Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-34768883

RESUMO

Malignant mesothelioma (MM) is a highly aggressive cancer with limited therapeutic options. We have previously shown that the endocytic collagen receptor, uPARAP, is upregulated in certain cancers and can be therapeutically targeted. Public RNA expression data display uPARAP overexpression in MM. Thus, to evaluate its potential use in diagnostics and therapy, we quantified uPARAP expression by immunohistochemical H-score in formalin-fixed paraffin-embedded bioptic/surgical human tissue samples and tissue microarrays. We detected pronounced upregulation of uPARAP in the three main MM subtypes compared to non-malignant reactive mesothelial proliferations, with higher expression in sarcomatoid and biphasic than in epithelioid MM. The upregulation appeared to be independent of patients' asbestos exposure and unaffected after chemotherapy. Using immunoblotting, we demonstrated high expression of uPARAP in MM cell lines and no expression in a non-malignant mesothelial cell line. Moreover, we showed the specific internalization of an anti-uPARAP monoclonal antibody by the MM cell lines using flow cytometry-based assays and confocal microscopy. Finally, we demonstrated the sensitivity of these cells towards sub-nanomolar concentrations of an antibody-drug conjugate formed with the uPARAP-directed antibody and a potent cytotoxin that led to efficient, uPARAP-specific eradication of the MM cells. Further studies on patient cohorts and functional preclinical models will fully reveal whether uPARAP could be exploited in diagnostics and therapeutic targeting of MM.


Assuntos
Lectinas de Ligação a Manose/metabolismo , Glicoproteínas de Membrana/metabolismo , Mesotelioma Maligno/metabolismo , Receptores de Superfície Celular/metabolismo , Adulto , Idoso , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Humanos , Imunoconjugados/metabolismo , Masculino , Lectinas de Ligação a Manose/fisiologia , Glicoproteínas de Membrana/fisiologia , Mesotelioma Maligno/diagnóstico , Mesotelioma Maligno/fisiopatologia , Pessoa de Meia-Idade , Receptores de Superfície Celular/fisiologia , Receptores de Colágeno/genética , Receptores de Colágeno/metabolismo , Receptores de Colágeno/fisiologia , Receptores Mitogênicos/genética , Transcriptoma , Regulação para Cima
2.
Oncogene ; 35(15): 1899-908, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-26148229

RESUMO

Integrin α11ß1 is a stromal cell-specific receptor for fibrillar collagens and is overexpressed in carcinoma-associated fibroblasts (CAFs). We have investigated its direct role in cancer progression by generating severe combined immune deficient (SCID) mice deficient in integrin α11 (α11) expression. The growth of A549 lung adenocarcinoma cells and two patient-derived non-small cell lung carcinoma (NSCLC) xenografts in these α11 knockout (α11(-/-)) mice was significantly impeded, as compared with wild-type (α11(+/+)) SCID mice. Orthotopic implantation of a spontaneously metastatic NCI-H460SM cell line into the lungs of α11(-/-) and α11(+/+) mice showed significant reduction in the metastatic potential of these cells in the α11(-/-) mice. We identified that collagen cross-linking is associated with stromal α11 expression, and the loss of tumor stromal α11 expression was correlated with decreased collagen reorganization and stiffness. This study shows the role of integrin α11ß1, a receptor for fibrillar collagen in differentiation of fibroblasts into CAFs. Furthermore, our data support an important role for α11 signaling pathway in CAFs, promoting tumor growth and metastatic potential of NSCLC cells and being closely associated with collagen cross-linking and the organization and stiffness of fibrillar collagen matrices.


Assuntos
Adenocarcinoma/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Fibroblastos/fisiologia , Integrina beta1/fisiologia , Integrinas/fisiologia , Neoplasias Pulmonares/patologia , Receptores de Colágeno/fisiologia , Células Estromais/fisiologia , Animais , Linhagem Celular Tumoral , Colágeno/metabolismo , Cruzamentos Genéticos , Elasticidade , Proteínas da Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Cadeias alfa de Integrinas , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos SCID , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais
3.
Adv Exp Med Biol ; 819: 73-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25023168

RESUMO

Integrin α11 is the last addition to the vertebrate integrin family. In this chapter we will summarize some basic facts about this integrin and update with information that has been gained in the last decade. Integrin α11ß1 is a major collagen receptor on a subset of fibroblasts. Extensive characterization of the expression pattern in developing mouse embryos has demonstrated expression restricted to subsets of fibroblasts and a transient expression in odontoblasts, but comprehensive characterization of corresponding expression in adult tissues is still lacking. Mice lacking integrin α11 are dwarfed, primarily due to defective incisor eruption defect, which can be traced back to need for α11 on periodontal ligament fibroblasts during incisor eruption. Separate studies have suggested reduced levels of IGF-1 in mice lacking α11. Analysis of lung cancer has identified α11ß1 as a functional important collagen receptor on carcinoma associated fibroblasts (CAFs) and a number of disease models are awaiting analysis to see the importance of this collagen receptor in pathological models.


Assuntos
Fibroblastos/fisiologia , Integrinas/fisiologia , Receptores de Colágeno/fisiologia , Animais , Éxons , Humanos , Integrinas/genética , Camundongos , Regiões Promotoras Genéticas , Receptores de Colágeno/genética
4.
J Immunol ; 191(12): 5941-50, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24244022

RESUMO

Th17 cells play a critical role in the pathogenesis of rheumatoid arthritis (RA), but the mechanisms by which these cells regulate the development of RA are not fully understood. We have recently shown that α2ß1 integrin, the receptor of type I collagen, is the major collagen-binding integrin expressed by human Th17 cells. In this study, we examined the role of α2ß1 integrin in Th17-mediated destructive arthritis in the murine model of collagen-induced arthritis (CIA). We found that α2ß1 integrin is expressed on synovial Th17 cells from CIA mice and its neutralization with a specific mAb significantly reduced inflammation and cartilage degradation, and protected the mice from bone erosion. Blockade of α2ß1 integrin led to a decrease in the number of Th17 cells in the joints and to a reduction of IL-17 levels in CIA mice. This was associated with an inhibition of receptor activator of NF-κB ligand levels and osteoclast numbers, and reduction of bone loss. We further show that α2ß1 integrin is expressed on synovial Th17 cells from RA patients, and that its ligation with collagen costimulated the production of IL-17 by polarized human Th17 cells by enhancing the expression of retinoic acid receptor-related orphan receptor C through ERK and PI3K/AKT. Our findings provide the first evidence, to our knowledge, that α2ß1 integrin is an important pathway in Th17 cell activation in the pathogenesis of CIA, suggesting that its blockade can be beneficial for the treatment of RA and other Th17-associated autoimmune diseases.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Artrite Experimental/terapia , Artrite Reumatoide/metabolismo , Integrina alfa2beta1/fisiologia , Osteólise/prevenção & controle , Receptores de Colágeno/fisiologia , Células Th17/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Reumatoide/imunologia , Cartilagem Articular/patologia , Colágeno/farmacologia , Cricetinae , Regulação para Baixo , Feminino , Humanos , Inflamação , Integrina alfa2beta1/antagonistas & inibidores , Interleucina-17/sangue , Ativação Linfocitária , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos DBA , NF-kappa B/fisiologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Osteoclastos/patologia , Osteólise/etiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ligante RANK/sangue , Receptores de Colágeno/antagonistas & inibidores , Transdução de Sinais , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Células Th17/fisiologia
5.
J Clin Invest ; 123(12): 5071-81, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24270421

RESUMO

von Willebrand disease type 2B (vWD-type 2B) is characterized by gain-of-function mutations in von Willebrand factor (vWF) that enhance its binding to the glycoprotein Ib-IX-V complex on platelets. Patients with vWD-type 2B have a bleeding tendency that is linked to loss of vWF multimers and/or thrombocytopenia. In this study, we uncovered evidence that platelet dysfunction is a third possible mechanism for bleeding tendency. We found that platelet aggregation, secretion, and spreading were diminished due to inhibition of integrin αIIbß3 in platelets from mice expressing a vWD-type 2B-associated vWF (vWF/p.V1316M), platelets from a patient with the same mutation, and control platelets pretreated with recombinant vWF/p.V1316M. Impaired platelet function coincided with reduced thrombus growth. Further, αIIbß3 activation and activation of the small GTPase Rap1 were impaired by vWF/p.V1316M following exposure to platelet agonists (thrombin, ADP, or convulxin). Conversely, thrombin- or ADP-induced Ca2+ store release, which is required for αIIbß3 activation, was normal, indicating that vWF/p.V1316M acts downstream of Ca2+ release and upstream of Rap1. We found normal Syk phosphorylation and PLCγ2 activation following collagen receptor signaling, further implying that vWF/p.V1316M acts directly on or downstream of Ca2+ release. These data indicate that the vWD-type 2B mutation p.V1316M is associated with severe thrombocytopathy, which likely contributes to the bleeding tendency in vWD-type 2B.


Assuntos
Substituição de Aminoácidos , Transtornos Hemorrágicos/etiologia , Mutação de Sentido Incorreto , Agregação Plaquetária/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Mutação Puntual , Doença de von Willebrand Tipo 2/genética , Fator de von Willebrand/genética , Trifosfato de Adenosina/metabolismo , Animais , Plaquetas/metabolismo , Sinalização do Cálcio/fisiologia , Transtornos Hemorrágicos/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipase C gama/fisiologia , Fosforilação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/fisiologia , Receptores de Colágeno/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Quinase Syk , Proteínas rap1 de Ligação ao GTP/metabolismo , Doença de von Willebrand Tipo 2/sangue , Fator de von Willebrand/fisiologia
6.
Am J Physiol Cell Physiol ; 303(12): C1207-17, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23015544

RESUMO

As increase in collagen deposition is no longer taken as simply a consequence but, rather, an inducer of disease progression; therefore, the understanding of collagen signal transduction is fundamentally important. Cells contain at least two types of collagen receptors: integrins and discoidin domain receptors (DDRs). The integrin heterodimers α(1)ß(1), α(2)ß(1), α(10)ß(1), and α(11)ß(1) are recognized as the non-tyrosine kinase collagen receptors. DDR1 and 2, the tyrosine kinase receptors of collagen, are specifically expressed in epithelium and mesenchyme, respectively. While integrin ß(1) and DDR1 are both required for cell adhesion on collagen, their roles in epithelial cell differentiation during development and disease progression seem to counteract each other, with integrin ß(1) favoring epithelium mesenchyme transition (EMT) and DDR1 inducing epithelial cell differentiation. The in vitro evidence shows that the integrin ß(1) and DDR1 exert opposing actions in regulation of membrane stability of E-cadherin, which itself is a critical regulator of epithelial cell differentiation. Here, we review the functional roles of integrin ß(1) and DDR1 in regulation of epithelial cell differentiation during development and disease progression, and explore the underlining mechanisms regarding to the regulation of membrane stability of E-cadherin.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Integrina beta1/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Colágeno/fisiologia , Receptores Mitogênicos/fisiologia , Animais , Caderinas/fisiologia , Colágeno/fisiologia , Receptores com Domínio Discoidina , Feminino , Fibrose , Humanos , Mesoderma/fisiologia , Camundongos , Neoplasias/patologia
7.
Acta Haematol ; 128(4): 233-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22922528

RESUMO

INTRODUCTION: Ligands acting at the platelet collagen receptor, glycoprotein (GP)VI, induce intracellular FcRγ/Syk-dependent signaling pathways and Syk-dependent or Syk-independent generation of intracellular reactive oxygen species (ROS). Additional signaling-dependent or signaling-independent pathways lead to metalloproteinase-mediated shedding of GPVI. AIM: Analysis of platelet GPVI expression and signaling in a patient with a collagen-selective defect associated with myelodysplastic syndrome (MDS) uniquely demonstrates divergent pathways leading to ROS generation and Syk phosphorylation in human platelets. METHODS: Surface expression of GPVI and ligand-induced ROS generation was quantitated by flow cytometry. GPVI shedding and Syk phosphorylation were analyzed by Western blot. RESULTS: Despite platelet count/size and GPVI surface expression within normal ranges, platelet-rich plasma showed no aggregation in response to collagen or GPVI-selective agonist collagen-related peptide, but aggregated in response to other agonists, consistent with dysfunctional GPVI signaling. We observed rapid GPVI-dependent Syk-independent ROS generation and disulfide-dependent GPVI homodimerization, but not Syk-dependent ROS or ligand-induced shedding. Temporal analysis showed a gradual decline in platelet count and the appearance of ligand-induced phosphorylation of an ∼40-kDa Syk fragment. CONCLUSIONS: These studies show that GPVI ligation in platelets induces intracellular ROS production independent of either Syk activation or divergent pathways leading to platelet aggregation or ectodomain shedding.


Assuntos
Síndromes Mielodisplásicas/fisiopatologia , Glicoproteínas da Membrana de Plaquetas/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Colágeno/fisiologia , Transdução de Sinais/fisiologia , Idoso , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes , Proteínas Tirosina Quinases/metabolismo , Quinase Syk
8.
Internist (Berl) ; 51(9): 1109-14, 1116-7, 2010 Sep.
Artigo em Alemão | MEDLINE | ID: mdl-20725709

RESUMO

Inherited thrombocytopathies are much less frequent in comparison to acquired platelet function disorders. However, congenital disorders can lead to severe bleeding tendency and are often not diagnosed. They are induced by different platelet defects based on disorders of platelet adhesion, receptors, secretion and signal transduction. In some cases they are associated with thrombocytopenia, giant platelets and various comorbidities. This article gives an overview regarding diverse defects, their diagnosis and treatment options.


Assuntos
Transtornos Plaquetários/genética , Plaquetas/fisiologia , Genótipo , Transtornos Hemorrágicos/genética , Síndrome de Bernard-Soulier/sangue , Síndrome de Bernard-Soulier/diagnóstico , Síndrome de Bernard-Soulier/genética , Síndrome de Bernard-Soulier/terapia , Transtornos Plaquetários/sangue , Transtornos Plaquetários/diagnóstico , Transtornos Plaquetários/terapia , Plaquetas/patologia , Diagnóstico Diferencial , Transtornos Hemorrágicos/sangue , Transtornos Hemorrágicos/diagnóstico , Transtornos Hemorrágicos/terapia , Humanos , Programas de Rastreamento , Agregação Plaquetária/genética , Agregação Plaquetária/fisiologia , Testes de Função Plaquetária , Deficiência do Pool Plaquetário/sangue , Deficiência do Pool Plaquetário/diagnóstico , Deficiência do Pool Plaquetário/genética , Deficiência do Pool Plaquetário/terapia , Prostaglandina-Endoperóxido Sintases/genética , Receptores de Colágeno/genética , Receptores de Colágeno/fisiologia , Trombastenia/sangue , Trombastenia/diagnóstico , Trombastenia/genética , Trombastenia/terapia , Tromboxano-A Sintase/genética
9.
Oncogene ; 29(16): 2368-80, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20101206

RESUMO

Matrix remodeling, degradation, inflammation and invasion liberate peptide fragments that can subsequently interact with cells in an attachment-independent manner. Such 'soluble' matrix components, including collagens, fibronectin and laminin, induced Smad activation (termed crosstalk signaling), which follows a similar chronological sequence and R-Smad specificity as induced by transforming growth factor (TGF)-beta1. Smad4 nuclear translocation occurred in response to collagen binding, indicating downstream signal propagation. TGF-beta scavenging antibody affected only TGF-beta1, but not crosstalk-induced responses. TGF-beta type II receptor mutation (DR26Delta25), which is deficient in TGF-beta type I receptor recruitment to the ligand, induced a heterotetramer signaling complex, and propagated Smad2 activation only through collagen induction and not TGF-beta signaling. Consequentially, TGF-beta ligand participation is not required for crosstalk signaling. This signaling requires a functional integrin beta1 receptor as showed by RNA interference. Co-immunoprecipitation (co-IP) and fluorescent microscopy indicate the involvement of focal adhesion kinase (FAK) and Src activity in collagen-induced signal propagation, and suggest a membrane signaling complex formation that includes both TGF-beta receptors and integrins. The related gene expressional responses are distinct from that evoked by TGF-beta1, supporting its separate function. This signaling mechanism expands and partially explains TGF-beta receptor dynamics and consequential signaling diversity-related gene expressional plasticity.


Assuntos
Matriz Extracelular/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais/fisiologia , Linhagem Celular Tumoral , Colágeno/farmacologia , Humanos , Integrina beta1/fisiologia , Modelos Biológicos , Fosforilação , Receptor Cross-Talk/fisiologia , Receptores de Colágeno/fisiologia , Proteínas Smad/metabolismo
10.
J Dent Res ; 88(7): 621-6, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19641149

RESUMO

We previously demonstrated a role for alpha11beta1 integrin in periodontal ligament (PDL)-driven tooth eruption in the mouse. To explore a possible role for alpha11beta1 in the human periodontium, we have characterized the expression and function of alpha11 in human PDL tissue, in human PDL fibroblasts (hPDLF), and in human gingival fibroblasts (hGF). alpha11 expression was detected in PDL tissue, in hPDLF, and in hGF cells. Platelet-derived growth factor-BB and insulin-like growth factor II stimulated contraction of collagen lattices by both types of fibroblasts. alpha2 integrin blocking antibodies and the use of alpha11 siRNA demonstrated a role for both alpha2beta1 and alpha11beta1 in collagen lattice remodeling. Analysis of the proximal ITGA11 promoter from persons with chronic periodontal disease failed to reveal any polymorphism. Analysis of our data shows that alpha11beta1 is a major collagen receptor on cultured human PDL cells and implies that it is also functionally important in the PDL in vivo.


Assuntos
Periodontite Crônica/metabolismo , Colágeno Tipo I/metabolismo , Integrinas/fisiologia , Ligamento Periodontal/metabolismo , Receptores de Colágeno/fisiologia , Adolescente , Adulto , Idoso , Sequência de Bases , Becaplermina , Estudos de Casos e Controles , Células Cultivadas , Colágeno Tipo I/efeitos dos fármacos , Fibroblastos/fisiologia , Gengiva/citologia , Gengiva/metabolismo , Humanos , Fator de Crescimento Insulin-Like II/farmacologia , Cadeias alfa de Integrinas/antagonistas & inibidores , Cadeias alfa de Integrinas/genética , Integrinas/genética , Pessoa de Meia-Idade , Dados de Sequência Molecular , Ligamento Periodontal/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Reação em Cadeia da Polimerase , Polimorfismo Genético , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-sis , Interferência de RNA , Receptores de Colágeno/genética , Adulto Jovem
11.
J Leukoc Biol ; 83(4): 799-803, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18063695

RESUMO

The immune system protects the body from invaders such as viruses and bacteria. Immune cells must be activated in the correct context to function properly. It is critical that the receptors, costimulatory molecules, and cytokines that orchestrate this activation are carefully regulated to prevent uncontrolled inflammation and autoimmunity. Inhibitory receptors play an important role in regulation of immune cell function, usually upon interaction with ligands present on other cells. In contrast, the function of the inhibitory leukocyte-associated Ig-like receptor (LAIR)-1 can be regulated by extracellular matrix collagens. LAIR-1 is expressed on most cells of the immune system, and its function has been studied on multiple cell types. This review summarizes current literature about LAIR-1, a receptor that potentially is able to regulate multiple steps of an immune response.


Assuntos
Receptores Imunológicos/fisiologia , Proteína Tirosina Quinase CSK , Clonagem Molecular , Homeostase , Humanos , Células Matadoras Naturais/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Colágeno/fisiologia , Receptores Imunológicos/química , Receptores Imunológicos/genética , Linfócitos T/imunologia , Domínios de Homologia de src , Quinases da Família src
12.
Circ Res ; 100(12): 1673-85, 2007 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-17585075

RESUMO

Platelet adhesion is an essential function in response to vascular injury and is generally viewed as the first step during which single platelets bind through specific membrane receptors to cellular and extracellular matrix constituents of the vessel wall and tissues. This response initiates thrombus formation that arrests hemorrhage and permits wound healing. Pathological conditions that cause vascular alterations and blood flow disturbances may turn this beneficial process into a disease mechanism that results in arterial occlusion, most frequently in atherosclerotic vessels of the heart and brain. Besides their relevant role in hemostasis and thrombosis, platelet adhesive properties are central to a variety of pathophysiological processes that extend from inflammation to immune-mediated host defense and pathogenic mechanisms as well as cancer metastasis. All of these activities depend on the ability of platelets to circulate in blood as sentinels of vascular integrity, adhere where alterations are detected, and signal the abnormality to other platelets and blood cells. In this respect, therefore, platelet adhesion to vascular wall structures, to one another (aggregation), or to other blood cells, represent different aspects of the same fundamental biological process. Detailed studies by many investigators over the past several years have been aimed to dissect the complexity of these functions, and the results obtained now permit an attempt to integrate all the available information into a picture that highlights the balanced diversity and synergy of distinct platelet adhesive interactions.


Assuntos
Plaquetas/fisiologia , Adesividade Plaquetária/fisiologia , Animais , Aterosclerose/fisiopatologia , Moléculas de Adesão Celular/fisiologia , Matriz Extracelular/fisiologia , Hemostasia/fisiologia , Humanos , Inflamação/fisiopatologia , Agregação Plaquetária/fisiologia , Receptores de Colágeno/fisiologia , Trombose/fisiopatologia
13.
Mol Cell Biol ; 27(12): 4306-16, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17420280

RESUMO

The fibroblast integrin alpha11beta1 is a key receptor for fibrillar collagens. To study the potential function of alpha11 in vivo, we generated a null allele of the alpha11 gene. Integrin alpha11(-/-) mice are viable and fertile but display dwarfism with increased mortality, most probably due to severely defective incisors. Mutant incisors are characterized by disorganized periodontal ligaments, whereas molar ligaments appear normal. The primary defect in the incisor ligament leads to halted tooth eruption. alpha11beta1-defective embryonic fibroblasts displayed severe defects in vitro, characterized by (i) greatly reduced cell adhesion and spreading on collagen I, (ii) reduced ability to retract collagen lattices, and (iii) reduced cell proliferation. Analysis of matrix metalloproteinase in vitro and in vivo revealed disturbed MMP13 and MMP14 synthesis in alpha11(-/-) cells. We show that alpha11beta1 is the major receptor for collagen I on mouse embryonic fibroblasts and suggest that alpha11beta1 integrin is specifically required on periodontal ligament fibroblasts for cell migration and collagen reorganization to help generate the forces needed for axial tooth movement. Our data show a unique role for alpha11beta1 integrin during tooth eruption.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Incisivo/fisiologia , Integrinas/fisiologia , Ligamento Periodontal/metabolismo , Receptores de Colágeno/fisiologia , Erupção Dentária , Animais , Blastocisto , Linhagem Celular Transformada , Movimento Celular , Transformação Celular Viral , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Homozigoto , Imuno-Histoquímica , Incisivo/citologia , Integrinas/deficiência , Integrinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Microinjeções , Ligamento Periodontal/citologia , Receptores de Colágeno/deficiência , Receptores de Colágeno/genética
14.
Arterioscler Thromb Vasc Biol ; 26(12): 2588-93, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17023681

RESUMO

The immunoreceptor signaling pathway has classically been defined by its role in mediating intracellular signals downstream of immune receptors on circulating cells, but recent studies have revealed new and unexpected roles for this pathway in vascular biology. In platelets the immunoreceptor signaling pathway is coupled to 2 structurally distinct platelet collagen receptors, glycoprotein VI and integrin alpha2beta1, and is required for the activation of platelets after exposure to vessel wall collagen during plaque rupture. During vascular development immunoreceptor signaling is required for proper formation of the lymphatic system, a role that has revealed the contribution of hematopoietic endothelial progenitors to that process. In conjunction with the identification of new biological roles in vascular cell types, new molecular mechanisms of activating this signaling pathway have been discovered, including activation by integrins and immunoreceptor tyrosine activation motifs (ITAMs) on receptors that do not function as part of the immune response. Here we discuss some of these recent findings and their implications for vascular biology and the treatment of human vascular diseases.


Assuntos
Plaquetas/fisiologia , Células Endoteliais/fisiologia , Receptores Imunológicos/fisiologia , Transdução de Sinais/fisiologia , Animais , Sistema Hematopoético/citologia , Sistema Hematopoético/fisiologia , Humanos , Sistema Linfático/irrigação sanguínea , Receptores de Colágeno/fisiologia
15.
Cancer Res ; 66(17): 8648-54, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16951179

RESUMO

The most frequent site of prostate cancer metastasis is the bone. Adhesion to bone-specific factors may facilitate the selective metastasis of prostate cancer to the skeleton. Therefore, we tested whether prostate cancer bone metastasis is mediated by binding to type I collagen, the most abundant bone protein. We observed that only bone metastatic prostate cancer cells bound collagen I, whereas cells that form only visceral metastases failed to bind collagen. To confirm the relationship between collagen adhesion and bone metastatic potential, a collagen-binding variant of human LNCaP prostate cancer cells was derived through serial passage on type I collagen (LNCaP(col)). Fluorescence-activated cell sorting analysis showed that LNCaP(col) cells express increased levels of the integrin collagen I receptor alpha(2)beta(1) compared with LNCaP cells. Antibodies to the alpha(2)beta(1) complex inhibited LNCaP(col) binding to collagen, confirming that integrins mediated the attachment. Correspondingly, LNCaP(col) cells displayed enhanced chemotactic migration toward collagen I compared with LNCaP cells, an activity that could be blocked with alpha(2)beta(1) antibodies. To directly test the role of alpha(2)beta(1)-dependent collagen binding in bone metastasis, LNCaP and LNCaP(col) cells were injected into the tibia of nude mice. After 9 weeks, 7 of 13 (53%) mice injected with LNCaP(col) developed bone tumors, whereas 0 of 8 mice injected with LNCaP cells had evidence of boney lesions. LNCaP(col) cells were found to express increased levels of the metastasis-promoting RhoC GTPase compared with parental LNCaP. We conclude that collagen I attachment mediated by alpha(2)beta(1) initiates motility programs through RhoC and suggest a mechanism for prostate cancer metastasis to the bone.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Próstata/patologia , Receptores de Colágeno/fisiologia , Animais , Neoplasias Ósseas/fisiopatologia , Adesão Celular , Divisão Celular , Linhagem Celular Tumoral , Variação Genética , Humanos , Integrina alfa2beta1 , Integrinas/fisiologia , Masculino , Camundongos , Camundongos Nus , Metástase Neoplásica , Transdução de Sinais , Transplante Heterólogo
16.
J Biol Chem ; 279(52): 53955-62, 2004 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-15456756

RESUMO

Vav family proteins are guanine nucleotide exchange factors for the Rho/Rac family of small GTP-binding proteins. In addition, they have domains that mediate protein-protein interactions, including one Src homology 2 (SH2) and two Src homology 3 (SH3) domains. Vav1, Vav2, and Vav3 play a crucial role in the regulation of phospholipase C gamma (PLC gamma) isoforms by immuno-tyrosine-based activation motif (ITAM)-coupled receptors, including the T- and B-cell antigen receptors. We have reported in platelets, however, that Vav1 and Vav2 are not required for activation of PLC gamma 2 in response to stimulation of the ITAM-coupled collagen receptor glycoprotein VI (GPVI). Here we report that Vav3 is tyrosinephosphorylated upon activation of GPVI but that Vav3-deficient platelets also exhibit a normal response upon activation of the ITAM receptor. In sharp contrast, platelets deficient in both Vav1 and Vav3 show a marked inhibition of aggregation and spreading upon activation of GPVI, which is associated with a reduction in tyrosine phosphorylation of PLC gamma 2. The phenotype of Vav1/2/3 triple-deficient platelets is similar to that of Vav1/3 double-deficient cells. These results demonstrate that Vav3 and Vav1 play crucial but redundant roles in the activation of PLC gamma 2 by GPVI. This is the first time that absolute redundancy between two protein isoforms has been observed with respect to the regulation of PLC gamma 2 in platelets.


Assuntos
Colágeno/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Tirosina Quinase da Agamaglobulinemia , Animais , Plaquetas/química , Plaquetas/fisiologia , Antígenos CD36/química , Antígenos CD36/fisiologia , Proteínas de Ciclo Celular , Ativação Enzimática/fisiologia , Precursores Enzimáticos/metabolismo , Fatores de Troca do Nucleotídeo Guanina , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Proteínas Oncogênicas/deficiência , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/fisiologia , Fosfolipase C gama , Fosforilação , Fosfotirosina/metabolismo , Ativação Plaquetária/fisiologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Proto-Oncogênicas c-vav , Receptores de Colágeno/química , Receptores de Colágeno/fisiologia , Quinase Syk , Fosfolipases Tipo C/metabolismo , Quinases da Família src/metabolismo
17.
Leuk Lymphoma ; 44(9): 1569-77, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14565661

RESUMO

The leflunomide metabolite analog alpha-cyano-beta-hydroxy-beta-methyl-N-(2,5-dibromophenyl)-propenamide (LFM-A13) is a rationally-designed specific inhibitor of the TEC family protein tyrosine kinase, Bruton's tyrosine kinase (BTK) which plays an important role in platelet physiology by regulating the glycoprotein GPVI-FcRgamma-coupled collagen receptor signaling pathway. At low micromolar concentrations, LFM-A13 inhibited collagen-induced ultrastructural changes indicative of activation. LFM-A13 inhibited collagen (but not thrombin, TRAP-6, or ADP)-induced platelet aggregation in a concentration-dependent fashion with an IC50 value of 2.8 microM. LFM-A13 was not toxic to mice when administered systemically at dose levels ranging from 1 to 100 mg/kg. At nontoxic dose levels, LFM-A13 prolonged the tail bleeding times of mice and improved event-free survival in two mouse models of agonist-induced invariably fatal pulmonary thromboembolism. To our knowledge, LFM-A13 is the first anti-thrombotic agent which prevents platelet aggregation by inhibiting BTK.


Assuntos
Amidas/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Fibrinolíticos/uso terapêutico , Nitrilas/uso terapêutico , Inibidores da Agregação Plaquetária/uso terapêutico , Agregação Plaquetária/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Embolia Pulmonar/prevenção & controle , Tromboembolia/prevenção & controle , Difosfato de Adenosina/farmacologia , Tirosina Quinase da Agamaglobulinemia , Amidas/farmacologia , Animais , Aspirina/farmacologia , Aspirina/uso terapêutico , Tempo de Sangramento , Domínio Catalítico/efeitos dos fármacos , Colágeno/antagonistas & inibidores , Colágeno/farmacologia , Colágeno/toxicidade , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Fibrinolíticos/farmacologia , Humanos , Janus Quinase 3 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nitrilas/farmacologia , Fragmentos de Peptídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/fisiologia , Proteínas Tirosina Quinases/química , Embolia Pulmonar/induzido quimicamente , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Receptores de Colágeno/efeitos dos fármacos , Receptores de Colágeno/fisiologia , Transdução de Sinais/efeitos dos fármacos , Trombina/farmacologia , Tromboembolia/induzido quimicamente , Tromboplastina/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA