Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 119(11): 3384-94, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19841537

RESUMO

When plasma levels of homocysteine (HC), a thiol amino acid formed upon methionine demethylation, exceed 12 muM, individuals are at increased risk of developing large vessel atherothrombosis and small vessel dysfunction. The annexin A2 complex (termed "A2") is the cell surface coreceptor for plasminogen and TPA and accelerates the catalytic activation of plasmin, the major fibrinolytic agent in mammals. We previously showed that HC prevents A2-mediated, TPA-dependent activation of plasminogen in vitro by disulfide derivatization of the "tail" domain of A2. We also demonstrated that fibrinolysis and angiogenesis are severely impaired in A2-deficient mice. We now report here that, although hyperhomocysteinemic mice had a normal coagulation profile and normal platelet function, fibrin accumulated in their tissues due to reduced perivascular fibrinolytic activity and angiogenesis was impaired. A2 isolated from hyperhomocysteinemic mice failed to fully support TPA-dependent plasmin activation. However, infusion of hyperhomocysteinemic mice with fresh recombinant A2, which localized to neoangiogenic endothelial cells, resulted in normalization of angiogenesis and disappearance of peri- and intravascular fibrin. We therefore conclude that hyperhomocysteinemia impairs postnatal angiogenesis by derivatizing A2, preventing perivascular fibrinolysis, and inhibiting directed endothelial cell migration. These findings provide a mechanistic explanation for microvascular dysfunction and macrovascular occlusion in individuals with hyperhomocysteinemia.


Assuntos
Anexina A2/metabolismo , Fibrinólise/fisiologia , Homocisteína/metabolismo , Neovascularização Fisiológica/fisiologia , Animais , Anexina A2/farmacologia , Movimento Celular/efeitos dos fármacos , Neovascularização da Córnea/fisiopatologia , Dieta , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Fibrina/metabolismo , Fibrinólise/efeitos dos fármacos , Homocisteína/farmacologia , Hiper-Homocisteinemia/fisiopatologia , Masculino , Metionina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Recombinantes/farmacologia
2.
J Biol Chem ; 283(28): 19192-200, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18434302

RESUMO

The annexin A2 (A2) heterotetramer, consisting of two copies of A2 and two copies of S100A10/p11, promotes fibrinolytic activity on the surface of vascular endothelial cells by assembling plasminogen and tissue plasminogen activator (tPA) and accelerating the generation of plasmin. In humans, overexpression of A2 by acute promyelocytic leukemia cells is associated with excessive fibrinolysis and hemorrhage, whereas anti-A2 autoantibodies appear to accentuate the risk of thrombosis in patients with anti-phospholipid syndrome. Complete deficiency of A2 in mice leads to a lack of tPA cofactor activity, accumulation of intravascular fibrin, and failure to clear arterial thrombi. Within the endothelial cell, p11 is required for Src kinase-mediated tyrosine phosphorylation of A2, which signals translocation of both proteins to the cell surface. Here we show that p11 is expressed at very low levels in the absence of A2 both in vitro and in vivo. We demonstrate further that unpartnered p11 becomes polyubiquitinated and degraded via a proteasome-dependent mechanism. A2 stabilizes intracellular p11 through direct binding, thus masking an autonomous p11 polyubiquitination signal that triggers proteasomal degradation. This interaction requires both the p11-binding N-terminal domain of A2 and the C-terminal domain of p11. This mechanism prevents accumulation of free p11 in the endothelial cell and suggests that regulation of tPA-dependent cell surface fibrinolytic activity is precisely tuned to the intracellular level of p11.


Assuntos
Anexina A2/metabolismo , Células Endoteliais/metabolismo , Proteínas S100/metabolismo , Ubiquitinação , Animais , Anexina A2/genética , Síndrome Antifosfolipídica/sangue , Síndrome Antifosfolipídica/genética , Autoanticorpos/sangue , Linhagem Celular Tumoral , Fibrina/genética , Fibrina/metabolismo , Fibrinolisina/genética , Fibrinolisina/metabolismo , Fibrinólise/genética , Hemorragia/sangue , Hemorragia/etiologia , Hemorragia/genética , Humanos , Leucemia Promielocítica Aguda/sangue , Leucemia Promielocítica Aguda/complicações , Leucemia Promielocítica Aguda/genética , Camundongos , Camundongos Knockout , Proteína Oncogênica pp60(v-src)/genética , Proteína Oncogênica pp60(v-src)/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica/genética , Transporte Proteico/genética , Proteínas S100/genética , Trombose/sangue , Trombose/etiologia , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinação/genética
3.
Blood ; 107(11): 4375-82, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16493010

RESUMO

The association of thrombosis and gestational morbidity with antiphospholipid antibodies is termed antiphospholipid syndrome (APS). Annexin 2 (A2) is a profibrinolytic endothelial cell surface receptor that binds plasminogen, its tissue activator (tPA), and beta(2)-glycoprotein I (beta2GPI), the main antigen for antiphospholipid antibodies. Here, we evaluate A2 as a target antigen in APS. Serum samples from 434 individuals (206 patients with systemic lupus erythematosus without thrombosis, 62 with APS, 21 with nonautoimmune thrombosis, and 145 healthy individuals) were analyzed by enzyme-linked immunosorbent assay (ELISA) and immunoblot for antiphospholipid and A2 antibodies. Anti-A2 antibodies (titer > 3 SDs) were significantly more prevalent in patients with APS (22.6%; venous, 17.5%; arterial, 34.3%; and mixed thrombosis, 40.4%) than in healthy individuals (2.1%, P < .001), patients with nonautoimmune thrombosis (0%, P = .017), or patients with lupus without thrombosis (6.3%, P < .001). Anti-A2 IgG enhanced the expression of tissue factor on endothelial cells (6.4-fold +/- 0.13-fold SE), blocked A2-supported plasmin generation in a tPA-dependent generation assay (19%-71%) independently of beta2GPI, and inhibited cell surface plasmin generation on human umbilical vein endothelial cells (HUVECs) by 34% to 83%. We propose that anti-A2 antibodies contribute to the prothrombotic diathesis in antiphospholipid syndrome.


Assuntos
Anexina A2/imunologia , Síndrome Antifosfolipídica/etiologia , Autoanticorpos/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Síndrome Antifosfolipídica/complicações , Síndrome Antifosfolipídica/imunologia , Estudos de Casos e Controles , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Feminino , Fibrinolisina/biossíntese , Humanos , Imunoensaio , Masculino , Pessoa de Meia-Idade , Gravidez , Complicações na Gravidez , Tromboplastina/biossíntese , Trombose/etiologia
4.
J Biol Chem ; 279(42): 43411-8, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15302870

RESUMO

Annexin 2 is a profibrinolytic co-receptor for plasminogen and tissue plasminogen activator that stimulates activation of the major fibrinolysin, plasmin, at cell surfaces. In human subjects, overexpression of annexin 2 in acute promyelocytic leukemia leads to a bleeding diathesis reflective of excessive cell surface annexin 2-dependent generation of plasmin (Menell, J. S., Cesarman, G. M., Jacovina, A. T., McLaughlin, M. A., Lev, E. A., and Hajjar, K. A. (1999) N. Engl. J. Med. 340, 994-1004). In addition, mice completely deficient in annexin 2 display fibrin accumulation within blood vessels and impaired clearance of injury-induced thrombi (Ling Q., Jacovina, A.T., Deora, A.B., Febbraio, M., Simantov, R., Silverstein, R. L., Hempstead, B. L., Mark, W., and Hajjar, K. A. (2004) J. Clin. Investig. 113, 38-48). Here, we show that endothelial cell annexin 2, a protein that lacks a typical signal peptide, translocates from the cytoplasm to the extracytoplasmic plasma membrane in response to brief temperature stress both in vitro and in vivo in the absence of cell death or cell lysis. This regulated response is independent of new protein or mRNA synthesis and does not require the classical endoplasmic reticulum-Golgi pathway. Temperature stress-induced annexin 2 translocation is dependent on both expression of protein p11 (S100A10) and tyrosine phosphorylation of annexin 2 because annexin 2 release is completely eliminated on depletion of p11, inactivation of tyrosine kinase, or mutation of tyrosine 23. Translocation of annexin 2 to the cell surface dramatically increases tissue plasminogen activator-dependent plasminogen activation potential and may represent a novel stress-induced protein secretion pathway.


Assuntos
Anexina A2/metabolismo , Membrana Celular/metabolismo , Endotélio Vascular/metabolismo , Proteínas S100/metabolismo , Anexina A2/genética , Apoptose , Células Cultivadas , Clonagem Molecular , DNA Complementar , Humanos , Cinética , Fosforilação , Transporte Proteico , Proteínas Recombinantes/metabolismo , Termodinâmica , Veias Umbilicais
5.
Blood ; 103(1): 317-24, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14504107

RESUMO

Monocytes and macrophages participate in a wide variety of host defense mechanisms. Annexin II, a fibrinolytic receptor, binds plasminogen and tissue plasminogen activator (t-PA) independently at the cell surface, thereby enhancing the catalytic efficiency of plasmin production. We demonstrated previously that annexin II on the surface of both cultured monocytoid cells and monocyte-derived macrophages promotes their ability to remodel extracellular matrix. Here, we demonstrate that human peripheral blood monocytes represent the major circulating annexin II-expressing cell. Annexin II supported t-PA-dependent generation of cell surface plasmin and the matrix-penetrating activity of human monocytes. Compared to polymorphonuclear leukocytes, monocytes supported a 12.9-fold greater rate of plasmin generation in the presence of exogenous t-PA, and this activity was largely attributable to annexin II. Likewise, anti-annexin II IgG directed against the t-PA-binding tail domain inhibited plasminogen-dependent, cytokine-directed monocyte migration through extracellular matrix. On differentiation of monocytes to macrophages, there was a 2.4-fold increase in annexin II-specific mRNA, and a 7.9-fold increase in surface annexin II. Thioglycolate-elicited peritoneal macrophages, furthermore, displayed an additional 3.8-fold increase in annexin II surface expression compared with resident cells. Thus, annexin II-mediated assembly of plasminogen and t-PA on monocyte/macrophages contributes to plasmin generation, matrix remodeling, and directed migration.


Assuntos
Anexina A2/fisiologia , Macrófagos/fisiologia , Monócitos/fisiologia , Plasminogênio/fisiologia , Anexina A2/antagonistas & inibidores , Diferenciação Celular , Movimento Celular/fisiologia , Matriz Extracelular/fisiologia , Humanos , Imunoglobulina G/farmacologia , Técnicas In Vitro , Macrófagos/efeitos dos fármacos , Monócitos/citologia , Tioglicolatos/farmacologia , Ativador de Plasminogênio Tecidual/fisiologia
6.
Dev Biol ; 265(1): 140-54, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14697359

RESUMO

Epithelial-mesenchymal transformation (EMT), the process by which epithelial cells are converted into motile, invasive mesenchymal cells, is critical to valvulogenesis. Transforming growth factor-beta3 (TGF-beta3), an established mediator of avian atrioventricular (AV) canal EMT, is secreted as a latent complex. In vitro, plasmin-mediated proteolysis has been shown to release active TGF-betas from the latent complex. Annexin II, a co-receptor for tissue plasminogen activator (tPA) and plasminogen, promotes cell-surface generation of the serine protease plasmin. Here, we show that annexin II-mediated plasmin activity regulates release of active TGF-beta3 during chick AV canal EMT. Primary embryonic endocardial-derived cells express annexin II which promotes plasminogen activation in vitro. Incubation of heart explant cultures with either alpha(2)antiplasmin (alpha(2)AP), a major physiological plasmin inhibitor, or anti-annexin II IgG, blocked EMT by approximately 80%, and 50%, respectively. Anti-annexin II IgG-mediated inhibition of EMT was overcome by the addition of recombinant TGF-beta3. Upon treatment with anti-annexin II IgG or alpha(2)AP, conditioned medium from heart explant cultures showed absence of the active fragment of TGF-beta3 by Western blot analysis and a approximately 50% decrease in TGF-beta specific bioactivity. Our results suggest that annexin II-mediated plasmin activity regulates the release of active TGF-beta during cardiac valve development in the avian heart.


Assuntos
Anexina A2/metabolismo , Fibrinolisina/biossíntese , Valvas Cardíacas/embriologia , Mesoderma/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Bioensaio , Northern Blotting , Western Blotting , Embrião de Galinha , Epitélio/embriologia , Epitélio/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Fator de Crescimento Transformador beta3 , alfa 2-Antiplasmina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...