Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 41(1): 234-249, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33176450

RESUMO

OBJECTIVE: Platelet transfusion is a life-saving therapy to prevent or treat bleeding in patients with thrombocytopenia or platelet dysfunction. However, for >6 decades, safe and effective strategies for platelet storage have been an impediment to widespread use of platelet transfusion. Refrigerated platelets are cleared rapidly from circulation, precluding cold storage of platelets for transfusion. Consequently, platelets are stored at room temperature with an upper limit of 5 days due to risks of bacterial contamination and loss of platelet function. This practice severely limits platelet availability for transfusion. This study is to identify the mechanism of platelet clearance after cold storage and develop a method for platelet cold storage. Approach and Results: We found that rapid clearance of cold-stored platelets was largely due to integrin activation and apoptosis. Deficiency of integrin ß3 or caspase-3 prolonged cold-stored platelets in circulation. Pretreatment of platelets with EGTA, a cell impermeable calcium ion chelator, reversely inhibited cold storage-induced platelet activation and consequently prolonged circulation of cold-stored platelets. Moreover, transfusion of EGTA-treated, cold-stored platelets, but not room temperature-stored platelets, into the mice deficient in glycoprotein Ibα significantly shortened tail-bleeding times and diminished blood loss. CONCLUSIONS: Integrin activation and apoptosis is the underlying mechanism of rapid clearance of platelets after cold storage. Addition of a cell impermeable calcium ion chelator to platelet products is potentially a simple and effective method to enable cold storage of platelets for transfusion.


Assuntos
Plaquetas/efeitos dos fármacos , Preservação de Sangue , Quelantes de Cálcio/farmacologia , Cálcio/sangue , Temperatura Baixa , Ácido Egtázico/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Plaquetas/metabolismo , Feminino , Humanos , Integrinas/sangue , Integrinas/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transfusão de Plaquetas , Fatores de Tempo
2.
Immunity ; 50(6): 1401-1411.e4, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31076358

RESUMO

Inflammasome activation and subsequent pyroptosis are critical defense mechanisms against microbes. However, overactivation of inflammasome leads to death of the host. Although recent studies have uncovered the mechanism of pyroptosis following inflammasome activation, how pyroptotic cell death drives pathogenesis, eventually leading to death of the host, is unknown. Here, we identified inflammasome activation as a trigger for blood clotting through pyroptosis. We have shown that canonical inflammasome activation by the conserved type III secretion system (T3SS) rod proteins from Gram-negative bacteria or noncanonical inflammasome activation by lipopolysaccharide (LPS) induced systemic blood clotting and massive thrombosis in tissues. Following inflammasome activation, pyroptotic macrophages released tissue factor (TF), an essential initiator of coagulation cascades. Genetic or pharmacological inhibition of TF abolishes inflammasome-mediated blood clotting and protects against death. Our data reveal that blood clotting is the major cause of host death following inflammasome activation and demonstrate that inflammasome bridges inflammation with thrombosis.


Assuntos
Coagulação Sanguínea , Inflamassomos/metabolismo , Piroptose , Trombose/etiologia , Trombose/metabolismo , Animais , Infecções Bacterianas/complicações , Infecções Bacterianas/microbiologia , Biomarcadores , Caspases/metabolismo , Micropartículas Derivadas de Células/imunologia , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Humanos , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Transdução de Sinais , Tromboplastina/metabolismo , Trombose/sangue , Trombose/mortalidade
3.
Sci Rep ; 7(1): 17112, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29215030

RESUMO

CD40 ligand (CD40L), a member of the tumor necrosis factor (TNF) superfamily, binds to CD40, leading to many effects depending on target cell type. Platelets express CD40L and are a major source of soluble CD40L. CD40L has been shown to potentiate platelet activation and thrombus formation, involving both CD40-dependent and -independent mechanisms. A family of proteins called TNF receptor associated factors (TRAFs) plays key roles in mediating CD40L-CD40 signaling. Platelets express several TRAFs. It has been shown that TRAF2 plays a role in CD40L-mediated platelet activation. Here we show that platelet also express TRAF3, which plays a negative role in regulating platelet activation. Thrombin- or collagen-induced platelet aggregation and secretion are increased in TRAF3 knockout mice. The expression levels of collagen receptor GPVI and integrin αIIbß3 in platelets were not affected by deletion of TRAF3, suggesting that increased platelet activation in the TRAF3 knockout mice was not due to increased expression platelet receptors. Time to formation of thrombi in a FeCl3-induced thrombosis model was significantly shortened in the TRAF3 knockout mice. However, mouse tail-bleeding times were not affected by deletion of TRAF3. Thus, TRAF3 plays a negative role in platelet activation and in thrombus formation in vivo.


Assuntos
Agregação Plaquetária , Fator 3 Associado a Receptor de TNF/metabolismo , Trombose/metabolismo , Animais , Plaquetas/metabolismo , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Fator 3 Associado a Receptor de TNF/genética , Trombose/sangue , Trombose/genética
4.
Blood ; 127(11): 1459-67, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26738539

RESUMO

Platelet and megakaryocyte endocytosis is important for loading certain granule cargo (ie, fibrinogen [Fg] and vascular endothelial growth factor); however, the mechanisms of platelet endocytosis and its functional acute effects are understudied. Adenosine 5'-diphosphate-ribosylation factor 6 (Arf6) is a small guanosine triphosphate-binding protein that regulates endocytic trafficking, especially of integrins. To study platelet endocytosis, we generated platelet-specific Arf6 knockout (KO) mice. Arf6 KO platelets had less associated Fg suggesting that Arf6 affects αIIbß3-mediated Fg uptake and/or storage. Other cargo was unaffected. To measure Fg uptake, mice were injected with biotinylated- or fluorescein isothiocyanate (FITC)-labeled Fg. Platelets from the injected Arf6 KO mice showed lower accumulation of tagged Fg, suggesting an uptake defect. Ex vivo, Arf6 KO platelets were also defective in FITC-Fg uptake and storage. Immunofluorescence analysis showed initial trafficking of FITC-Fg to a Rab4-positive compartment followed by colocalization with Rab11-positive structures, suggesting that platelets contain and use both early and recycling endosomes. Resting and activated αIIbß3 levels, as measured by flow cytometry, were unchanged; yet, Arf6 KO platelets exhibited enhanced spreading on Fg and faster clot retraction. This was not the result of alterations in αIIbß3 signaling, because myosin light-chain phosphorylation and Rac1/RhoA activation were unaffected. Consistent with the enhanced clot retraction and spreading, Arf6 KO mice showed no deficits in tail bleeding or FeCl3-induced carotid injury assays. Our studies present the first mouse model for defining the functions of platelet endocytosis and suggest that altered integrin trafficking may affect the efficacy of platelet function.


Assuntos
Fatores de Ribosilação do ADP/fisiologia , Plaquetas/fisiologia , Endocitose/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/deficiência , Fatores de Ribosilação do ADP/genética , Animais , Biotinilação , Plaquetas/ultraestrutura , Membrana Celular/metabolismo , Tamanho Celular , Retração do Coágulo , Grânulos Citoplasmáticos , Fibrinogênio/metabolismo , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia
5.
Circulation ; 132(24): 2334-44, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26399659

RESUMO

BACKGROUND: Integrins are heterodimeric (α/ß) membrane proteins that play fundamental roles in many biological processes, for example, cell adhesion and spreading, which are important for platelet function and hemostasis. The molecular mechanism that regulates integrin activation is not completely understood. METHODS AND RESULTS: Here, we show that VPS33B, a member of the Sec1/Munc18 family, binds directly to the integrin ß subunit. Overexpression of VPS33B in Chinese hamster ovary cells potentiated αIIbß3 outside-in signaling but not inside-out signaling. Platelets, from megakaryocyte- and platelet-specific VPS33B conditional knockout mice, had normal morphology, yet their spreading on fibrinogen was impaired and they failed to support clot retraction. Platelet aggregation and ATP secretion in response to low-dose agonists were reduced in the VPS33B knockout mice. αIIbß3-mediated endocytosis of fibrinogen was also defective. Tail bleeding times and times to occlusion in an FeCl3-induced thrombosis model were prolonged in the VPS33B knockout mice. Furthermore, VPS33B acted upstream of the RhoA-ROCK-MLC and Rac1-dependent pathways that lead to clot retraction and cell spreading, respectively. CONCLUSIONS: Our work demonstrates that vesicular trafficking complexes, containing VPS33B, are a novel class of modifiers of integrin function. Our data also provide insights into the molecular mechanism and treatment of arthrogryposis, renal dysfunction, and cholestasis syndrome.


Assuntos
Hemostasia/fisiologia , Ativação Plaquetária/fisiologia , Trombose/metabolismo , Proteínas de Transporte Vesicular/biossíntese , Animais , Células CHO , Proteínas de Transporte/metabolismo , Cricetinae , Cricetulus , Integrinas/metabolismo , Camundongos , Camundongos Knockout , Transporte Proteico/fisiologia
6.
Blood ; 126(10): 1224-33, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26209658

RESUMO

Autophagy is important for maintaining cellular homeostasis, and thus its deficiency is implicated in a broad spectrum of human diseases. Its role in platelet function has only recently been examined. Our biochemical and imaging studies demonstrate that the core autophagy machinery exists in platelets, and that autophagy is constitutively active in resting platelets. Moreover, autophagy is induced upon platelet activation, as indicated by agonist-induced loss of the autophagy marker LC3II. Additional experiments, using inhibitors of platelet activation, proteases, and lysosomal acidification, as well as platelets from knockout mouse strains, show that agonist-induced LC3II loss is a consequence of platelet signaling cascades and requires proteases, acidic compartments, and membrane fusion. To assess the physiological role of platelet autophagy, we generated a mouse strain with a megakaryocyte- and platelet-specific deletion of Atg7, an enzyme required for LC3II production. Ex vivo analysis of platelets from these mice shows modest defects in aggregation and granule cargo packaging. Although these mice have normal platelet numbers and size distributions, they exhibit a robust bleeding diathesis in the tail-bleeding assay and a prolonged occlusion time in the FeCl3-induced carotid injury model. Our results demonstrate that autophagy occurs in platelets and is important for hemostasis and thrombosis.


Assuntos
Autofagia/fisiologia , Hemostasia/fisiologia , Ativação Plaquetária/fisiologia , Trombose/fisiopatologia , Animais , Plaquetas/fisiologia , Western Blotting , Células Cultivadas , Humanos , Camundongos , Camundongos Mutantes
7.
Nat Commun ; 4: 2657, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24150174

RESUMO

Although it has long been known that patients with sepsis often have thrombocytopenia and that septic patients with severe thrombocytopenia have a poor prognosis and higher mortality, the role of platelets in the pathogenesis of sepsis is poorly understood. Here we report a protective role of platelets in septic shock. We show that experimental thrombocytopenia induced by intraperitoneal injection of an anti-glycoprotein Ibα monoclonal antibody increases mortality and aggravates organ failure, whereas transfusion of platelets reduces mortality in lipopolysaccharide-induced endotoxemia and a bacterial infusion mouse sepsis model. Plasma concentrations of proinflammatory cytokines TNF-α and IL-6 are elevated by thrombocytopenia and decreased by platelet transfusion in septic mice. Furthermore, we identify that platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the COX1/PGE2/EP4-dependent pathway. Thus, these findings demonstrate a previously unappreciated role for platelets in septic shock and suggest that platelet transfusion may be effective in treating severely septic patients.


Assuntos
Plaquetas/imunologia , Macrófagos Peritoneais/imunologia , Transfusão de Plaquetas , Choque Séptico/terapia , Trombocitopenia/prevenção & controle , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Plaquetas/citologia , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/imunologia , Dinoprostona/genética , Dinoprostona/imunologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Inflamação/prevenção & controle , Interleucina-6/biossíntese , Interleucina-6/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/imunologia , Choque Séptico/induzido quimicamente , Choque Séptico/genética , Choque Séptico/patologia , Transdução de Sinais , Trombocitopenia/induzido quimicamente , Trombocitopenia/genética , Trombocitopenia/patologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
8.
PLoS One ; 7(12): e51037, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23236426

RESUMO

BACKGROUND: ADP is an important physiological agonist that induces integrin activation and platelet aggregation through its receptors P2Y(1) (Gα(q)-coupled) and P2Y(12) (Gα(i)-coupled). P2Y(12) plays a critical role in platelet activation and thrombosis. Adenosine-based P2Y(12) antagonists, 2-methylthioadenosine 5'-monophosphate triethylammonium salt hydrate (2MeSAMP) and Cangrelor (AR-C69931MX) have been widely used to demonstrate the role of P2Y(12) in platelet function. Cangrelor is being evaluated in clinical trials of thrombotic diseases. However, a recent study reported that both 2MeSAMP and Cangrelor raise intra-platelet cAMP levels and inhibit platelet aggregation through a P2Y(12)-independent mechanism. METHODOLOGY/PRINCIPAL FINDINGS: The present work, using P2Y(12) deficient mice, sought to clarify previous conflicting reports and to elucidate the mechanisms by which 2MeSAMP and Cangrelor inhibit platelet activation and thrombosis. 2MeSAMP and Cangrelor inhibited aggregation and ATP release of wild-type but not P2Y(12) deficient platelets. 2MeSAMP and Cangrelor neither raised intracellular cAMP concentrations nor induced phosphorylation of vasodilator-stimulated phosphoprotein (VASP) in washed human or mouse platelets. Furthermore, unlike the activators (PGI(2) and forskolin) of the cAMP pathway, 2MeSAMP and Cangrelor failed to inhibit Ca(2+) mobilization, Akt phosphorylation, and Rap1b activation in P2Y(12) deficient platelets. Importantly, while injection of Cangrelor inhibited thrombus formation in a FeCl(3)-induced thrombosis model in wild-type mice, it failed to affect thrombus formation in P2Y(12) deficient mice. CONCLUSIONS: These data together demonstrate that 2MeSAMP and Cangrelor inhibit platelet function through the P2Y(12)-dependent mechanism both in vitro and in vivo.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Adenosina/análogos & derivados , Plaquetas/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Tionucleosídeos/farmacologia , Adenosina/farmacologia , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/farmacologia , Animais , Plaquetas/metabolismo , Moléculas de Adesão Celular/metabolismo , Colforsina/farmacologia , AMP Cíclico/metabolismo , Epoprostenol/farmacologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Purinérgicos P2Y/genética , Receptores Purinérgicos P2Y/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
J Biol Chem ; 287(49): 41277-87, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23066026

RESUMO

The Src family kinases (SFKs) play essential roles in collagen- and von Willebrand factor (VWF)-mediated platelet activation. However, the roles of SFKs in G protein-coupled receptor-mediated platelet activation and the molecular mechanisms whereby SFKs are activated by G protein-coupled receptor stimulation are not fully understood. Here we show that the thrombin receptor protease-activated receptor 4 agonist peptide AYPGKF elicited SFK phosphorylation in P2Y(12) deficient platelets but stimulated minimal SFK phosphorylation in platelets lacking G(q). We have previously shown that thrombin-induced SFK phosphorylation was inhibited by the calcium chelator 5,5'-dimethyl-bis-(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (dimethyl-BAPTA). The calcium ionophore A23187 induced SFK phosphorylation in both wild-type and G(q) deficient platelets. Together, these results indicate that SFK phosphorylation in response to thrombin receptor stimulation is downstream from G(q)/Ca(2+) signaling. Moreover, A23187-induced thromboxane A(2) synthesis, platelet aggregation, and secretion were inhibited by preincubation of platelets with a selective SFK inhibitor, PP2. AYPGKF-induced thromboxane A(2) production in wild-type and P2Y(12) deficient platelets was abolished by PP2, and AYPGKF-mediated P-selectin expression, integrin α(IIb)ß(3) activation, and aggregation of P2Y(12) deficient platelets were partially inhibited by the PKC inhibitor Ro-31-8220, PP2, dimethyl-BAPTA, or LY294002, but were abolished by Ro-31-8220 plus PP2, dimethyl-BAPTA, or LY294002. These data indicate that Ca(2+)/SFKs/PI3K and PKC represent two alternative signaling pathways mediating G(q)-dependent platelet activation.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Regulação Enzimológica da Expressão Gênica , Oligopeptídeos/metabolismo , Proteína Quinase C/metabolismo , Quinases da Família src/metabolismo , Animais , Plaquetas/metabolismo , Antígenos CD18/metabolismo , Calcimicina/farmacologia , Cálcio/metabolismo , Inibidores Enzimáticos/farmacologia , Fibrinogênio/metabolismo , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ativação Plaquetária , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Ligação Proteica , Receptores Purinérgicos P2Y12/metabolismo
10.
J Biol Chem ; 286(45): 39466-77, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21940635

RESUMO

Rap1b is activated by platelet agonists and plays a critical role in integrin α(IIb)ß(3) inside-out signaling and platelet aggregation. Here we show that agonist-induced Rap1b activation plays an important role in stimulating secretion of platelet granules. We also show that α(IIb)ß(3) outside-in signaling can activate Rap1b, and integrin outside-in signaling-mediated Rap1b activation is important in facilitating platelet spreading on fibrinogen and clot retraction. Rap1b-deficient platelets had diminished ATP secretion and P-selectin expression induced by thrombin or collagen. Importantly, addition of low doses of ADP and/or fibrinogen restored aggregation of Rap1b-deficient platelets. Furthermore, we found that Rap1b was activated by platelet spreading on immobilized fibrinogen, a process that was not affected by P2Y(12) or TXA(2) receptor deficiency, but was inhibited by the selective Src inhibitor PP2, the PKC inhibitor Ro-31-8220, or the calcium chelator demethyl-1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis. Clot retraction was abolished, and platelet spreading on fibrinogen was diminished in Rap1b-deficient platelets compared with wild-type controls. The defects in clot retraction and spreading on fibrinogen of Rap1b-deficient platelets were not rescued by addition of MnCl(2), which elicits α(IIb)ß(3) outside-in signaling in the absence of inside-out signaling. Thus, our results reveal two different activation mechanisms of Rap1b as well as novel functions of Rap1b in platelet secretion and in integrin α(IIb)ß(3) outside-in signaling.


Assuntos
Plaquetas/metabolismo , Retração do Coágulo/fisiologia , Adesividade Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Proteínas rap de Ligação ao GTP/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Animais , Quelantes/farmacologia , Retração do Coágulo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , Fibrinogênio/metabolismo , Indóis/farmacologia , Camundongos , Camundongos Mutantes , Selectina-P/genética , Selectina-P/metabolismo , Adesividade Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Pirimidinas/farmacologia , Receptores Purinérgicos P2Y12/genética , Receptores Purinérgicos P2Y12/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas rap de Ligação ao GTP/genética
11.
Blood ; 118(13): 3670-9, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21803853

RESUMO

Nitric oxide (NO) stimulates cGMP synthesis by activating its intracellular receptor, soluble guanylyl cyclase (sGC). It is a currently prevailing concept that No and cGMP inhibits platelet function. However, the data supporting the inhibitory role of NO/sGC/cGMP in platelets have been obtained either in vitro or using whole body gene deletion that affects vessel wall function. Here we have generated mice with sGC gene deleted only in megakaryocytes and platelets. Using the megakaryocyte- and platelet-specific sGC-deficient mice, we identify a stimulatory role of sGC in platelet activation and in thrombosis in vivo. Deletion of sGC in platelets abolished cGMP production induced by either NO donors or platelet agonists, caused a marked defect in aggregation and attenuated secretion in response to low doses of collagen or thrombin. Importantly, megakaryocyte- and platelet-specific sGC deficient mice showed prolonged tail-bleeding times and impaired FeCl3-induced carotid artery thrombosis in vivo. Interestingly, the inhibitory effect of the NO donor SNP on platelet activation was sGC-dependent only at micromolar concentrations, but sGC-independent at millimolar concentrations. Together, our data demonstrate important roles of sGC in stimulating platelet activation and in vivo thrombosis and hemostasis, and sGC-dependent and -independent inhibition of platelets by NO donors.


Assuntos
Guanilato Ciclase/fisiologia , Ativação Plaquetária/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Plaquetas/fisiologia , Cálcio/metabolismo , GMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/farmacologia , Proteína Oncogênica v-akt/metabolismo , Especificidade de Órgãos/genética , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Guanilil Ciclase Solúvel
12.
EMBO J ; 29(22): 3773-86, 2010 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-20924358

RESUMO

The molecular chaperone heat shock protein 90 (Hsp90) and the co-chaperone/ubiquitin ligase carboxyl terminus of Hsc70-interacting protein (CHIP) control the turnover of client proteins. How this system decides to stabilize or degrade the client proteins under particular physiological or pathological conditions is unclear. We report here a novel client protein, the SUMO2/3 protease SENP3, that is sophisticatedly regulated by CHIP and Hsp90. SENP3 is maintained at a low basal level under non-stress condition due to Hsp90-independent CHIP-mediated ubiquitination. Upon mild oxidative stress, SENP3 undergoes thiol modification, which recruits Hsp90. Hsp90/SENP3 association protects SENP3 from CHIP-mediated ubiquitination and subsequent degradation, but this effect of Hsp90 requires the presence of CHIP. Our data demonstrate for the first time that CHIP and Hsp90 interplay with a client alternately under non-stress and stress conditions, and the choice between stabilization and degradation is made by the redox state of the client. In addition, enhanced SENP3/Hsp90 association is found in cancer. These findings provide new mechanistic insight into how cells regulate the SUMO protease in response to oxidative stress.


Assuntos
Cisteína Endopeptidases/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Cisteína/metabolismo , Estabilidade Enzimática , Células HEK293 , Células HeLa , Humanos , Oxirredução , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Ubiquitinação
13.
J Biol Chem ; 285(17): 12906-15, 2010 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-20181954

RESUMO

Small ubiquitin-like modifier (SUMO) 2/3 is known to conjugate to substrates in response to a variety of cellular stresses. However, whether and how SUMO2/3-specific proteases are involved in de-conjugation under cell stress is unclear. Here, we show that low doses of hydrogen peroxide (H(2)O(2)) induce an increase of the SENP3 protein, which removes SUMO2/3 from promyelocytic leukemia (PML). Low dose H(2)O(2) causes SENP3 to co-localize with PML bodies and reduces the number of PML bodies in a SENP3-dependent manner. Furthermore, de-conjugation of SUMO2/3 from PML is responsible for the accelerated cell proliferation caused by low dose H(2)O(2). Knocking down PML promotes basal cell proliferation as expected. This can be reversed by reconstitution with wild-type PML but not its mutant lacking SUMOylation, indicating that only the SUMOylated PML can play an inhibitory role for cell proliferation. Thus, SENP3 appears to be a key mediator in mild oxidative stress-induced cell proliferation via regulation of the SUMOylation status of PML. Furthermore, SENP3 is over-accumulated in a variety of primary human cancers including colon adenocarcinoma in which PML is hypo-SUMOylated. These results reveal an important role of SENP3 and the SUMOylation status of PML in the regulation of cell proliferation under oxidative stress.


Assuntos
Proliferação de Células , Cisteína Endopeptidases/metabolismo , Proteínas Nucleares/metabolismo , Estresse Oxidativo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animais , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Cisteína Endopeptidases/genética , Relação Dose-Resposta a Droga , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Peróxido de Hidrogênio/farmacologia , Camundongos , Células NIH 3T3 , Proteínas Nucleares/genética , Oxidantes/farmacologia , Proteína da Leucemia Promielocítica , Processamento de Proteína Pós-Traducional , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Ubiquitinas/genética
14.
Cell Res ; 19(4): 449-57, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19238172

RESUMO

Intracellular reactive oxygen species (ROS) are known to regulate apoptosis. Activation of caspase-9, the initial caspase in the mitochondrial apoptotic cascade, is closely associated with ROS, but it is unclear whether ROS regulate caspase-9 via direct oxidative modification. The present study aims to elucidate the molecular mechanisms by which ROS mediate caspase-9 activation. Our results show that the cellular oxidative state facilitates caspase-9 activation. Hydrogen peroxide treatment causes the activation of caspase-9 and apoptosis, and promotes an interaction between caspase-9 and apoptotic protease-activating factor 1 (Apaf-1) via disulfide formation. In addition, in an in vitro mitochondria-free system, the thiol-oxidant diamide promotes auto-cleavage of caspase-9 and the caspase-9/Apaf-1 interaction by facilitating the formation of disulfide-linked complexes. Finally, a point mutation at C403 of caspase-9 impairs both H(2)O(2)-promoted caspase-9 activation and interaction with Apaf-1 through the abolition of disulfide formation. The association between cytochrome c and the C403S mutant is significantly weaker than that between cytochrome c and wild-type caspase-9, indicating that oxidative modification of caspase-9 contributes to apoptosome formation under oxidative stress. Taken together, oxidative modification of caspase-9 by ROS can mediate its interaction with Apaf-1, and can thus promote its auto-cleavage and activation. This mechanism may facilitate apoptosome formation and caspase-9 activation under oxidative stress.


Assuntos
Fator Apoptótico 1 Ativador de Proteases/metabolismo , Caspase 9/metabolismo , Dissulfetos/metabolismo , Estresse Oxidativo , Substituição de Aminoácidos , Apoptose , Linhagem Celular , Citocromos c/metabolismo , Diamida/farmacologia , Células HeLa , Humanos , Peróxido de Hidrogênio/farmacologia , Proteínas Mutantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA