Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Int J Mol Sci ; 22(13)2021 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-34281225

RESUMO

Thyroid hormones (THs) are key regulators of different biological processes. Their action involves genomic and non-genomic mechanisms, which together mediate the final effects of TH in target tissues. However, the proportion of the two processes and their contribution to the TH-mediated effects are still poorly understood. Skeletal muscle is a classical target tissue for TH, which regulates muscle strength and contraction, as well as energetic metabolism of myofibers. Here we address the different contribution of genomic and non-genomic action of TH in skeletal muscle cells by specifically silencing the deiodinase Dio2 or the ß3-Integrin expression via CRISPR/Cas9 technology. We found that myoblast proliferation is inversely regulated by integrin signal and the D2-dependent TH activation. Similarly, inhibition of the nuclear receptor action reduced myoblast proliferation, confirming that genomic action of TH attenuates proliferative rates. Contrarily, genomic and non-genomic signals promote muscle differentiation and the regulation of the redox state. Taken together, our data reveal that integration of genomic and non-genomic signal pathways finely regulates skeletal muscle physiology. These findings not only contribute to the understanding of the mechanisms involved in TH modulation of muscle physiology but also add insight into the interplay between different mechanisms of action of TH in muscle cells.


Assuntos
Células Musculares/fisiologia , Músculo Esquelético/fisiologia , Hormônios Tireóideos/fisiologia , Animais , Diferenciação Celular , Integrina beta3/fisiologia , Iodeto Peroxidase/fisiologia , Camundongos , Músculo Esquelético/citologia , Iodotironina Desiodinase Tipo II
2.
Sci Signal ; 14(665)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436498

RESUMO

The spike protein of SARS-CoV-2 binds the angiotensin-converting enzyme 2 (ACE2) on the host cell surface and subsequently enters host cells through receptor-mediated endocytosis. Additional cell receptors may be directly or indirectly involved, including integrins. The cytoplasmic tails of ACE2 and integrins contain several predicted short linear motifs (SLiMs) that may facilitate internalization of the virus as well as its subsequent propagation through processes such as autophagy. Here, we measured the binding affinity of predicted interactions between SLiMs in the cytoplasmic tails of ACE2 and integrin ß3 with proteins that mediate endocytic trafficking and autophagy. We validated that a class I PDZ-binding motif mediated binding of ACE2 to the scaffolding proteins SNX27, NHERF3, and SHANK, and that a binding site for the clathrin adaptor AP2 µ2 in ACE2 overlaps with a phospho-dependent binding site for the SH2 domains of Src family tyrosine kinases. Furthermore, we validated that an LC3-interacting region (LIR) in integrin ß3 bound to the ATG8 domains of the autophagy receptors MAP1LC3 and GABARAP in a manner enhanced by LIR-adjacent phosphorylation. Our results provide molecular links between cell receptors and mediators of endocytosis and autophagy that may facilitate viral entry and propagation.


Assuntos
Enzima de Conversão de Angiotensina 2/fisiologia , COVID-19/virologia , Integrina beta3/fisiologia , Receptores Virais/fisiologia , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , Internalização do Vírus , Sequência de Aminoácidos , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/genética , Autofagia/fisiologia , Endocitose/fisiologia , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Integrina beta3/química , Integrina beta3/genética , Modelos Moleculares , Pandemias , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Receptores Virais/química , Receptores Virais/genética , SARS-CoV-2/genética
3.
J Cancer Res Clin Oncol ; 147(2): 361-371, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33083904

RESUMO

Integrin ß3 (ITGB3) is probably related to skeletal metastasis, which is the most frequent complication in breast cancer progression. We aimed to define its role and suitability as target for anti-metastatic therapy. We generated two MDA-MB-231 cell clones with conditional miRNA-mediated ITGB3 knockdown for analyzing the resulting effects in vitro regarding mRNA expression, proliferation and migration, as well the impact on skeletal metastasis in a nude rat model. Furthermore, ITGB3 levels were analyzed in exosomes from plasma of rats with skeletal metastases, and from MDA-MB-231 cells incubated with these vesicles, as well as from exosomes secreted by cells with conditional ITGB3 knockdown. This inhibition of ITGB3 expression decreased cellular proliferation and more distinctly inhibited cellular migration. Reduction and even complete remissions of respective soft tissue and osteolytic lesions were detected after ITGB3 knockdown in vivo. Furthermore, ITGB3 levels were increased in exosomes isolated from plasma of rats harboring MDA-MB-231 lesions as well as in respective cells incubated with these vesicles in vitro. ITGB3 was distinctly decreased in exosomes from cells with ITGB3 knockdown. The observed in vitro and in vivo anti-ITGB3 effects can be explained by downregulation of specific genes, which have roles in angiogenesis (NPTN, RRM2), tumor growth (NPTN), energy metabolism (ISCA1), cytokinesis (SEPT11), migration (RRM2, STX6), cell proliferation, invasiveness, senescence, tumorigenesis (RRM2) and vesicle trafficking (SEPT11, STX6). ITGB3 has a role in breast cancer skeletal metastasis via gene expression modulation, as mirrored for ITGB3 in exosomes, thus it could serve as target for anti-metastatic therapy.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Integrina beta3/fisiologia , Osteólise/etiologia , Animais , Neoplasias Ósseas/prevenção & controle , Linhagem Celular Tumoral , Exossomos , Feminino , Humanos , Masculino , Ratos , Lesões dos Tecidos Moles/etiologia
4.
Biomedica ; 40(2): 362-381, 2020 06 15.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-32673463

RESUMO

INTRODUCTION: Cancer is the second leading cause of death in the United States, surpassed only by cardiovascular disease. However, cancer has now overtaken cardiovascular disease as the main cause of death in 12 countries in Western Europe. The burden of cancer is posing a major challenge to health care systems worldwide and demanding improvements in methods for cancer prevention, diagnosis, and treatment. Alternative and complementary strategies for orthodox surgery, radiotherapy, and chemotherapy need to be developed. OBJECTIVE: To determine the oncolytic potential of tumor cell-adapted rotavirus in terms of their ability to infect and lysate murine myeloma Sp2/0-Ag14 cells. MATERIALS AND METHODS: We inoculated rotaviruses Wt1-5, WWM, TRUYO, ECwt-O, and WTEW in Sp2/0-Ag14 cells and we examined their infectious effects by immunocytochemistry, immunofluorescence, flow cytometry, and DNA fragmentation assays. RESULTS: Rotavirus infection involved the participation of some heat shock proteins, of protein disulfide isomerase (PDI), and integrin ß3. We detected the accumulation of viral antigens within the virus-inoculated cells and in the culture medium in all the rotavirus isolates examined. The rotavirus-induced cell death mechanism in Sp2/0-Ag14 cells involved changes in cell membrane permeability, chromatin condensation, and DNA fragmentation, which were compatible with cytotoxicity and apoptosis. CONCLUSIONS: The ability of the rotavirus isolates Wt1-5, WWM, TRUYO, ECwt-O, and WTEW to infect and cause cell death of Sp2/0-Ag14 cells through mechanisms that are compatible with virus-induced apoptosis makes them potential candidates as oncolytic agents.


Introducción. El cáncer es la segunda causa de muerte en los Estados Unidos, solamente superado por la enfermedad cardiovascular. Sin embargo, el cáncer aventaja a la enfermedad cardiovascular como primera causa de muerte en doce países de Europa occidental. Se requieren mejores métodos de prevención, diagnóstico y tratamiento para afrontar el gran desafío que el cáncer representa mundialmente para los sistemas de salud, y se necesita desarrollar estrategias alternativas y complementarias a la cirugía, la radioterapia y la quimioterapia convencionales. Objetivo. Evaluar el potencial oncolítico de rotavirus adaptados a células tumorales por su capacidad para infectar y lisar células Sp2/0-Ag14 de mieloma de ratón. Materiales and métodos. Los aislamientos de rotavirus Wt1-5, WWM, TRUYO, ECwt-O y WTEW se inocularon en células Sp2/0-Ag14 y se examinaron sus efectos infecciosos mediante inmunocitoquímica, inmunofluorescencia, citometría de flujo y ensayos de fragmentación del ADN. Resultados. La infección con los rotavirus Wt1-5, WWM, TRUYO, ECwt-O y WTEW implicó la participación de algunas proteínas de choque térmico, la proteína disulfuro isomerasa y la integrina ß3. La acumulación de antígenos virales intracelulares y extracelulares se detectó en todos los virus utilizados. Los mecanismos de muerte inducidos por los rotavirus en células Sp2/0-Ag14 indujeron cambios en la permeabilidad de la membrana celular, la condensación de cromatina y la fragmentación de ADN, los cuales fueron compatibles con citotoxicidad y apoptosis. Conclusiones. La capacidad de los rotavirus estudiados para infectar y causar la muerte de células Sp2/0-Ag14 mediante mecanismos compatibles con la apoptosis inducida viralmente los convierte en candidatos potenciales para ser utilizados como agentes oncolíticos.


Assuntos
Efeito Citopatogênico Viral , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Rotavirus , Sequência de Aminoácidos , Animais , Antígenos Virais/análise , Apoptose , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Cromatina/ultraestrutura , Meios de Cultura/química , Fragmentação do DNA , Proteínas de Choque Térmico/fisiologia , Integrina beta3/fisiologia , Camundongos , Mieloma Múltiplo/patologia , Isomerases de Dissulfetos de Proteínas/fisiologia , Rotavirus/imunologia , Rotavirus/fisiologia , Replicação Viral
5.
J Am Soc Nephrol ; 31(8): 1762-1780, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32709711

RESUMO

BACKGROUND: Diabetic nephropathy (dNP), now the leading cause of ESKD, lacks efficient therapies. Coagulation protease-dependent signaling modulates dNP, in part via the G protein-coupled, protease-activated receptors (PARs). Specifically, the cytoprotective protease-activated protein C (aPC) protects from dNP, but the mechanisms are not clear. METHODS: A combination of in vitro approaches and mouse models evaluated the role of aPC-integrin interaction and related signaling in dNP. RESULTS: The zymogen protein C and aPC bind to podocyte integrin-ß3, a subunit of integrin-αvß3. Deficiency of this integrin impairs thrombin-mediated generation of aPC on podocytes. The interaction of aPC with integrin-αvß3 induces transient binding of integrin-ß3 with G α13 and controls PAR-dependent RhoA signaling in podocytes. Binding of aPC to integrin-ß3via its RGD sequence is required for the temporal restriction of RhoA signaling in podocytes. In podocytes lacking integrin-ß3, aPC induces sustained RhoA activation, mimicking the effect of thrombin. In vivo, overexpression of wild-type aPC suppresses pathologic renal RhoA activation and protects against dNP. Disrupting the aPC-integrin-ß3 interaction by specifically deleting podocyte integrin-ß3 or by abolishing aPC's integrin-binding RGD sequence enhances RhoA signaling in mice with high aPC levels and abolishes aPC's nephroprotective effect. Pharmacologic inhibition of PAR1, the pivotal thrombin receptor, restricts RhoA activation and nephroprotects RGE-aPChigh and wild-type mice.Conclusions aPC-integrin-αvß3 acts as a rheostat, controlling PAR1-dependent RhoA activation in podocytes in diabetic nephropathy. These results identify integrin-αvß3 as an essential coreceptor for aPC that is required for nephroprotective aPC-PAR signaling in dNP.


Assuntos
Nefropatias Diabéticas/prevenção & controle , Integrina beta3/fisiologia , Podócitos/fisiologia , Proteína C/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Citoproteção , Receptor de Proteína C Endotelial/fisiologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptor PAR-1/fisiologia
6.
Leukemia ; 34(8): 2087-2101, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32439895

RESUMO

Therapy resistance in leukemia may be due to cancer cell-intrinsic and/or -extrinsic mechanisms. Mutations within BCR-ABL1, the oncogene giving rise to chronic myeloid leukemia (CML), lead to resistance to tyrosine kinase inhibitors (TKI), and some are associated with clinically more aggressive disease and worse outcome. Using the retroviral transduction/transplantation model of CML and human cell lines we faithfully recapitulate accelerated disease course in TKI resistance. We show in various models, that murine and human imatinib-resistant leukemia cells positive for the oncogene BCR-ABL1T315I differ from BCR-ABL1 native (BCR-ABL1) cells with regards to niche location and specific niche interactions. We implicate a pathway via integrin ß3, integrin-linked kinase (ILK) and its role in deposition of the extracellular matrix (ECM) protein fibronectin as causative of these differences. We demonstrate a trend towards a reduced BCR-ABL1T315I+ tumor burden and significantly prolonged survival of mice with BCR-ABL1T315I+ CML treated with fibronectin or an ILK inhibitor in xenogeneic and syngeneic murine transplantation models, respectively. These data suggest that interactions with ECM proteins via the integrin ß3/ILK-mediated signaling pathway in BCR-ABL1T315I+ cells differentially and specifically influence leukemia progression. Niche targeting via modulation of the ECM may be a feasible therapeutic approach to consider in this setting.


Assuntos
Mesilato de Imatinib/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Animais , Resistencia a Medicamentos Antineoplásicos , Fibronectinas/análise , Fibronectinas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Proteínas de Fusão bcr-abl/análise , Proteínas de Fusão bcr-abl/fisiologia , Humanos , Imidazóis/farmacologia , Integrina beta3/fisiologia , Camundongos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/fisiologia , Piridazinas/farmacologia
7.
Shock ; 53(3): 335-343, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31135705

RESUMO

Sepsis is a major challenge in clinical practice and responsible for high mortality. Recent studies indicated that integrins participated in toll-like-receptor (TLR)-mediated innate immunity. In the present study, we investigated the mechanism of integrin ß3 and TLR4 signaling using a cecal ligation and puncture (CLP)-induced sepsis and lipopolysaccharide (LPS)-treated macrophage cell model. In a lethal CLP model, the survival rate of integrin ß3 mice was higher than that of wild-type mice. The levels of alanine aminotransferase, aspartate transaminase, creatinine, blood urea nitrogen , and lactate dehydrogenase in the serum and cluster of differentiation 14 (CD14) protein expression in the tissues were significantly decreased in integrin ß3 mice. A similar effect with regard to CD14 down-regulation was observed in septic TLR4 mice. In wild-type macrophages, the inhibition of integrin ß3 by P11 or with a specific antibody, inhibited TNF-α, and IL-6 release at the early time period of LPS stimulation. However, during the late periods of LPS stimulation this effect was not noted. CD14 expression levels had no change in such treatment. In contract, LPS-induced TNF-α and IL-6 release and LPS-induced CD14 expression were significantly decreased in integrin ß3macrophages. The inhibition of the TLR4 pathway by TAK-242, or in TLR4 mutant macrophages abolished LPS-induced CD14 expression. Integrin ß3 pathway activation by vitronectin exhibited no effect in CD14 expression. Furthermore, recombinant CD14 protein stimulation reversed integrin ß3 deficiency and caused lower TNF-α and IL-6 release. Moreover, the molecular interaction of TLR4 and integrin ß3 was significantly increased following LPS stimulation. In conclusion, integrin ß3 positively regulated TLR4-mediated inflammatory responses via CD14 expression in macrophages in septic condition. Specifically targeting integrin ß3/TLR4-CD14 signaling pathway may be a potential treatment strategy for polymicrobial sepsis.


Assuntos
Integrina beta3/fisiologia , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Sepse/metabolismo , Receptor 4 Toll-Like/fisiologia , Animais , Modelos Animais de Doenças , Mediadores da Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Sepse/etiologia , Sepse/patologia , Transdução de Sinais
8.
Pharmacol Ther ; 203: 107393, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31356909

RESUMO

Antiplatelet drugs serve as a first-line antithrombotic therapy for the management of acute ischemic events and the prevention of secondary complications in vascular diseases. Numerous antiplatelet therapies have been developed; however, currently available agents are still associated with inadequate efficacy, risk of bleeding, and variability in individual response. Understanding the mechanisms of platelet involvement in thrombosis and the clinical development process of antiplatelet agents is critical for the discovery of novel agents. The functions of platelets in thrombosis are regulated by two major mechanisms: the interaction between surface receptors and their ligands, and the downstream intracellular signaling pathways. Recently, most of the progress made in antiplatelet drug development has been achieved with P2Y receptor antagonists. Additionally, the usage of GP IIb/IIIa receptor antagonists has decreased, because it is associated with a higher risk of bleeding and thrombocytopenia. Agents targeting other platelet surface receptors such as PARs, TP receptor, EP3 receptor, GPIb-IX-V receptor, P-selectin, as well as intracellular signaling factors, such as PI3Kß, have been evaluated in an attempt to develop the next generation of antiplatelet drugs, reduce or eliminate interpatient variability of drug efficacy and significantly lower the risk of drug-induced bleeding. The aim of this review is to describe the pathways of platelet activation in thrombosis, and summarize the development process of antiplatelet agents, as well as the preclinical and clinical evaluations performed on these agents.


Assuntos
Plaquetas/efeitos dos fármacos , Inibidores da Agregação Plaquetária/uso terapêutico , Trombose/prevenção & controle , Animais , Plaquetas/fisiologia , Desenvolvimento de Medicamentos , Humanos , Integrina beta3/fisiologia , Terapia de Alvo Molecular , Ativação Plaquetária/efeitos dos fármacos , Glicoproteína IIb da Membrana de Plaquetas/fisiologia , Trombose/fisiopatologia
9.
Clin Exp Metastasis ; 36(4): 351-363, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31119444

RESUMO

The infiltration of tumor-associated macrophages (TAMs) is associated with tumor progression and poor prognosis in endometrial cancer (EC). Collagen triple helix repeat containing 1 (CTHRC1), a secreted ECM protein, has been reported to have important roles in promoting cancer invasion and metastasis, but the functional role of CTHRC1 and its association with TAMs in EC remain unclear. Here we report that, in EC patients, CTHRC1 expression was up-regulated in endometrial cancer tissues compared with normal endometrium (P < 0.0001), and is positively correlated with tumor grade and depth of myometrial invasion (P = 0.024 and P = 0.0002, respectively). Meanwhile, CTHRC1 expression was positively correlated with an increased number of infiltrating TAMs, especially M2-like TAMs (P = 0.003, P = 0.001). In the tumor microenvironment of EC, CTHRC1 not only promoted myometrial invasion by interacting with Integrin ß3-Akt signaling pathway, but also promoted infiltration of M2-like TAMs by upregulating Fractalkine chemokine receptor (CX3CR1) expression in macrophages. Changing levels of recombinant CTHRC1 protein (rCTHRC1) promoted tumor migration and invasion via enhancing macrophage recruitment in vitro. In summary, our findings eventually provided a novel role for CTHRC1 in remodeling the tumor immune microenvironment to promote tumor metastasis in EC patients.


Assuntos
Neoplasias do Endométrio/patologia , Proteínas da Matriz Extracelular/fisiologia , Integrina beta3/fisiologia , Macrófagos/fisiologia , Miométrio/patologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Linhagem Celular Tumoral , Movimento Celular , Neoplasias do Endométrio/imunologia , Feminino , Humanos , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/fisiologia , Receptores de Superfície Celular/análise , Transdução de Sinais/fisiologia , Microambiente Tumoral
10.
Hum Mol Genet ; 28(7): 1153-1161, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30535103

RESUMO

Elevated serotonin (5-HT) blood levels, the first biomarker identified in autism research, has been consistently found in 20-30% of patients with Autism Spectrum Disorder (ASD). Hyperserotonemia is mainly due to greater 5-HT uptake into platelets, mediated by the 5-HT transporter (SERT) located at the platelet plasma membrane. The protein complex involved in platelet SERT trafficking and externalization includes integrin ß3, the beta subunit of the platelet membrane adhesive GP IIb/IIIa. Integrin ß3 is encoded by the ITGB3 gene, previously identified as a quantitative trait locus (QTL) for 5-HT blood levels in ASD at single nucleotide polymorphism (SNP) rs2317385. The present study aims to identify the functional ITGB3 gene variants contributing to hyperserotonemia. ITGB3 gene sequencing in 20 individuals selected on the basis of rs2317385 genotypes defined four haplotypes encompassing six SNPs located in the ITGB3 gene promoter region, all in linkage disequilibrium with rs2317385. Luciferase assays in two hematopoietic cell lines, K-562 and HEL 92.1.7, demonstrate that ITGB3 gene promoter activity is enhanced by the presence of the C allele at rs55827077 specifically during differentiation into megakaryocytes (P < 0.01), with modulatory effects by flanking SNPs. This same allele is strongly associated with (a) higher 5-HT blood levels in 176 autistic individuals (P < 0.001), (b) greater platelet integrin ß3 protein expression (P < 0.05) and (c) enhanced SERT trafficking from the cytosol toward the platelet plasma membrane (P = 4.05 × 10-11). Our results support rs55827077 as the functional ITGB3 gene promoter variant contributing to elevated 5-HT blood levels in ASD and define a mechanistic chain of events linking ITGB3 to hyperserotonemia.


Assuntos
Transtorno do Espectro Autista/genética , Integrina beta3/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/fisiologia , Adolescente , Adulto , Transtorno Autístico/genética , Criança , Pré-Escolar , Distúrbios do Sono por Sonolência Excessiva/genética , Feminino , Predisposição Genética para Doença/genética , Variação Genética/genética , Genótipo , Haplótipos , Humanos , Integrina beta3/fisiologia , Desequilíbrio de Ligação/genética , Masculino , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Transporte Proteico/fisiologia , Serotonina/análise , Serotonina/sangue , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
11.
Blood ; 132(5): 533-543, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-29853537

RESUMO

It is currently unclear why agonist-stimulated platelets require shear force to efficiently externalize the procoagulant phospholipid phosphatidylserine (PS) and release PS-exposed microvesicles (MVs). We reveal that integrin outside-in signaling is an important mechanism for this requirement. PS exposure and MV release were inhibited in ß3-/- platelets or by integrin antagonists. The impaired MV release and PS exposure in ß3-/- platelets were rescued by expression of wild-type ß3 but not a Gα13 binding-deficient ß3 mutant (E733EE to AAA), which blocks outside-in signaling but not ligand binding. Inhibition of Gα13 or Src also diminished agonist/shear-dependent PS exposure and MV release, further indicating a role for integrin outside-in signaling. PS exposure in activated platelets was induced by application of pulling force via an integrin ligand, which was abolished by inhibiting Gα13-integrin interaction, suggesting that Gα13-dependent transmission of mechanical signals by integrins induces PS exposure. Inhibition of Gα13 delayed coagulation in vitro. Furthermore, inhibition or platelet-specific knockout of Gα13 diminished laser-induced intravascular fibrin formation in arterioles in vivo. Thus, ß3 integrins serve as a shear sensor activating the Gα13-dependent outside-in signaling pathway to facilitate platelet procoagulant function. Pharmacological targeting of Gα13-integrin interaction prevents occlusive thrombosis in vivo by inhibiting both coagulation and platelet thrombus formation.


Assuntos
Coagulação Sanguínea , Plaquetas/fisiologia , Micropartículas Derivadas de Células/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Integrina beta3/fisiologia , Fosfatidilserinas/metabolismo , Resistência ao Cisalhamento , Animais , Fenômenos Biomecânicos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Trombose/fisiopatologia
12.
Nat Commun ; 9(1): 2073, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29802249

RESUMO

Smooth muscle cells (SMCs) play a key role in atherogenesis. However, mechanisms regulating expansion and fate of pre-existing SMCs in atherosclerotic plaques remain poorly defined. Here we show that multiple SMC progenitors mix to form the aorta during development. In contrast, during atherogenesis, a single SMC gives rise to the smooth muscle-derived cells that initially coat the cap of atherosclerotic plaques. Subsequently, highly proliferative cap cells invade the plaque core, comprising the majority of plaque cells. Reduction of integrin ß3 (Itgb3) levels in SMCs induces toll-like receptor 4 expression and thereby enhances Cd36 levels and cholesterol-induced transdifferentiation to a macrophage-like phenotype. Global Itgb3 deletion or transplantation of Itgb3(-/-) bone marrow results in recruitment of multiple pre-existing SMCs into plaques. Conditioned medium from Itgb3-silenced macrophages enhances SMC proliferation and migration. Together, our results suggest SMC contribution to atherogenesis is regulated by integrin ß3-mediated pathways in both SMCs and bone marrow-derived cells.


Assuntos
Aterosclerose/patologia , Proliferação de Células , Integrina beta3/fisiologia , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/patologia , Animais , Aorta/citologia , Aorta/patologia , Aterosclerose/cirurgia , Transplante de Medula Óssea , Movimento Celular , Transdiferenciação Celular , Células Cultivadas , Colesterol/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/patologia , Placa Aterosclerótica/cirurgia
13.
Mol Biol Cell ; 28(21): 2887-2903, 2017 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-28835374

RESUMO

Pathogenic hantaviruses bind to the plexin-semaphorin-integrin (PSI) domain of inactive, ß3 integrins. Previous studies have implicated a cognate cis interaction between the bent conformation ß5/ß3 integrins and an arginine-glycine-aspartic acid (RGD) sequence in the first extracellular loop of P2Y2R. With single-molecule atomic force microscopy, we show a specific interaction between an atomic force microscopy tip decorated with recombinant αIIbß3 integrins and (RGD)P2Y2R expressed on cell membranes. Mutation of the RGD sequence to RGE in the P2Y2R removes this interaction. Binding of inactivated and fluorescently labeled Sin Nombre virus (SNV) to the integrin PSI domain stimulates higher affinity for (RGD)P2Y2R on cells, as measured by an increase in the unbinding force. In CHO cells, stably expressing αIIbß3 integrins, virus engagement at the integrin PSI domain, recapitulates physiologic activation of the integrin as indicated by staining with the activation-specific mAB PAC1. The data also show that blocking of the Gα13 protein from binding to the cytoplasmic domain of the ß3 integrin prevents outside-in signaling and infection. We propose that the cis interaction with P2Y2R provides allosteric resistance to the membrane-normal motion associated with the switchblade model of integrin activation, where the development of tensile force yields physiological integrin activation.


Assuntos
Infecções por Hantavirus/metabolismo , Integrina beta3/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Animais , Células CHO , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Cricetulus , Células Endoteliais , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Orthohantavírus/isolamento & purificação , Humanos , Cadeias beta de Integrinas/metabolismo , Integrina beta3/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Domínios Proteicos , Receptores Purinérgicos P2Y2/genética , Semaforinas/metabolismo , Transdução de Sinais
14.
J Am Soc Nephrol ; 28(5): 1475-1490, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27920156

RESUMO

De novo expression in the kidney of periostin, a protein involved in odontogenesis and osteogenesis, has been suggested as a biomarker of renal disease. In this study, we investigated the mechanism(s) of induction and the role of periostin in renal disease. Using a combination of bioinformatics, reporter assay, and chromatin immunoprecipitation analyses, we found that NFκB and other proinflammatory transcription factors induce periostin expression in vitro and that binding of these factors on the periostin promoter is enriched in glomeruli during experimental GN. Mice lacking expression of periostin displayed preserved renal function and structure during GN. Furthermore, delayed administration of periostin antisense oligonucleotides in wild-type animals with GN reversed already established proteinuria, diminished tissue inflammation, and improved renal structure. Lack of periostin expression also blunted the de novo renal expression of integrin-ß3 and phosphorylation of focal adhesion kinase and AKT, known mediators of integrin-ß3 signaling that affect cell motility and survival, observed during GN in wild-type animals. In vitro, recombinant periostin increased the expression of integrin-ß3 and the concomitant phosphorylation of focal adhesion kinase and AKT in podocytes. Notably, periostin and integrin-ß3 were highly colocalized in biopsy specimens from patients with inflammatory GN. These results demonstrate that interplay between periostin and renal inflammation orchestrates inflammatory and fibrotic responses, driving podocyte damage through downstream activation of integrin-ß3 signaling. Targeting periostin may be a novel therapeutic strategy for treating CKD.


Assuntos
Moléculas de Adesão Celular/fisiologia , Integrina beta3/fisiologia , Nefropatias/etiologia , NF-kappa B/fisiologia , Animais , Feminino , Glomerulonefrite/complicações , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
15.
Oncotarget ; 7(34): 55572-55584, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27487131

RESUMO

Osteopontin (OPN), a phosphorylated glycoprotein, is frequently overexpressed in cancer. Among the three OPN isoforms, OPN-a is the most highly expressed in lung cancer cell lines and lung tumors. Overexpression of OPN-a greatly reduced CL1-5 lung adenocarcinoma cell growth, but had no effect on growth in A549 lung adenocarcinoma cells. Examination of the expression of integrins and CD44, which are possible OPN-a receptors, revealed that differences in integrin ß3 levels might explain this discrepancy between CL1-5 and A549 cells. When integrin ß3 was ectopically expressed in A549 cells, OPN-a inhibited their growth, whereas OPN-a increased cell growth following integrin ß3 knockdown in CL1-5 cells. This OPN-a-induced increase in growth appeared to result from activation of the CD44/NFκB pathway. Our results demonstrated that OPN-a inhibits growth of cells with high integrin ß3 levels and increases growth via activation of the CD44/NFκB pathway in cells with low integrin ß3 levels. Thus, OPN-a, integrin ß3, and CD44 interact to affect lung cancer cell growth, and this study may aid in the development of cancer treatment strategies involving these molecules.


Assuntos
Receptores de Hialuronatos/fisiologia , Integrina beta3/fisiologia , Neoplasias Pulmonares/patologia , Osteopontina/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Receptores de Hialuronatos/análise , Integrina beta3/análise , NF-kappa B/fisiologia , Osteopontina/análise , Splicing de RNA
16.
Cancer Res ; 76(12): 3484-95, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27216180

RESUMO

Integrin ß3 is critical for tumor invasion, neoangiogenesis, and inflammation, making it a promising cancer target. However, preclinical and clinical data of integrin ß3 antagonists have demonstrated no benefit or worse outcomes. We hypothesized that integrin ß3 could affect tumor immunity and evaluated tumors in mice with deletion of integrin ß3 in macrophage lineage cells (ß3KOM). ß3KOM mice had increased melanoma and breast cancer growth with increased tumor-promoting M2 macrophages and decreased CD8(+) T cells. Integrin ß3 antagonist, cilengitide, also enhanced tumor growth and increased M2 function. We uncovered a negative feedback loop in M2 myeloid cells, wherein integrin ß3 signaling favored STAT1 activation, an M1-polarizing signal, and suppressed M2-polarizing STAT6 activation. Finally, disruption of CD8(+) T cells, macrophages, or macrophage integrin ß3 signaling blocked the tumor-promoting effects of integrin ß3 antagonism. These results suggest that effects of integrin ß3 therapies on immune cells should be considered to improve outcomes. Cancer Res; 76(12); 3484-95. ©2016 AACR.


Assuntos
Tolerância Imunológica , Integrina beta3/fisiologia , Neoplasias/imunologia , Animais , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT6/fisiologia , Quinase Syk/metabolismo , Microambiente Tumoral
17.
Ann Rheum Dis ; 75(6): 1211-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26141367

RESUMO

OBJECTIVE: Periarticular and subchondral bone erosion in rheumatoid arthritis caused by osteoclast differentiation and activation is a critical index for diagnosis, therapy and monitoring of the disease. Sorting nexin (SNX) 10, a member of the SNX family which functions in regulation of endosomal sorting, has been implicated to play an important clinical role in malignant osteopetrosis. Here we studied the roles and precise mechanisms of SNX10 in the bone destruction of collagen-induced arthritis (CIA) mice. METHODS: The role of SNX10 in bone destruction was evaluated by a CIA mice model which was induced in male SNX10(-/-) mice and wild type littermates. The mechanism was explored in osteoclasts induced by receptor activator of nuclear factor κB ligand from bone marrow mononuclear cells of wild type and SNX10(-/-) mice. RESULTS: SNX10 knockout prevented bone loss and joint destruction in CIA mice with reduced serum levels of TNF-α, interleukin 1ß and anticollagen IgG 2α antibody. SNX10 deficiency did not block osteoclastogenesis, but significantly impaired osteoclast maturation and bone-resorption function by disturbing the formation of actin belt. The production of TRAP, CtsK and MMP9 in SNX10(-/-) osteoclasts was significantly inhibited, and partially restored by SNX10 overexpression. We further demonstrated that the degradation of NFATc1 was accelerated in SNX10(-/-) osteoclasts causing an inhibition of integrin ß3-Src-PYK2 signalling. CONCLUSIONS: Our study discloses a crucial role and novel mechanism for SNX10 in osteoclast function, and provides evidence for SNX10 as a promising novel therapeutic target for suppression of immune inflammation and bone erosion in rheumatoid arthritis.


Assuntos
Artrite Experimental/complicações , Reabsorção Óssea/prevenção & controle , Fatores de Transcrição NFATC/metabolismo , Nexinas de Classificação/fisiologia , Animais , Artrite Experimental/diagnóstico por imagem , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/etiologia , Reabsorção Óssea/patologia , Integrina beta3/fisiologia , Masculino , Camundongos Knockout , Osteoclastos/patologia , Osteoclastos/fisiologia , Osteogênese/fisiologia , Transdução de Sinais/fisiologia , Nexinas de Classificação/deficiência , Membrana Sinovial/patologia , Tomografia Computadorizada por Raios X
18.
Biomaterials ; 64: 33-44, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26115412

RESUMO

Cancer patients frequently develop skeletal metastases that significantly impact quality of life. Since bone metastases remain incurable, a clearer understanding of molecular mechanisms regulating skeletal metastases is required to develop new therapeutics that block establishment of tumors in bone. While many studies have suggested that the microenvironment contributes to bone metastases, the factors mediating tumors to progress from a quiescent to a bone-destructive state remain unclear. In this study, we hypothesized that the "soil" of the bone microenvironment, specifically the rigid mineralized extracellular matrix, stimulates the transition of the tumor cells to a bone-destructive phenotype. To test this hypothesis, we synthesized 2D polyurethane (PUR) films with elastic moduli ranging from the basement membrane (70 MPa) to cortical bone (3800 MPa) and measured expression of genes associated with mechanotransduction and bone metastases. We found that expression of Integrin ß3 (Iß3), as well as tumor-produced factors associated with bone destruction (Gli2 and parathyroid hormone related protein (PTHrP)), significantly increased with matrix rigidity, and that blocking Iß3 reduced Gli2 and PTHrP expression. To identify the mechanism by which Iß3 regulates Gli2 and PTHrP (both are also known to be regulated by TGF-ß), we performed Förster resonance energy transfer (FRET) and immunoprecipitation, which indicated that Iß3 co-localized with TGF-ß Receptor Type II (TGF-ß RII) on rigid but not compliant films. Finally, transplantation of tumor cells expressing Iß3 shRNA into the tibiae of athymic nude mice significantly reduced PTHrP and Gli2 expression, as well as bone destruction, suggesting a crucial role for tumor-produced Iß3 in disease progression. This study demonstrates that the rigid mineralized bone matrix can alter gene expression and bone destruction in an Iß3/TGF-ß-dependent manner, and suggests that Iß3 inhibitors are a potential therapeutic approach for blocking tumor transition to a bone destructive phenotype.


Assuntos
Integrina beta3/fisiologia , Proteínas de Neoplasias/fisiologia , Osteólise/etiologia , Maleabilidade , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Microambiente Tumoral/fisiologia , Adenocarcinoma/patologia , Adenocarcinoma/secundário , Animais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/secundário , Linhagem Celular Tumoral , Módulo de Elasticidade , Matriz Extracelular/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Integrina beta3/efeitos dos fármacos , Integrina beta3/genética , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína Gli2 com Dedos de Zinco
19.
Exp Dermatol ; 23(9): 682-4, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24828603

RESUMO

Although fibronectin (FN) is known as a chemoattractant for human dermal fibroblasts (HDFs), it is unclear whether HDF migration is stimulated by FN produced by HDFs (autocrine manner) or by keratinocytes (paracrine manner). In this study, we investigated HDF migration by Boyden chamber assay using conditioned media from HDFs and HaCaT cells (keratinocyte cell line). Immunoblotting and enzyme-linked immunosorbent assay revealed that FN existed in both conditioned media. Boyden chamber assay showed both conditioned media stimulated HDF migration, which was inhibited by anti-FN antibody. Antibodies to both integrin ß1and ß3 subunits inhibited HDF migration induced by HDF-conditioned medium almost completely and that by HaCaT cell-conditioned medium with 50-60%. These results suggested that HDF migration was stimulated by FN in both autocrine and paracrine manners. However, the mechanisms of HDF migration by FN, particularly the role of integrin ß1 and ß3 subunits, were slightly different between autocrine and paracrine manners.


Assuntos
Fibroblastos/fisiologia , Fibronectinas/fisiologia , Pele/citologia , Comunicação Autócrina , Linhagem Celular , Movimento Celular/fisiologia , Meios de Cultivo Condicionados , Humanos , Integrina beta1/fisiologia , Integrina beta3/fisiologia , Queratinócitos/fisiologia , Comunicação Parácrina , Fenômenos Fisiológicos da Pele
20.
Exp Eye Res ; 120: 61-70, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24462632

RESUMO

As an early responder to an inflammatory stimulus, neutrophils (PMNs) must exit the vasculature and migrate through the extravascular tissue to the site of insult, which is often remote from the point of extravasation. Following a central epithelial corneal abrasion, PMNs recruited from the peripheral limbal vasculature migrate into the avascular corneal stroma. In vitro studies suggest PMN locomotion over 2-D surfaces is dependent on integrin binding while migration within 3-D matrices can be integrin-independent. Electron micrographs of injured mouse corneas show migrating PMNs make extensive surface contact not only with collagen fibrils in the extracellular matrix (ECM), but also keratocytes. Evidence supporting involvement of integrins in corneal inflammation has prompted research and development of integrin blocking agents for use as anti-inflammatory therapies. However, the role of integrin binding (cell-cell; cell-ECM) during stromal migration in the inflamed cornea has previously not been clearly defined. In this study in vivo time lapse imaging sequences provided the means to quantify cell motility while observing PMN interactions with keratocytes and other stromal components in the living eye. The relative contribution of ß1, ß2 and ß3 integrins to PMN locomotion in the inflamed mouse cornea was investigated using blocking antibodies against the respective integrins. Of the 3 integrin families (ß1, ß2 and ß3) investigated for their potential role in PMN migration, only ß1 antibody blockade produced a significant, but partial, reduction in PMN motility. The preferential migration of PMNs along the keratocyte network was not affected by integrin blockade. Hence, the dominant mechanism for PMN motility within the corneal stroma appears to be integrin-independent as does the restriction of PMN migration paths to the keratocyte network.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Lesões da Córnea , Traumatismos Oculares/metabolismo , Integrinas/fisiologia , Neutrófilos/fisiologia , Ferimentos não Penetrantes/metabolismo , Animais , Anticorpos Bloqueadores , Antígenos CD18/fisiologia , Ceratócitos da Córnea/metabolismo , Substância Própria/citologia , Traumatismos Oculares/fisiopatologia , Feminino , Integrina beta1/fisiologia , Integrina beta3/fisiologia , Ceratite/metabolismo , Ceratite/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Ferimentos não Penetrantes/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA