Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gastroenterology ; 162(6): 1690-1704, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35031299

RESUMO

BACKGROUND & AIMS: Crohn's disease (CD) globally emerges with Westernization of lifestyle and nutritional habits. However, a specific dietary constituent that comprehensively evokes gut inflammation in human inflammatory bowel diseases remains elusive. We aimed to delineate how increased intake of polyunsaturated fatty acids (PUFAs) in a Western diet, known to impart risk for developing CD, affects gut inflammation and disease course. We hypothesized that the unfolded protein response and antioxidative activity of glutathione peroxidase 4 (GPX4), which are compromised in human CD epithelium, compensates for metabolic perturbation evoked by dietary PUFAs. METHODS: We phenotyped and mechanistically dissected enteritis evoked by a PUFA-enriched Western diet in 2 mouse models exhibiting endoplasmic reticulum (ER) stress consequent to intestinal epithelial cell (IEC)-specific deletion of X-box binding protein 1 (Xbp1) or Gpx4. We translated the findings to human CD epithelial organoids and correlated PUFA intake, as estimated by a dietary questionnaire or stool metabolomics, with clinical disease course in 2 independent CD cohorts. RESULTS: PUFA excess in a Western diet potently induced ER stress, driving enteritis in Xbp1-/-IEC and Gpx4+/-IEC mice. ω-3 and ω-6 PUFAs activated the epithelial endoplasmic reticulum sensor inositol-requiring enzyme 1α (IRE1α) by toll-like receptor 2 (TLR2) sensing of oxidation-specific epitopes. TLR2-controlled IRE1α activity governed PUFA-induced chemokine production and enteritis. In active human CD, ω-3 and ω-6 PUFAs instigated epithelial chemokine expression, and patients displayed a compatible inflammatory stress signature in the serum. Estimated PUFA intake correlated with clinical and biochemical disease activity in a cohort of 160 CD patients, which was similarly demonstrable in an independent metabolomic stool analysis from 199 CD patients. CONCLUSIONS: We provide evidence for the concept of PUFA-induced metabolic gut inflammation which may worsen the course of human CD. Our findings provide a basis for targeted nutritional therapy.


Assuntos
Doença de Crohn , Enterite , Ácidos Graxos Ômega-3 , Animais , Doença de Crohn/tratamento farmacológico , Endorribonucleases , Enterite/induzido quimicamente , Enterite/tratamento farmacológico , Ácidos Graxos Insaturados , Humanos , Inflamação/tratamento farmacológico , Camundongos , Proteínas Serina-Treonina Quinases , Receptor 2 Toll-Like
2.
Cell Metab ; 34(1): 106-124.e10, 2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34986329

RESUMO

Still's disease, the paradigm of autoinflammation-cum-autoimmunity, predisposes for a cytokine storm with excessive T lymphocyte activation upon viral infection. Loss of function of the purine nucleoside enzyme FAMIN is the sole known cause for monogenic Still's disease. Here we discovered that a FAMIN-enabled purine metabolon in dendritic cells (DCs) restrains CD4+ and CD8+ T cell priming. DCs with absent FAMIN activity prime for enhanced antigen-specific cytotoxicity, IFNγ secretion, and T cell expansion, resulting in excessive influenza A virus-specific responses. Enhanced priming is already manifest with hypomorphic FAMIN-I254V, for which ∼6% of mankind is homozygous. FAMIN controls membrane trafficking and restrains antigen presentation in an NADH/NAD+-dependent manner by balancing flux through adenine-guanine nucleotide interconversion cycles. FAMIN additionally converts hypoxanthine into inosine, which DCs release to dampen T cell activation. Compromised FAMIN consequently enhances immunosurveillance of syngeneic tumors. FAMIN is a biochemical checkpoint that protects against excessive antiviral T cell responses, autoimmunity, and autoinflammation.


Assuntos
Autoimunidade , Purinas , Linfócitos T CD8-Positivos , Células Dendríticas , Ativação Linfocitária , Purinas/metabolismo
3.
Gut ; 71(3): 509-520, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33758004

RESUMO

OBJECTIVE: Primary sclerosing cholangitis (PSC) is in 70% of cases associated with inflammatory bowel disease. The hypermorphic T108M variant of the orphan G protein-coupled receptor GPR35 increases risk for PSC and ulcerative colitis (UC), conditions strongly predisposing for inflammation-associated liver and colon cancer. Lack of GPR35 reduces tumour numbers in mouse models of spontaneous and colitis associated cancer. The tumour microenvironment substantially determines tumour growth, and tumour-associated macrophages are crucial for neovascularisation. We aim to understand the role of the GPR35 pathway in the tumour microenvironment of spontaneous and colitis-associated colon cancers. DESIGN: Mice lacking GPR35 on their macrophages underwent models of spontaneous colon cancer or colitis-associated cancer. The role of tumour-associated macrophages was then assessed in biochemical and functional assays. RESULTS: Here, we show that GPR35 on macrophages is a potent amplifier of tumour growth by stimulating neoangiogenesis and tumour tissue remodelling. Deletion of Gpr35 in macrophages profoundly reduces tumour growth in inflammation-associated and spontaneous tumour models caused by mutant tumour suppressor adenomatous polyposis coli. Neoangiogenesis and matrix metalloproteinase activity is promoted by GPR35 via Na/K-ATPase-dependent ion pumping and Src activation, and is selectively inhibited by a GPR35-specific pepducin. Supernatants from human inducible-pluripotent-stem-cell derived macrophages carrying the UC and PSC risk variant stimulate tube formation by enhancing the release of angiogenic factors. CONCLUSIONS: Activation of the GPR35 pathway promotes tumour growth via two separate routes, by directly augmenting proliferation in epithelial cells that express the receptor, and by coordinating macrophages' ability to create a tumour-permissive environment.


Assuntos
Colangite Esclerosante/patologia , Colite Ulcerativa/patologia , Neoplasias do Colo/etiologia , Neovascularização Patológica/etiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Colangite Esclerosante/genética , Colite Ulcerativa/genética , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Macrófagos/fisiologia , Camundongos , Microambiente Tumoral
4.
Cell ; 180(2): 278-295.e23, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-31978345

RESUMO

Mutations in FAMIN cause arthritis and inflammatory bowel disease in early childhood, and a common genetic variant increases the risk for Crohn's disease and leprosy. We developed an unbiased liquid chromatography-mass spectrometry screen for enzymatic activity of this orphan protein. We report that FAMIN phosphorolytically cleaves adenosine into adenine and ribose-1-phosphate. Such activity was considered absent from eukaryotic metabolism. FAMIN and its prokaryotic orthologs additionally have adenosine deaminase, purine nucleoside phosphorylase, and S-methyl-5'-thioadenosine phosphorylase activity, hence, combine activities of the namesake enzymes of central purine metabolism. FAMIN enables in macrophages a purine nucleotide cycle (PNC) between adenosine and inosine monophosphate and adenylosuccinate, which consumes aspartate and releases fumarate in a manner involving fatty acid oxidation and ATP-citrate lyase activity. This macrophage PNC synchronizes mitochondrial activity with glycolysis by balancing electron transfer to mitochondria, thereby supporting glycolytic activity and promoting oxidative phosphorylation and mitochondrial H+ and phosphate recycling.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Adenina/metabolismo , Adenosina/metabolismo , Adenosina Desaminase/metabolismo , Cromatografia Líquida/métodos , Células HEK293 , Células Hep G2 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Espectrometria de Massas/métodos , Enzimas Multifuncionais/genética , Fosforilação , Proteínas/genética , Nucleotídeos de Purina/metabolismo , Purinas/metabolismo
5.
Sci Signal ; 12(562)2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30600262

RESUMO

The sodium potassium pump (Na/K-ATPase) ensures the electrochemical gradient of a cell through an energy-dependent process that consumes about one-third of regenerated ATP. We report that the G protein-coupled receptor GPR35 interacted with the α chain of Na/K-ATPase and promotes its ion transport and Src signaling activity in a ligand-independent manner. Deletion of Gpr35 increased baseline Ca2+ to maximal levels and reduced Src activation and overall metabolic activity in macrophages and intestinal epithelial cells (IECs). In contrast, a common T108M polymorphism in GPR35 was hypermorphic and had the opposite effects to Gpr35 deletion on Src activation and metabolic activity. The T108M polymorphism is associated with ulcerative colitis and primary sclerosing cholangitis, inflammatory diseases with a high cancer risk. GPR35 promoted homeostatic IEC turnover, whereas Gpr35 deletion or inhibition by a selective pepducin prevented inflammation-associated and spontaneous intestinal tumorigenesis in mice. Thus, GPR35 acts as a central signaling and metabolic pacesetter, which reveals an unexpected role of Na/K-ATPase in macrophage and IEC biology.


Assuntos
Proliferação de Células , Glicólise , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Carcinogênese , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Células Epiteliais/metabolismo , Células HEK293 , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos Knockout , Polimorfismo de Nucleotídeo Único , Receptores Acoplados a Proteínas G/genética , ATPase Trocadora de Sódio-Potássio/genética , Células THP-1 , Quinases da Família src/genética , Quinases da Família src/metabolismo
6.
Gut ; 66(5): 930-938, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-26858343

RESUMO

OBJECTIVE: Alcoholic steatohepatitis is a life-threatening condition with short-term mortality up to 40%. It features hepatic neutrophil infiltration and blood neutrophilia, and may evolve from ethanol-induced breakdown of the enteric barrier and consequent bacteraemia. Signalling through CXCR1/2 G-protein-coupled-receptors (GPCRs), the interleukin (IL)-8 receptors, is critical for the recruitment and activation of neutrophils. We have developed short lipopeptides (pepducins), which inhibit post-ligand GPCR activation precisely targeting individual GPCRs. DESIGN: Experimental alcoholic liver disease was induced by administering alcohol and a Lieber-DeCarli high-fat diet. CXCR1/2 GPCRs were blocked via pepducins either from onset of the experiment or after disease was fully established. Hepatic inflammatory infiltration, hepatocyte lipid accumulation and overall survival were assessed as primary outcome parameters. Neutrophil activation was assessed by myeloperoxidase activity and liver cell damage by aspartate aminotransferase and alanine aminotransferase plasma levels. Chemotaxis assays were performed to identify chemoattractant signals derived from alcohol-exposed hepatocytes. RESULTS: Here, we show that experimental alcoholic liver disease is driven by CXCR1/2-dependent activation of neutrophils. CXCR1/2-specific pepducins not only protected mice from liver inflammation, weight loss and mortality associated with experimental alcoholic liver disease, but therapeutic administration cured disease and prevented further mortality in fully established disease. Hepatic neutrophil infiltration and triglyceride accumulation was abrogated by CXCR1/2 blockade. Moreover, CXCL-1 plasma levels were decreased with the pepducin therapy as was the transcription of hepatic IL-1ß mRNA. CONCLUSIONS: We propose that high circulating IL-8 in human alcoholic hepatitis may cause pathogenic overzealous neutrophil activation, and therapeutic blockade via pepducins merits clinical study.


Assuntos
Fígado Gorduroso Alcoólico/tratamento farmacológico , Lipopeptídeos/farmacologia , Receptores de Interleucina-8A/antagonistas & inibidores , Receptores de Interleucina-8B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Caspase 1/metabolismo , Quimiocina CXCL1/genética , Quimiotaxia/efeitos dos fármacos , Fígado Gorduroso Alcoólico/complicações , Fígado Gorduroso Alcoólico/metabolismo , Fígado Gorduroso Alcoólico/patologia , Feminino , Células Hep G2 , Hepatite/etiologia , Hepatite/prevenção & controle , Hepatócitos/metabolismo , Humanos , Interleucina-1beta/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipopeptídeos/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Peroxidase/metabolismo , Receptores de Interleucina-8A/sangue , Taxa de Sobrevida , Transcrição Gênica/efeitos dos fármacos , Triglicerídeos/metabolismo , Fator de Necrose Tumoral alfa/genética , Redução de Peso/efeitos dos fármacos
7.
Nat Immunol ; 17(9): 1046-56, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27478939

RESUMO

Single-nucleotide variations in C13orf31 (LACC1) that encode p.C284R and p.I254V in a protein of unknown function (called 'FAMIN' here) are associated with increased risk for systemic juvenile idiopathic arthritis, leprosy and Crohn's disease. Here we set out to identify the biological mechanism affected by these coding variations. FAMIN formed a complex with fatty acid synthase (FASN) on peroxisomes and promoted flux through de novo lipogenesis to concomitantly drive high levels of fatty-acid oxidation (FAO) and glycolysis and, consequently, ATP regeneration. FAMIN-dependent FAO controlled inflammasome activation, mitochondrial and NADPH-oxidase-dependent production of reactive oxygen species (ROS), and the bactericidal activity of macrophages. As p.I254V and p.C284R resulted in diminished function and loss of function, respectively, FAMIN determined resilience to endotoxin shock. Thus, we have identified a central regulator of the metabolic function and bioenergetic state of macrophages that is under evolutionary selection and determines the risk of inflammatory and infectious disease.


Assuntos
Artrite Juvenil/genética , Doença de Crohn/genética , Infecções/genética , Hanseníase/genética , Macrófagos/imunologia , Proteínas/genética , Choque Séptico/genética , Trifosfato de Adenosina/metabolismo , Animais , Bacteriólise , Células Cultivadas , Metabolismo Energético , Ácido Graxo Sintase Tipo I/metabolismo , Predisposição Genética para Doença , Humanos , Inflamassomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidases/metabolismo , Oxirredução , Polimorfismo de Nucleotídeo Único , Risco
8.
J Exp Med ; 210(10): 2041-56, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-24043762

RESUMO

Unresolved endoplasmic reticulum (ER) stress in the epithelium can provoke intestinal inflammation. Hypomorphic variants of ER stress response mediators, such as X-box-binding protein 1 (XBP1), confer genetic risk for inflammatory bowel disease. We report here that hypomorphic Xbp1 function instructs a multilayered regenerative response in the intestinal epithelium. This is characterized by intestinal stem cell (ISC) expansion as shown by an inositol-requiring enzyme 1α (Ire1α)-mediated increase in Lgr5(+) and Olfm4(+) ISCs and a Stat3-dependent increase in the proliferative output of transit-amplifying cells. These consequences of hypomorphic Xbp1 function are associated with an increased propensity to develop colitis-associated and spontaneous adenomatous polyposis coli (APC)-related tumors of the intestinal epithelium, which in the latter case is shown to be dependent on Ire1α. This study reveals an unexpected role for Xbp1 in suppressing tumor formation through restraint of a pathway that involves an Ire1α- and Stat3-mediated regenerative response of the epithelium as a consequence of ER stress. As such, Xbp1 in the intestinal epithelium not only regulates local inflammation but at the same time also determines the propensity of the epithelium to develop tumors.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/genética , Estresse do Retículo Endoplasmático , Mucosa Intestinal/metabolismo , Intestinos/patologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Fatores de Transcrição/genética , Animais , Comunicação Autócrina/genética , Transformação Celular Neoplásica/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proteínas de Ligação a DNA/metabolismo , Endorribonucleases/metabolismo , Ativação Enzimática , Deleção de Genes , Genes APC , Interleucina-11/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/patologia , Janus Quinase 1/metabolismo , MAP Quinase Quinase 4/antagonistas & inibidores , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição de Fator Regulador X , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição/metabolismo , Carga Tumoral/genética , Proteína 1 de Ligação a X-Box
9.
Blood ; 119(7): 1717-25, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-22186993

RESUMO

The chemokine receptor CXCR4, which normally regulates stromal stem cell interactions in the bone marrow, is highly expressed on a variety of malignant hematologic cells, including lymphoma and lymphocytic leukemias. A new treatment concept has arisen wherein CXCR4 may be an effective therapeutic target as an adjunct to treatment of hematologic neoplasms with chemo- and immunotherapy. In the present study, we developed pepducins, cell-penetrating lipopeptide antagonists of CXCR4, to interdict CXCL12-CXCR4 transmembrane signaling to intracellular G-proteins. We demonstrate that pepducins targeting the first (i1) or third (i3) intracellular loops of CXCR4 completely abrogate CXCL12-mediated cell migration of lymphocytic leukemias and lymphomas. Stromal-cell coculture protects lymphoma cells from apoptosis in response to treatment with the CD20-targeted Ab rituximab. However, combination treatment with CXCR4 pepducins and rituximab significantly increases the apoptotic effect of rituximab. Furthermore, treatment of mice bearing disseminated lymphoma xenografts with pepducins alone or in combination with rituximab significantly increased their survival. These data demonstrate that CXCL12-CXCR4 signaling can be effectively inhibited by cell-penetrating pepducins, which represents a potential new treatment strategy for lymphoid malignancies.


Assuntos
Leucemia Linfoide/tratamento farmacológico , Lipopeptídeos/uso terapêutico , Linfoma/tratamento farmacológico , Receptores CXCR4/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Leucemia Linfoide/metabolismo , Leucemia Linfoide/patologia , Lipopeptídeos/administração & dosagem , Lipopeptídeos/síntese química , Lipopeptídeos/química , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Modelos Moleculares , Terapia de Alvo Molecular , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cell ; 137(2): 332-43, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19379698

RESUMO

Matrix metalloproteases (MMPs) play important roles in normal and pathological remodeling processes including atherothrombotic disease, inflammation, angiogenesis, and cancer. MMPs have been viewed as matrix-degrading enzymes, but recent studies have shown that they possess direct signaling capabilities. Platelets harbor several MMPs that modulate hemostatic function and platelet survival; however their mode of action remains unknown. We show that platelet MMP-1 activates protease-activated receptor-1 (PAR1) on the surface of platelets. Exposure of platelets to fibrillar collagen converts the surface-bound proMMP-1 zymogen to active MMP-1, which promotes aggregation through PAR1. Unexpectedly, MMP-1 cleaves PAR1 at a distinct site that strongly activates Rho-GTP pathways, cell shape change and motility, and MAPK signaling. Blockade of MMP1-PAR1 curtails thrombogenesis under arterial flow conditions and inhibits thrombosis in animals. These studies provide a link between matrix-dependent activation of metalloproteases and platelet-G protein signaling and identify MMP1-PAR1 as a potential target for the prevention of arterial thrombosis.


Assuntos
Receptor PAR-1/metabolismo , Trombose/metabolismo , Animais , Plaquetas/metabolismo , Colágeno/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Cobaias , Humanos , Ligantes , Metaloproteinase 1 da Matriz/metabolismo , Estrutura Terciária de Proteína , Receptor PAR-1/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Mol Cancer Ther ; 7(9): 2746-57, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18790755

RESUMO

Gene chip and proteomic analyses of tumors and stromal tissue has led to the identification of dozens of candidate tumor and host components potentially involved in tumor-stromal interactions, angiogenesis, and progression of invasive disease. In particular, matrix metalloproteases (MMP) have emerged as important biomarkers and prognostic factors for invasive and metastatic cancers. From an initial screen of benign versus malignant patient fluids, we delineated a metalloprotease cascade comprising MMP-14, MMP-9, and MMP-1 that culminates in activation of PAR1, a G protein-coupled protease-activated receptor up-regulated in diverse cancers. In xenograft models of advanced peritoneal ovarian cancer, PAR1-dependent angiogenesis, ascites formation, and metastasis were effectively inhibited by i.p. administration of cell-penetrating pepducins based on the intracellular loops of PAR1. These data provide an in vivo proof-of-concept that targeting the metalloprotease-PAR1 signaling system may be a novel therapeutic approach in the treatment of ovarian cancer.


Assuntos
Ascite/enzimologia , Metaloproteases/antagonistas & inibidores , Neovascularização Patológica/enzimologia , Oligopeptídeos/farmacologia , Neoplasias Ovarianas/patologia , Receptor PAR-1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Líquidos Corporais/enzimologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Progressão da Doença , Docetaxel , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Humanos , Camundongos , Camundongos Nus , Células NIH 3T3 , Invasividade Neoplásica , Oligopeptídeos/administração & dosagem , Oligopeptídeos/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Peritônio/enzimologia , Peritônio/patologia , Taxoides/administração & dosagem , Taxoides/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Thromb Haemost ; 93(6): 1120-7, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15968398

RESUMO

Platelet recruitment crucially depends on amplification systems provided by autocrine and paracrine factors such as adenosine diphosphate. In inflammatory states, consumption of coagulation proteins, such as antithrombin aggravates the procoagulant state. In this study, we report that platelets express syndecan-4, an antithrombin-binding cell surface heparan sulphate proteoglycan, whose ligation with antithrombin inhibits activated platelet-dependent superoxide anion release from neutrophils by the limitation of adenosine diphosphate and adenosine triphosphate secretion in activated platelets. Adenosine triphosphate-induced platelet aggregation is reduced after treatment of platelets with antithrombin, which is reversed by blockade of syndecan-4. We further observed that antithrombin limits CD40 ligand expression in adenosine diphosphate-activated platelets and inhibits the shedding of syndecan-4 from activated platelets. Syndecan-4 appears to be directly involved in regulating platelet aggregation as anti-syndecan-4 antibody augments platelet aggregation. We suggest that antithrombin might exert beneficial effects in disseminated intravascular coagulation by reducing platelet activation, observed as inhibited CD40 ligand expression, syndecan-4 shedding, and adenosine diphosphate- and adenosine triphosphate-release from activated platelets with subsequent inhibition of neutrophil respiratory burst. From these data it is concluded that syndecan-4 may play important roles in the regulation of inflammatory effects of platelets.


Assuntos
Plaquetas/metabolismo , Glicoproteínas de Membrana/sangue , Proteoglicanas/sangue , Difosfato de Adenosina/sangue , Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/sangue , Antitrombinas/farmacologia , Plaquetas/efeitos dos fármacos , Ligante de CD40/sangue , Expressão Gênica , Humanos , Técnicas In Vitro , Mediadores da Inflamação/sangue , Glicoproteínas de Membrana/genética , Neutrófilos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Proteoglicanas/genética , RNA Mensageiro/sangue , RNA Mensageiro/genética , Explosão Respiratória , Transdução de Sinais , Sindecana-4
13.
Nat Med ; 11(6): 661-5, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15880119

RESUMO

We describe a new therapeutic approach for the treatment of lethal sepsis using cell-penetrating lipopeptides-termed pepducins-that target either individual or multiple chemokine receptors. Interleukin-8 (IL-8), a ligand for the CXCR1 and CXCR2 receptors, is the most potent endogenous proinflammatory chemokine in sepsis. IL-8 levels rise in blood and lung fluids to activate neutrophils and other cells, and correlate with shock, lung injury and high mortality. We show that pepducins derived from either the i1 or i3 intracellular loops of CXCR1 and CXCR2 prevent the IL-8 response of both receptors and reverse the lethal sequelae of sepsis, including disseminated intravascular coagulation and multi-organ failure in mice. Conversely, pepducins selective for CXCR4 cause a massive leukocytosis that does not affect survival. CXCR1 and CXCR2 pepducins conferred nearly 100% survival even when treatment was postponed, suggesting that our approach might be beneficial in the setting of advanced disease.


Assuntos
Peptídeos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Receptores de Interleucina-8A/antagonistas & inibidores , Receptores de Interleucina-8B/antagonistas & inibidores , Síndrome de Resposta Inflamatória Sistêmica/tratamento farmacológico , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Feminino , Humanos , Interleucina-8/fisiologia , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Neutrófilos/efeitos dos fármacos , Conformação Proteica , Subunidades Proteicas , Receptores CXCR4/fisiologia , Receptores de Interleucina-8A/química , Receptores de Interleucina-8A/fisiologia , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/fisiologia
14.
Blood Coagul Fibrinolysis ; 16(2): 105-10, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15741797

RESUMO

Increasing evidence shows the importance of platelet-endothelial cell interactions in the progression of atherosclerosis. Platelets contribute to coronary events both as major components of thrombi and as a triggering factor in inflammation that leads to plaque vulnerability. Recent data suggest that statins, besides their lipid-lowering properties, exert pleiotropic effects that may be beneficial in atherosclerosis. Whether activated platelets influence cyclooxygenase-2 (COX-2) expression in human umbilical vein endothelial cells (HUVEC), the effect of atorvastatin, and possible mechanisms were investigated. COX-2 gene expression in HUVEC was studied using real-time polymerase chain reaction. CD40 ligand surface expression of platelets was tested by fluorescence-activated cell sorting analyses. Activated platelets significantly up-regulated COX-2 gene expression in HUVEC. Co-incubation of platelets with atorvastatin was shown to reverse this up-regulation via reduction of CD40 ligand surface expression on platelets. Data suggest that atorvastatin influences CD40-CD40-ligand-dependent platelet-endothelial interaction and that this influence affects platelet-induced COX-2 expression in HUVEC.


Assuntos
Anticolesterolemiantes/farmacologia , Plaquetas/fisiologia , Ligante de CD40/biossíntese , Células Endoteliais/fisiologia , Ácidos Heptanoicos/farmacologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Pirróis/farmacologia , Arteriosclerose/tratamento farmacológico , Arteriosclerose/fisiopatologia , Atorvastatina , Antígenos CD40/metabolismo , Células Cultivadas , Ciclo-Oxigenase 2 , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Membrana , Ativação Plaquetária/efeitos dos fármacos
15.
J Clin Endocrinol Metab ; 90(5): 2964-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15728209

RESUMO

Chronic inflammation is characterized by tissue infiltration with monocytes/macrophages, which possess broad proinflammatory, destructive, and remodeling capacities. Elevated levels of osteoprotegerin, an important regulator of differentiation and activation of osteoclasts that also affects different cells of the immune system, were found in the serum of patients with chronic inflammatory diseases. The study of whether osteoprotegerin affects monocyte locomotion in vitro and the possible mechanisms and pathways involved was investigated using Boyden microchemotaxis chambers and Western blot analyses. Osteoprotegerin significantly stimulated monocyte chemotaxis, whereas preincubation of monocytes with osteoprotegerin inhibited monocyte migration toward optimal concentrations of regulated upon activation normal T cell expressed and secreted, monocyte chemotactic protein -1, and procalcitonin. The effects of osteoprotegerin were abolished by pretreating cells with heparinase I and chondroitinase or antibodies against the ectodomain of syndecan-1. Osteoprotegerin signaling was shown to involve protein kinase C, phosphatidylinositol 3-kinase/Akt, and tyrosine kinase. Data suggest that osteoprotegerin affects monocyte mi-gration and protein kinase C and phosphatidylinositol 3-kinase/Akt activation via syndecan-1. Osteoprotegerin-induced deactivation of monocyte chemotaxis toward different chemokines is due to interaction of osteoprotegerin with heparan sulfate and chondroitin sulfate.


Assuntos
Quimiotaxia de Leucócito/efeitos dos fármacos , Glicoproteínas/farmacologia , Glicoproteínas de Membrana/fisiologia , Monócitos/imunologia , Proteoglicanas/fisiologia , Movimento Celular/efeitos dos fármacos , Condroitinases e Condroitina Liases/farmacologia , Heparina Liase/farmacologia , Humanos , Osteoprotegerina , Fosfatidilinositol 3-Quinases/fisiologia , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores Citoplasmáticos e Nucleares , Receptores do Fator de Necrose Tumoral , Sindecana-1 , Sindecanas , Fator A de Crescimento do Endotélio Vascular/fisiologia , Quinases da Família src/metabolismo
16.
Thromb Res ; 114(3): 185-94, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15342215

RESUMO

BACKGROUND: Thrombin downregulates endothelial ectonucleotidase activity resulting in high levels of adenosine diphosphate (ADP) and adenosine triphosphate (ATP) which lead to platelet, leukocyte and endothelial activation. Depending on adenosine nucleotide levels, resting platelets inhibit and thrombin-activated platelets increase respiratory burst of neutrophils. Whether the red wine polyphenols quercetin and resveratrol affect thrombin-dependent adenosine nucleotide, metabolism and thrombin-induced signaling is unknown. MATERIALS AND METHODS: ATP and ADP secretion by platelets, the impact on subsequent oxidative burst activity in neutrophils and CD39/ATPdase function in endothelial cells (ECs)was studied. Cell migration was measured in modified Boyden chambers; adenosine metabolites were quantified by high-performance liquid chromatography (HPLC). Signal transduction was studied by Western blotting. RESULTS: Quercetin and resveratrol inhibited thrombin-induced ADP and ATP secretion from platelets in a concentration-dependent manner. Augmented respiratory burst of neutrophils in response to thrombin-activated platelets was also inhibited by the two polyphenols as was neutrophil migration toward thrombin-induced supernatants of platelets. Quercetin and resveratrol restored the decreased CD39/ATPdase activity in human umbilical vein endothelial cells, in response to thrombin as demonstrated by adenosine monophosphate (AMP) and adenosine increases in endothelial culture supernatants. Both polyphenols inhibited thrombin-induced MAPK, JNK and focal adhesion kinase activities in endothelial cells. CONCLUSION: Quercetin and resveratrol interfere with the proinflammatory signaling of thrombin resulting in the inhibition of adenosine nucleotide secretion from activated platelets and decreased neutrophil function. Moreover, the polyphenols protect endothelial adenosine nucleotide metabolism when downregulated by thrombin. These observations may explain cardioprotective effects of grape products.


Assuntos
Nucleotídeos de Adenina/metabolismo , Plaquetas/fisiologia , Células Endoteliais/fisiologia , Neutrófilos/fisiologia , Quercetina/farmacologia , Estilbenos/farmacologia , Trombina/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Antioxidantes/farmacologia , Plaquetas/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/fisiologia , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Humanos , Neutrófilos/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Arthritis Rheum ; 50(7): 2309-16, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15248232

RESUMO

OBJECTIVE: RANKL, a member of the tumor necrosis factor superfamily, is a central regulator of osteoclast recruitment and activation. Whether RANKL affects monocyte locomotion in vitro via RANK and a possible signaling pathway were investigated. METHODS: Monocytes were obtained from venous blood of healthy donors. Cell migration was studied by micropore filter assays. The signaling mechanisms required for RANKL-dependent migration were tested using signaling enzyme blockers and Western blot analyses. Expression of RANK messenger RNA (mRNA) in monocytes was demonstrated by reverse transcriptase-polymerase chain reaction, and receptor expression on cell surface was investigated by fluorescence-activated cell sorting analyses. RESULTS: RANKL significantly stimulated monocyte chemotaxis via activation of phosphatidylinositol 3-kinase, phosphodiesterase, and Src kinase. The effect on migration was inhibited by osteoprotegerin, which is the decoy receptor for RANKL. Expression of RANK receptor mRNA was shown, and synthesis of RANK in monocytes was suggested by the detection of RANK immunoreactivity on the cell surface. CONCLUSION: These data suggest that RANK is expressed by monocytes whose activation by RANKL stimulates directed migration involving phosphatidylinositol 3-kinase, phosphodiesterase, and Src kinases.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Glicoproteínas/fisiologia , Monócitos/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Proteínas de Transporte/farmacologia , Movimento Celular/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , Ativação Enzimática , Enzimas/fisiologia , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Humanos , Membranas Intracelulares/enzimologia , Glicoproteínas de Membrana/farmacologia , Monócitos/metabolismo , Osteoprotegerina , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Quinases da Família src/metabolismo
18.
FASEB J ; 18(11): 1309-11, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15208267

RESUMO

Accumulation of inflammatory mononuclear phagocytes in Alzheimer's senile plaques, a hallmark of the innate immune response to beta-amyloid fibrils, can initiate and propagate neurodegeneration characteristic of Alzheimer's disease. Phagocytes migrate toward amyloid beta-protein involving formyl peptide receptor like-1-dependent signaling. Using human peripheral blood monocytes in Boyden chamber micropore filter assays, we show that the amyloid beta-protein- and amyloid beta-precursor protein-induced migration was abrogated by dimethylsphingosine, a sphingosine kinase inhibitor. Amyloid beta-protein stimulated in monocytes the gene expression for sphingosine-1-phosphate receptors 2 and 5, but not 1, 3, and 4. FTY720 that acts as a sphingosine-1-phosphate receptor agonist after endogenous phosphorylation by sphingosine kinase, as well as various neuropeptides that are known to be monocyte chemoattractants, dose-dependently inhibited amyloid beta-protein-induced migration. These data demonstrate that the migratory effects of beta-amyloid in human monocytes involve spingosine-1-phosphate signaling. Whereas endogenous neuropeptides may arrest and activate monocytes at sites of high beta-amyloid concentrations, interference with the amyloid beta-protein-dependent sphingosine-1-phosphate pathway in monocytes by FTY720, a novel immunomodulatory drug, suggests that FTY720 may be efficacious in beta-amyloid-related inflammatory diseases.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/farmacologia , Fatores Imunológicos/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/biossíntese , Esfingosina/análogos & derivados , 1-Metil-3-Isobutilxantina/farmacologia , Androstadienos/farmacologia , Bombesina/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Movimento Celular/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , Toxina da Cólera/farmacologia , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Cloridrato de Fingolimode , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Humanos , Indóis/farmacologia , Leucócitos Mononucleares/citologia , Maleimidas/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neuropeptídeos/farmacologia , Toxina Pertussis/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Mensageiro/biossíntese , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/fisiologia , Secretogranina II , Esfingosina/farmacologia , Estaurosporina/farmacologia , Tirfostinas/farmacologia , Peptídeo Intestinal Vasoativo/farmacologia , Wortmanina
19.
Am J Respir Cell Mol Biol ; 30(5): 729-35, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14607815

RESUMO

Vascular endothelial growth factor (VEGF) is highly expressed in the airway of patients with asthma. Whether VEGF affects eosinophil function in vitro and if VEGF receptors are involved was tested. Eosinophils were from venous blood of healthy donors. Cell migration was studied by micropore filter assays. Signaling mechanisms required for VEGF-dependent migration were tested using signaling enzyme blockers. Expression of flt-1 and KDR/flk-1 mRNA in eosinophils was demonstrated in reverse transcriptase-polymerase chain reaction, and receptor expression was investigated by fluorescence-activated cell sorting analysis. Eosinophil cationic protein release was measured in eosinophil supernatants by enzyme-linked immunosorbent assay. VEGF significantly stimulated eosinophil chemotaxis via activation of protein kinase C and phosphatidylinositol 3'-kinase. The effect on migration was reversed by an antibody against VEGF receptor flt-1, but not by an antibody against KDR/flk-1. Expression of VEGF receptor flt-1 mRNA was shown and synthesis of VEGF receptor in eosinophils is suggested by detection of VEGF receptor immunoreactivity on the cell surface. Data suggest that VEGF receptor flt-1 is expressed by eosinophils whose activation with VEGF stimulates directed migration and release of eosinophil cationic protein. Thus, VEGF may play an important role in the modulation of eosinophilic inflammation.


Assuntos
Movimento Celular/fisiologia , Eosinófilos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Anticorpos/metabolismo , Proteínas Sanguíneas/metabolismo , Células Cultivadas , Quimiocina CCL5/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Inibidores Enzimáticos/metabolismo , Proteínas Granulares de Eosinófilos , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribonucleases/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
20.
Regul Pept ; 115(2): 123-9, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12972327

RESUMO

G-protein-coupled bombesin receptors are capable of signaling through the G(i) protein even when receptor-coupling to G(q) is blocked by [D-Arg1,D-Phe5,D-Trp7,9,Leu11]substance P (SpD), a neurokinin-1 receptor antagonist and "biased" agonist to bombesin receptors. As bombesin is a monocyte and tumor cell attractant, we were interested in the effects of SpD on cell migration. Chemotaxis of monocytes was tested in micropore filter assays. SpD was a dose-dependent agonist in monocyte migration and was not inhibited by antagonists to neurokinin-1 or -2 receptors. SpD failed to inhibit chemotaxis toward bombesin, suggesting that inhibition of bombesin receptor coupling to G(q) with SpD does not impair migratory responses elicited by bombesin. As pertussis toxin inhibited migration, coupling of receptors to G(i) may signal migration. Chemotaxis toward SpD was inhibited by bombesin receptor antagonists as well as by blocking signaling enzymes downstream of G(q) (phospholipase-3 and protein kinase C with wortmannin and bisindolylmaleimide, respectively), suggesting transactivation of G(q)-mediated chemotaxis signaling by SpD via bombesin receptors. Protein kinase C that induces sphingosine kinase activation and production of sphingosine-1-phosphate, which may lead to G(q)-dependent chemoattraction, was involved in SpD-dependent migration. Inhibition of sphingosine-1-phosphate production with dimethylsphingosine inhibited monocyte migration toward SpD. Data suggest that SpD induces migration in monocytes and signaling events involving activation of sphingosine kinase in a G(i) protein- and protein kinase C-dependent fashion. "Biased" agonism of SpD at bombesin receptors may affect normal and tumor cell migration.


Assuntos
Bombesina/farmacologia , Movimento Celular/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Leucócitos Mononucleares/citologia , Antagonistas dos Receptores de Neurocinina-1 , Receptores da Bombesina/agonistas , Substância P/análogos & derivados , Substância P/farmacologia , Androstadienos/farmacologia , Bombesina/química , Bombesina/metabolismo , Células Cultivadas , Quimiotaxia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/química , Humanos , Indóis/farmacologia , Leucócitos Mononucleares/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Lisofosfolipídeos/metabolismo , Maleimidas/farmacologia , Toxina Pertussis/farmacologia , Fosfolipases/antagonistas & inibidores , Fosfolipases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Receptores da Bombesina/química , Receptores da Bombesina/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA