membrane proteins important for cell adhesion, differentiation, migration and phenotype stability. Here we produce and isolate a human recombinant LN-521 isoform and develop a cell culture matrix containing LN-521 and E-cadherin, which both localize to stem cell niches in vivo. This matrix allows clonal derivation, clonal survival and long-term self-renewal of hES cells under completely chemically defined and xeno-free conditions without ROCK inhibitors. Neither LN-521 nor E-cadherin alone enable clonal survival of hES cells. The LN-521/E-cadherin matrix allows hES cell line derivation from blastocyst inner cell mass and single blastomere cells without a need to destroy the embryo. This method can facilitate the generation of hES cell lines for development of different cell types for regenerative medicine purposes.


Assuntos
Caderinas , Técnicas de Cultura de Células , Células-Tronco Embrionárias/fisiologia , Laminina , Humanos , Integrina alfa6beta1/metabolismo , Cariotipagem , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
16.
Dev Cell ; 23(3): 587-99, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22975327

RESUMO

Angiogenesis, the process by which new blood vessels arise from preexisting ones, is critical for embryonic development and is an integral part of many disease processes. Recent studies have provided detailed information on how angiogenic sprouts initiate, elongate, and branch, but less is known about how these processes cease. Here, we show that S1PR1, a receptor for the blood-borne bioactive lipid sphingosine-1-phosphate (S1P), is critical for inhibition of angiogenesis and acquisition of vascular stability. Loss of S1PR1 leads to increased endothelial cell sprouting and the formation of ectopic vessel branches. Conversely, S1PR1 signaling inhibits angiogenic sprouting and enhances cell-to-cell adhesion. This correlates with inhibition of vascular endothelial growth factor-A (VEGF-A)-induced signaling and stabilization of vascular endothelial (VE)-cadherin localization at endothelial junctions. Our data suggest that S1PR1 signaling acts as a vascular-intrinsic stabilization mechanism, protecting developing blood vessels against aberrant angiogenic responses.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Neovascularização Fisiológica , Receptores de Lisoesfingolipídeo/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptores de Lisoesfingolipídeo/deficiência , Receptores de Esfingosina-1-Fosfato , Peixe-Zebra
17.
Cancer Res ; 70(23): 9905-15, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21118968

RESUMO

A previous in vitro study showed that sphingosine-1-phosphate (S1P), a ceramide antagonist, preserved endothelial cells in culture from radiation-induced apoptosis. We proposed to validate the role of S1P in tissue radioprotection by inhibiting acute gastrointestinal (GI) syndrome induced by endothelial cell apoptosis after high dose of radiation. Retro-orbital S1P was injected in mice exposed to 15 Gy, a dose-inducing GI syndrome within 10 days. Overall survival and apoptosis on intestines sections were studied. Intestinal cell type targeted by S1P and early molecular survival pathways were researched using irradiated in vitro cell models and in vivo mouse models. We showed that retro-orbital S1P injection before irradiation prevented GI syndrome by inhibiting endothelium collapse. We defined endothelium as a specific therapeutic target because only these cells and not intestinal epithelial cells, or B and T lymphocytes, were protected. Pharmacologic approaches using AKT inhibitor and pertussis toxin established that S1P affords endothelial cell protection in vitro and in vivo through a mechanism involving AKT and 7-pass transmembrane receptors coupled to Gi proteins. Our results provide strong pharmacologic and mechanistic proofs that S1P protects endothelial cells against acute radiation enteropathy.


Assuntos
Apoptose/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos da radiação , Western Blotting , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Medula Óssea/efeitos da radiação , Linhagem Celular , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/efeitos da radiação , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/patologia , Trato Gastrointestinal/efeitos da radiação , Humanos , Imuno-Histoquímica , Intestino Delgado/citologia , Intestino Delgado/efeitos da radiação , Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/patologia , Tecido Linfoide/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Lesões Experimentais por Radiação/tratamento farmacológico , Esfingosina/farmacologia , Síndrome
18.
J Nucl Med ; 51(4): 624-31, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20351352

RESUMO

UNLABELLED: A significant antitumor effect was previously observed with radioimmunotherapy using anti-carcinoembryonic antigen (131)I-F6 monoclonal antibody in medullary thyroid cancer-bearing nude mice. Nevertheless, no complete response was observed. As seen with chemotherapy, drugs targeting the tumor microenvironment might improve radioimmunotherapy efficacy. This study evaluated the toxicity and efficacy of combining radioimmunotherapy with thalidomide or a cyclopeptidic vascular endothelial growth inhibitor (CBOP11) in mice grafted with the TT human medullary thyroid cancer cell line. METHODS: Six to 10 nude mice treated with 92.5 MBq of (131)I-F6 in association with 200 mg/kg/d of oral thalidomide during 20 d by force-feeding or 0.45 mg/kg/d of CBOP11 during 25 d using subcutaneous minipumps were compared with control mice receiving either treatment or naked F6 or nonspecific (131)I-734. Combined therapies included (131)I-F6 at day 0 followed by thalidomide between days 20 and 40, thalidomide between days 0 and 20 followed by (131)I-F6 at day 25, (131)I-F6 at day 0 and CBOP11 between days 0 and 25, CBOP11 between days 0 and 25 followed by (131)I-F6 at day 25, and (131)I-F6 at day 0 followed by CBOP11 between days 20 and 45. Animal weight, hematologic toxicity, tumor volume, and serum calcitonin were monitored for the following 3 mo. Improvement of (125)I-F6 tumor biodistribution by antiangiogenic drug was studied after pretreatment by thalidomide. Follow-up of the tumor after combined antiangiogenic and radioimmunotherapy therapies was performed by histology studies. RESULTS: Combined associations, as compared with radioimmunotherapy alone, increased leukopenia but not thrombocytopenia. Tumor volume-quadrupling time (TVQT) was 22.8 +/- 3.3 d in the control group, 29.9 +/- 3.6 d in the group treated with thalidomide, 34.6 +/- 4.4 d in the group treated with CBOP11, and 51.0 +/- 2.8 d after radioimmunotherapy alone. As compared with radioimmunotherapy, TVQT was significantly longer (P < 0.01) after thalidomide followed by radioimmunotherapy (69.83 +/- 3.9), CBOP11 followed by radioimmunotherapy (71.3 +/- 6.1), and CBOP11-radioimmunotherapy in concomitance (64.2 +/- 6.1). Nevertheless, TVQT was not increased after radioimmunotherapy followed by thalidomide (48.8 +/- 4) and radioimmunotherapy followed by CBOP11 (56.8 +/- 4.8). Surprisingly, pretreatment by CBOP11 or thalidomide sensitized larger tumors (>300 mm(3)) to radioimmunotherapy. Change in calcitonin levels confirmed morphologic tumor response. Tumor uptake 24 h after injection of (125)I-F6 was 4.5 +/- 0.6 percentage injected dose per gram (%ID/g) without pretreatment and 8.7 +/- 1.3 %ID/g with pretreatment by thalidomide. An increase of the antitumor effect observed using the antiangiogenic drug combined with radioimmunotherapy was correlated with a decrease of blood vessels shown by von Willebrand immunostaining. CONCLUSION: Pretreatment with antiangiogenic therapies improved radioimmunotherapy efficacy, with acceptable toxicity. Future investigations will be performed to understand how antiangiogenic agents sensitize large tumors to radioimmunotherapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antígeno Carcinoembrionário/metabolismo , Regulação Neoplásica da Expressão Gênica , Radioimunoterapia , Neoplasias da Glândula Tireoide/terapia , Ensaios Antitumorais Modelo de Xenoenxerto , Inibidores da Angiogênese/imunologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Apoptose/efeitos dos fármacos , Antígeno Carcinoembrionário/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Fatores de Crescimento Endotelial/farmacocinética , Fatores de Crescimento Endotelial/farmacologia , Fatores de Crescimento Endotelial/uso terapêutico , Humanos , Radioisótopos do Iodo/química , Camundongos , Peptídeos Cíclicos/farmacocinética , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/uso terapêutico , Talidomida/farmacologia , Talidomida/uso terapêutico , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Distribuição Tecidual , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA