Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Signal ; 17(824): eadg9256, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38377179

RESUMO

High-density lipoprotein (HDL) nanoparticles promote endothelial cell (EC) function and suppress inflammation, but their utility in treating EC dysfunction has not been fully explored. Here, we describe a fusion protein named ApoA1-ApoM (A1M) consisting of apolipoprotein A1 (ApoA1), the principal structural protein of HDL that forms lipid nanoparticles, and ApoM, a chaperone for the bioactive lipid sphingosine 1-phosphate (S1P). A1M forms HDL-like particles, binds to S1P, and is signaling competent. Molecular dynamics simulations showed that the S1P-bound ApoM moiety in A1M efficiently activated EC surface receptors. Treatment of human umbilical vein ECs with A1M-S1P stimulated barrier function either alone or cooperatively with other barrier-enhancing molecules, including the stable prostacyclin analog iloprost, and suppressed cytokine-induced inflammation. A1M-S1P injection into mice during sterile inflammation suppressed neutrophil influx and inflammatory mediator secretion. Moreover, systemic A1M administration led to a sustained increase in circulating HDL-bound S1P and suppressed inflammation in a murine model of LPS-induced endotoxemia. We propose that A1M administration may enhance vascular endothelial barrier function, suppress cytokine storm, and promote resilience of the vascular endothelium.


Assuntos
Apolipoproteínas , Lipocalinas , Humanos , Camundongos , Animais , Apolipoproteínas/metabolismo , Apolipoproteínas/farmacologia , Lipocalinas/metabolismo , Lipocalinas/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Apolipoproteínas M , Inflamação , Lipoproteínas HDL/farmacologia , Lipoproteínas HDL/metabolismo , Lisofosfolipídeos/farmacologia , Lisofosfolipídeos/metabolismo , Esfingosina
2.
Am J Respir Cell Mol Biol ; 70(2): 119-128, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37934676

RESUMO

Respiratory viral infections are frequent causes of acute respiratory distress syndrome (ARDS), a disabling condition with a mortality of up to 46%. The pulmonary endothelium plays an important role in the development of ARDS as well as the pathogenesis of pulmonary fibrosis; however, the therapeutic potential to modulate endothelium-dependent signaling to prevent deleterious consequences has not been well explored. Here, we used a clinically relevant influenza A virus infection model, endothelial cell-specific transgenic gain-of-function and loss-of-function mice as well as pharmacologic approaches and in vitro modeling, to define the mechanism by which S1PR1 expression is dampened during influenza virus infection and determine whether therapeutic augmentation of S1PR1 has the potential to reduce long-term postviral fibrotic complications. We found that the influenza virus-induced inflammatory milieu promoted internalization of S1PR1, which was pharmacologically inhibited with paroxetine, an inhibitor of GRK2. Moreover, genetic overexpression or administration of paroxetine days after influenza virus infection was sufficient to reduce postviral pulmonary fibrosis. Taken together, our data suggest that endothelial S1PR1 signaling provides critical protection against long-term fibrotic complications after pulmonary viral infection. These findings support the development of antifibrotic strategies that augment S1PR1 expression in virus-induced ARDS to improve long-term patient outcomes.


Assuntos
Infecções por Orthomyxoviridae , Fibrose Pulmonar , Síndrome do Desconforto Respiratório , Animais , Humanos , Camundongos , Endotélio/metabolismo , Paroxetina , Receptores de Esfingosina-1-Fosfato/metabolismo
3.
Am J Respir Cell Mol Biol ; 66(1): 38-52, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34343038

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive disease which leads to significant morbidity and mortality from respiratory failure. The two drugs currently approved for clinical use slow the rate of decline in lung function but have not been shown to halt disease progression or reverse established fibrosis. Thus, new therapeutic targets are needed. Endothelial injury and the resultant vascular permeability are critical components in the response to tissue injury and are present in patients with IPF. However, it remains unclear how vascular permeability affects lung repair and fibrosis following injury. Lipid mediators such as sphingosine-1-phosphate (S1P) are known to regulate multiple homeostatic processes in the lung including vascular permeability. We demonstrate that endothelial cell-(EC) specific deletion of the S1P receptor 1 (S1PR1) in mice (EC-S1pr1-/-) results in increased lung vascular permeability at baseline. Following a low-dose intratracheal bleomycin challenge, EC-S1pr1-/- mice had increased and persistent vascular permeability compared with wild-type mice, which was strongly correlated with the amount and localization of resulting pulmonary fibrosis. EC-S1pr1-/- mice also had increased immune cell infiltration and activation of the coagulation cascade within the lung. However, increased circulating S1P ligand in ApoM-overexpressing mice was insufficient to protect against bleomycin-induced pulmonary fibrosis. Overall, these data demonstrate that endothelial cell S1PR1 controls vascular permeability in the lung, is associated with changes in immune cell infiltration and extravascular coagulation, and modulates the fibrotic response to lung injury.


Assuntos
Permeabilidade Capilar , Células Endoteliais/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Bleomicina , Coagulação Sanguínea , Deleção de Genes , Fibrose Pulmonar Idiopática/sangue , Pulmão/irrigação sanguínea , Pulmão/patologia , Lisofosfolipídeos/sangue , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , RNA-Seq , Análise de Célula Única , Esfingosina/análogos & derivados , Esfingosina/sangue
4.
Proc Natl Acad Sci U S A ; 117(6): 3157-3166, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31988136

RESUMO

Sphingosine 1-phosphate receptor-1 (S1PR1) is essential for embryonic vascular development and maturation. In the adult, it is a key regulator of vascular barrier function and inflammatory processes. Its roles in tumor angiogenesis, tumor growth, and metastasis are not well understood. In this paper, we show that S1PR1 is expressed and active in tumor vessels. Murine tumor vessels that lack S1PR1 in the vascular endothelium (S1pr1 ECKO) show excessive vascular sprouting and branching, decreased barrier function, and poor perfusion accompanied by loose attachment of pericytes. Compound knockout of S1pr1, 2, and 3 genes further exacerbated these phenotypes, suggesting compensatory function of endothelial S1PR2 and 3 in the absence of S1PR1. On the other hand, tumor vessels with high expression of S1PR1 (S1pr1 ECTG) show less branching, tortuosity, and enhanced pericyte coverage. Larger tumors and enhanced lung metastasis were seen in S1pr1 ECKO, whereas S1pr1 ECTG showed smaller tumors and reduced metastasis. Furthermore, antitumor activity of a chemotherapeutic agent (doxorubicin) and immune checkpoint inhibitor blocker (anti-PD-1 antibody) were more effective in S1pr1 ECTG than in the wild-type counterparts. These data suggest that tumor endothelial S1PR1 induces vascular normalization and influences tumor growth and metastasis, thus enhancing antitumor therapies in mouse models. Strategies to enhance S1PR1 signaling in tumor vessels may be an important adjunct to standard cancer therapy of solid tumors.


Assuntos
Antineoplásicos/farmacologia , Neovascularização Patológica/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Permeabilidade Capilar/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Camundongos , Camundongos Knockout , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Receptores de Esfingosina-1-Fosfato/genética
5.
Science ; 366(6463)2019 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-31624181

RESUMO

Sphingosine 1-phosphate (S1P), a metabolic product of cell membrane sphingolipids, is bound to extracellular chaperones, is enriched in circulatory fluids, and binds to G protein-coupled S1P receptors (S1PRs) to regulate embryonic development, postnatal organ function, and disease. S1PRs regulate essential processes such as adaptive immune cell trafficking, vascular development, and homeostasis. Moreover, S1PR signaling is a driver of multiple diseases. The past decade has witnessed an exponential growth in this field, in part because of multidisciplinary research focused on this lipid mediator and the application of S1PR-targeted drugs in clinical medicine. This has revealed fundamental principles of lysophospholipid mediator signaling that not only clarify the complex and wide ranging actions of S1P but also guide the development of therapeutics and translational directions in immunological, cardiovascular, neurological, inflammatory, and fibrotic diseases.


Assuntos
Lisofosfolipídeos/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Animais , Apolipoproteínas M/metabolismo , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/fisiopatologia , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares , Sistema Cardiovascular/embriologia , Sistema Cardiovascular/crescimento & desenvolvimento , Sistema Cardiovascular/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/fisiologia , Desenvolvimento de Medicamentos , Fibrose/tratamento farmacológico , Fibrose/fisiopatologia , Homeostase , Humanos , Fenômenos do Sistema Imunitário , Camundongos , Chaperonas Moleculares , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/fisiopatologia , Transdução de Sinais , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inibidores
6.
J Exp Med ; 216(7): 1582-1598, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31147448

RESUMO

Sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) activate G protein-coupled receptors (GPCRs) to regulate biological processes. Using a genome-wide CRISPR/dCas9-based GPCR signaling screen, LPAR1 was identified as an inducer of S1PR1/ß-arrestin coupling while suppressing Gαi signaling. S1pr1 and Lpar1-positive lymphatic endothelial cells (LECs) of lymph nodes exhibit constitutive S1PR1/ß-arrestin signaling, which was suppressed by LPAR1 antagonism. Pharmacological inhibition or genetic loss of function of Lpar1 reduced the frequency of punctate junctions at sinus-lining LECs. Ligand activation of transfected LPAR1 in endothelial cells remodeled junctions from continuous to punctate structures and increased transendothelial permeability. In addition, LPAR1 antagonism in mice increased lymph node retention of adoptively transferred lymphocytes. These data suggest that cross-talk between LPAR1 and S1PR1 promotes the porous junctional architecture of sinus-lining LECs, which enables efficient lymphocyte trafficking. Heterotypic inter-GPCR coupling may regulate complex cellular phenotypes in physiological milieu containing many GPCR ligands.


Assuntos
Células Endoteliais/metabolismo , Linfonodos/metabolismo , Receptor Cross-Talk , Animais , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Imunofluorescência , Edição de Genes , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Lisofosfolipídeos/metabolismo , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo
7.
Sci Immunol ; 4(33)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30877143

RESUMO

Sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs) regulate migration of lymphocytes out of thymus to blood and lymph nodes (LNs) to efferent lymph, whereas their role in other tissue sites is not known. Here, we investigated the question of how these molecules regulate leukocyte migration from tissues through afferent lymphatics to draining LNs (dLNs). S1P, but not other chemokines, selectively enhanced human and murine CD4 T cell migration across lymphatic endothelial cells (LECs). T cell S1PR1 and S1PR4, and LEC S1PR2, were required for migration across LECs and into lymphatic vessels and dLNs. S1PR1 and S1PR4 differentially regulated T cell motility and vascular cell adhesion molecule-1 (VCAM-1) binding. S1PR2 regulated LEC layer structure, permeability, and expression of the junction molecules VE-cadherin, occludin, and zonulin-1 through the ERK pathway. S1PR2 facilitated T cell transcellular migration through VCAM-1 expression and recruitment of T cells to LEC migration sites. These results demonstrated distinct roles for S1PRs in comodulating T cell and LEC functions in migration and suggest previously unknown levels of regulation of leukocytes and endothelial cells during homeostasis and immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Movimento Celular/imunologia , Células Endoteliais/imunologia , Vasos Linfáticos/imunologia , Receptores de Esfingosina-1-Fosfato/imunologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Linhagem Celular , Células Endoteliais/fisiologia , Humanos , Lisofosfolipídeos/imunologia , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esfingosina/análogos & derivados , Esfingosina/imunologia , Receptores de Esfingosina-1-Fosfato/genética , Proteínas de Junções Íntimas/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia
8.
Sci Signal ; 10(492)2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28811382

RESUMO

Endothelial dysfunction, a hallmark of vascular disease, is restored by plasma high-density lipoprotein (HDL). However, a generalized increase in HDL abundance is not beneficial, suggesting that specific HDL species mediate protective effects. Apolipoprotein M-containing HDL (ApoM+HDL), which carries the bioactive lipid sphingosine 1-phosphate (S1P), promotes endothelial function by activating G protein-coupled S1P receptors. Moreover, HDL-bound S1P is limiting in several inflammatory, metabolic, and vascular diseases. We report the development of a soluble carrier for S1P, ApoM-Fc, which activated S1P receptors in a sustained manner and promoted endothelial function. In contrast, ApoM-Fc did not modulate circulating lymphocyte numbers, suggesting that it specifically activated endothelial S1P receptors. ApoM-Fc administration reduced blood pressure in hypertensive mice, attenuated myocardial damage after ischemia/reperfusion injury, and reduced brain infarct volume in the middle cerebral artery occlusion model of stroke. Our proof-of-concept study suggests that selective and sustained targeting of endothelial S1P receptors by ApoM-Fc could be a viable therapeutic strategy in vascular diseases.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Hipertensão/prevenção & controle , Lisofosfolipídeos/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Esfingosina/análogos & derivados , Animais , Apolipoproteínas M/metabolismo , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipertensão/metabolismo , Hipertensão/patologia , Lipoproteínas HDL/metabolismo , Masculino , Camundongos , Camundongos Knockout , Ligação Proteica , Receptores Fc/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/farmacologia
9.
J Cell Biol ; 204(3): 377-93, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24493589

RESUMO

Export of newly synthesized G protein-coupled receptors (GPCRs) remains poorly characterized. We show in this paper that lipocalin-type prostaglandin D2 (PGD2) synthase (L-PGDS) interacts intracellularly with the GPCR DP1 in an agonist-independent manner. L-PGDS promotes cell surface expression of DP1, but not of other GPCRs, in HEK293 and HeLa cells, independent of L-PGDS enzyme activity. In addition, formation of a DP1-Hsp90 complex necessary for DP1 export to the cell surface is dependent on the interaction between L-PGDS and the C-terminal MEEVD residues of Hsp90. Surprisingly, PGD2 synthesis by L-PGDS is promoted by coexpression of DP1, suggesting a possible intracrine/autocrine signaling mechanism. In this regard, L-PGDS increases the formation of a DP1-ERK1/2 complex and increases DP1-mediated ERK1/2 signaling. Our findings define a novel cooperative mechanism in which a GPCR (DP1) promotes the activity of the enzyme (L-PGDS) that produces its agonist (PGD2) and in which this enzyme in turn acts as a cofactor (of Hsp90) to promote export and agonist-dependent activity of the receptor.


Assuntos
Espaço Intracelular/enzimologia , Oxirredutases Intramoleculares/metabolismo , Lipocalinas/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/enzimologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases , Proteínas Mutantes/metabolismo , Transporte Proteico , Rede trans-Golgi/metabolismo
10.
J Biol Chem ; 286(47): 40802-13, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21990357

RESUMO

Previous reports by us and others demonstrated that G protein-coupled receptors interact functionally with Rab GTPases. Here, we show that the ß(2)-adrenergic receptor (ß(2)AR) interacts with the Rab geranylgeranyltransferase α-subunit (RGGTA). Confocal microscopy showed that ß(2)AR co-localizes with RGGTA in intracellular compartments and at the plasma membrane. Site-directed mutagenesis revealed that RGGTA binds to the L(339)L(340) motif in the ß(2)AR C terminus known to be involved in the transport of the receptor from the endoplasmic reticulum to the cell surface. Modulation of the cellular levels of RGGTA protein by overexpression or siRNA-mediated knockdown of the endogenous protein demonstrated that RGGTA has a positive role in the maturation and anterograde trafficking of the ß(2)AR, which requires the interaction of RGGTA with the ß(2)AR L(339)L(340) motif. Furthermore, the ß(2)AR modulates the geranylgeranylation of Rab6a, Rab8a, and Rab11a, but not of other Rab proteins tested in this study. Regulation of Rab geranylgeranylation by the ß(2)AR was dependent on the RGGTA-interacting L(339)L(340) motif. Interestingly, a RGGTA-Y107F mutant was unable to regulate Rab geranylgeranylation but still promoted ß(2)AR maturation, suggesting that RGGTA may have functions independent of Rab geranylgeranylation. We demonstrate for the first time an interaction between a transmembrane receptor and RGGTA which regulates the maturation and anterograde transport of the receptor, as well as geranylgeranylation of Rab GTPases.


Assuntos
Alquil e Aril Transferases/metabolismo , Prenilação de Proteína , Receptores Adrenérgicos beta 2/metabolismo , Células HEK293 , Células HeLa , Humanos , Espaço Intracelular/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , Especificidade por Substrato , Proteínas rab de Ligação ao GTP/metabolismo
11.
J Cell Sci ; 124(Pt 19): 3292-304, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21940795

RESUMO

We identified the WD-repeat-containing protein, WDR36, as an interacting partner of the ß isoform of thromboxane A(2) receptor (TPß) by yeast two-hybrid screening. We demonstrated that WDR36 directly interacts with the C-terminus and the first intracellular loop of TPß by in vitro GST-pulldown assays. The interaction in a cellular context was observed by co-immunoprecipitation, which was positively affected by TPß stimulation. TPß-WDR36 colocalization was detected by confocal microscopy at the plasma membrane in non-stimulated HEK293 cells but the complex translocated to intracellular vesicles following receptor stimulation. Coexpression of WDR36 and its siRNA-mediated knockdown, respectively, increased and inhibited TPß-induced Gαq signalling. Interestingly, WDR36 co-immunoprecipitated with Gαq, and promoted TPß-Gαq interaction. WDR36 also associated with phospholipase Cß (PLCß) and increased the interaction between Gαq and PLCß, but prevented sequestration of activated Gαq by GRK2. In addition, the presence of TPß in PLCß immunoprecipitates was augmented by expression of WDR36. Finally, disease-associated variants of WDR36 affected its ability to modulate Gαq-mediated signalling by TPß. We report that WDR36 acts as a new scaffold protein tethering a G-protein-coupled receptor, Gαq and PLCß in a signalling complex.


Assuntos
Proteínas do Olho/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fosfolipase C beta/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Ativação Enzimática , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Células HEK293 , Humanos , Imunoprecipitação , Isoenzimas/metabolismo , Microscopia de Fluorescência , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Complexos Multiproteicos/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo , Transporte Proteico , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...