Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Mol Med ; 25(18): 8789-8795, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34363435

RESUMO

Osteosarcoma (OS) is a sarcoma with high rates of pulmonary metastases and mortality. The mechanisms underlying tumour generation and development in OS are not well-understood. Haematopoietic cell kinase (HCK), a vital member of the Src family of kinase proteins, plays crucial roles in cancer progression and may act as an anticancer target; however, the mechanism by which HCK enhances OS development remains unexplored. Therefore, we investigated the role of HCK in OS development in vitro and in vivo. Downregulation of HCK attenuated OS cell proliferation, migration and invasion and increased OS cell apoptosis, whereas overexpression of HCK enhanced these processes. Mechanistically, HCK expression enhanced OS tumorigenesis via the mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway; HCK upregulation increased the phosphorylation of MEK and ERK and promoted epithelial-mesenchymal transition, with a reduction in E-cadherin in vitro. Furthermore, HCK downregulation decreased the tumour volume and weight in mice transplanted with OS cells. In conclusion, HCK plays a crucial role in OS tumorigenesis, progression and metastasis via the MEK/ERK pathway, suggesting that HCK is a potential target for developing treatments for OS.


Assuntos
Neoplasias Ósseas/metabolismo , Osteossarcoma/metabolismo , Proteínas Proto-Oncogênicas c-hck/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
2.
J Am Soc Nephrol ; 28(5): 1385-1393, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27927780

RESUMO

Renal fibrosis is the common pathway of progression for patients with CKD and chronic renal allograft injury (CAI), but the underlying mechanisms remain obscure. We performed a meta-analysis in human kidney biopsy specimens with CAI, incorporating data available publicly and from our Genomics of Chronic Renal Allograft Rejection study. We identified an Src family tyrosine kinase, hematopoietic cell kinase (Hck), as upregulated in allografts in CAI. Querying the Kinase Inhibitor Resource database revealed that dasatinib, a Food and Drug Administration-approved drug, potently binds Hck with high selectivity. In vitro, Hck overexpression activated the TGF-ß/Smad3 pathway, whereas HCK knockdown inhibited it. Treatment of tubular cells with dasatinib reduced the expression of Col1a1 Dasatinib also reduced proliferation and α-SMA expression in fibroblasts. In a murine model with unilateral ureteric obstruction, pretreatment with dasatinib significantly reduced the upregulation of profibrotic markers, phosphorylation of Smad3, and renal fibrosis observed in kidneys pretreated with vehicle alone. Dasatinib treatment also improved renal function, reduced albuminuria, and inhibited expression of profibrotic markers in animal models with lupus nephritis and folic acid nephropathy. These data suggest that Hck is a key mediator of renal fibrosis and dasatinib could be developed as an antifibrotic drug.


Assuntos
Nefropatias/genética , Transplante de Rim , Rim/patologia , Complicações Pós-Operatórias/genética , Proteínas Proto-Oncogênicas c-hck/genética , Proteínas Proto-Oncogênicas c-hck/fisiologia , Animais , Feminino , Fibrose/genética , Genômica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Circulation ; 132(6): 490-501, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26068045

RESUMO

BACKGROUND: Leukocyte migration is critical for the infiltration of monocytes and accumulation of monocyte-derived macrophages in inflammation. Considering that Hck and Fgr are instrumental in this process, their impact on atherosclerosis and on lesion inflammation and stability was evaluated. METHODS AND RESULTS: Hematopoietic Hck/Fgr-deficient, LDLr(-/-) chimeras, obtained by bone marrow transplantation, had smaller but, paradoxically, less stable lesions with reduced macrophage content, overt cap thinning, and necrotic core expansion as the most prominent features. Despite a Ly6C(high)-skewed proinflammatory monocyte phenotype, Hck/Fgr deficiency led to disrupted adhesion of myeloid cells to and transmigration across endothelial monolayers in vitro and atherosclerotic plaques in vivo, as assessed by intravital microscopy, flow cytometry, and histological examination of atherosclerotic arteries. Moreover, Hck/Fgr-deficient macrophages showed blunted podosome formation and mesenchymal migration capacity. In consequence, transmigrated double-knockout macrophages were seen to accumulate in the fibrous cap, potentially promoting its focal erosion, as observed for double-knockout chimeras. CONCLUSIONS: The hematopoietic deficiency of Hck and Fgr led to attenuated atherosclerotic plaque formation by abrogating endothelial adhesion and transmigration; paradoxically, it also promoted plaque instability by causing monocyte subset imbalance and subendothelial accumulation, raising a note of caution regarding src kinase-targeted intervention in plaque inflammation.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Macrófagos Peritoneais/patologia , Monócitos/patologia , Placa Aterosclerótica/patologia , Proteínas Proto-Oncogênicas c-hck/deficiência , Proteínas Proto-Oncogênicas/deficiência , Quinases da Família src/deficiência , Animais , Apoptose , Adesão Celular , Extensões da Superfície Celular/ultraestrutura , Células Cultivadas , Células Endoteliais , Proteínas da Matriz Extracelular/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Migração e Rolagem de Leucócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Placa Aterosclerótica/enzimologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-hck/genética , Proteínas Proto-Oncogênicas c-hck/fisiologia , Quimera por Radiação , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de LDL/fisiologia , Migração Transendotelial e Transepitelial , Quinases da Família src/genética , Quinases da Família src/fisiologia
4.
Blood ; 125(10): 1611-22, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25527710

RESUMO

Macrophages are motile leukocytes, targeted by HIV-1, thought to play a critical role in host dissemination of the virus. However, whether infection impacts their migration capacity remains unknown. We show that 2-dimensional migration and the 3-dimensional (3D) amoeboid migration mode of HIV-1-infected human monocyte-derived macrophages were inhibited, whereas the 3D mesenchymal migration was enhanced. The viral protein Nef was necessary and sufficient for all HIV-1-mediated effects on migration. In Nef transgenic mice, tissue infiltration of macrophages was increased in a tumor model and in several tissues at steady state, suggesting a dominant role for mesenchymal migration in vivo. The mesenchymal motility involves matrix proteolysis and podosomes, cell structures constitutive of monocyte-derived cells. Focusing on the mechanisms used by HIV-1 Nef to control the mesenchymal migration, we show that the stability, size, and proteolytic function of podosomes are increased via the phagocyte-specific kinase Hck and Wiskott-Aldrich syndrome protein (WASP), 2 major regulators of podosomes. In conclusion, HIV-1 reprograms macrophage migration, which likely explains macrophage accumulation in several patient tissues, which is a key step for virus spreading and pathogenesis. Moreover, Nef points out podosomes and the Hck/WASP signaling pathway as good candidates to control tissue infiltration of macrophages, a detrimental phenomenon in several diseases.


Assuntos
HIV-1/patogenicidade , Macrófagos/fisiologia , Macrófagos/virologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Animais , Linhagem Celular Tumoral , Estruturas da Membrana Celular/patologia , Estruturas da Membrana Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Reprogramação Celular/fisiologia , Infecções por HIV/patologia , Infecções por HIV/fisiopatologia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-hck/fisiologia , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
5.
J Cell Physiol ; 227(3): 1090-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21567396

RESUMO

The interaction between HIV-1 Nef and the Src kinase Hck in macrophages has been shown to accelerate the progression to AIDS. We previously showed that Nef disturbed the N-glycosylation/trafficking of Fms, a cytokine receptor essential for maintaining macrophages in an anti-inflammatory state, in an Hck-dependent manner. Here, we show the underlying molecular mechanism of this effect. Using various Hck isoforms and their mutants and Golgi-targeting Hck mutants, we confirmed that Hck activation at the Golgi causes the Nef-induced Fms N-glycosylation defect. Importantly, we found that both the co-expression of Nef and Hck and the expression of a Golgi-targeted active Hck mutant caused alterations in the distribution of GM130, a Golgi protein that was shown to be required for efficient protein glycosylation. Moreover, the activation of Hck at the Golgi caused strong serine phosphorylation of the GM130-interacting Golgi structural protein GRASP65, which is known to induce Golgi cisternal unstacking. Using pharmacological inhibitors, we also found that the activation of Hck at the Golgi followed by the activation of the MAP kinase ERK-GRASP65 cascade is involved in the Fms N-glycosylation defect. These results suggest that Nef perturbs the structure and signaling of the Golgi by activating Hck at the Golgi, and thereby, induces the N-glycosylation/trafficking defect of Fms, which is in line with the idea that Src family kinases are crucial Golgi regulators.


Assuntos
Complexo de Golgi/patologia , Complexo de Golgi/virologia , Infecções por HIV/metabolismo , HIV-1/patogenicidade , Proteínas Proto-Oncogênicas c-hck/fisiologia , Transdução de Sinais/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Progressão da Doença , Complexo de Golgi/enzimologia , Células HEK293 , Infecções por HIV/patologia , Infecções por HIV/virologia , Humanos , Transporte Proteico/fisiologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo
6.
J Immunol ; 186(4): 2372-81, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21220696

RESUMO

We have recently reported that mice deficient in the myeloid Src-family tyrosine kinases Hck, Fgr, and Lyn (Src triple knockout [TKO]) had augmented innate lung clearance of Pneumocystis murina that correlated with a higher ability of alveolar macrophages (AMs) from these mice to kill P. murina. In this article, we show that despite possessing enhanced killing, AMs from naive Src TKO mice did not demonstrate enhanced inflammatory responses to P. murina. We subsequently discovered that both AMs and lungs from P. murina-infected Src TKO mice expressed significantly greater levels of the M2a markers RELM-α and Arg1, and the M2a-associated chemokines CCL17 and CCL22 than did wild-type mice. IL-4 and IL-13, the primary cytokines that promote M2a polarization, were not differentially produced in the lungs between wild-type and Src TKO mice. P. murina infection in Src TKO mice resulted in enhanced lung production of the novel IL-1 family cytokine IL-33. Immunohistochemical analysis of IL-33 in lung tissue revealed localization predominantly in the nucleus of alveolar epithelial cells. We further demonstrate that experimental polarization of naive AMs to M2a resulted in more efficient killing of P. murina compared with untreated AMs, which was further enhanced by the addition of IL-33. Administration of IL-33 to C57BL/6 mice increased lung RELM-α and CCL17 levels, and enhanced clearance of P. murina, despite having no effect on the cellular composition of the lungs. Collectively, these results indicate that M2a AMs are potent effector cells against P. murina. Furthermore, enhancing M2a polarization may be an adjunctive therapy for the treatment of Pneumocystis.


Assuntos
Interleucinas/fisiologia , Macrófagos Alveolares/imunologia , Pneumocystis/imunologia , Pneumocystis/patogenicidade , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/prevenção & controle , Animais , Polaridade Celular/genética , Polaridade Celular/imunologia , Células Cultivadas , Imunidade Inata/genética , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Interleucina-33 , Interleucinas/biossíntese , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos Alveolares/enzimologia , Macrófagos Alveolares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/genética , Fagocitose/imunologia , Pneumocystis/crescimento & desenvolvimento , Pneumonia por Pneumocystis/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-hck/deficiência , Proteínas Proto-Oncogênicas c-hck/fisiologia , Quinases da Família src/deficiência , Quinases da Família src/fisiologia
7.
Blood ; 116(3): 485-94, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20299514

RESUMO

In inflamed venules, neutrophils rolling on E-selectin induce integrin alpha(L)beta(2)-dependent slow rolling on intercellular adhesion molecule-1 by activating Src family kinases (SFKs), DAP12 and Fc receptor-gamma (FcRgamma), spleen tyrosine kinase (Syk), and p38. E-selectin signaling cooperates with chemokine signaling to recruit neutrophils into tissues. Previous studies identified P-selectin glycoprotein ligand-1 (PSGL-1) as the essential E-selectin ligand and Fgr as the only SFK that initiate signaling to slow rolling. In contrast, we found that E-selectin engagement of PSGL-1 or CD44 triggered slow rolling through a common, lipid raft-dependent pathway that used the SFKs Hck and Lyn as well as Fgr. We identified the Tec kinase Bruton tyrosine kinase as a key signaling intermediate between Syk and p38. E-selectin engagement of PSGL-1 was dependent on its cytoplasmic domain to activate SFKs and slow rolling. Although recruiting phosphoinositide-3-kinase to the PSGL-1 cytoplasmic domain was reported to activate integrins, E-selectin-mediated slow rolling did not require phosphoinositide-3-kinase. Studies in mice confirmed the physiologic significance of these events for neutrophil slow rolling and recruitment during inflammation. Thus, E-selectin triggers common signals through distinct neutrophil glycoproteins to induce alpha(L)beta(2)-dependent slow rolling.


Assuntos
Selectina E/fisiologia , Receptores de Hialuronatos/fisiologia , Migração e Rolagem de Leucócitos/fisiologia , Antígeno-1 Associado à Função Linfocitária/fisiologia , Glicoproteínas de Membrana/fisiologia , Tirosina Quinase da Agamaglobulinemia , Animais , Humanos , Receptores de Hialuronatos/genética , Técnicas In Vitro , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Microdomínios da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Neutrófilos/fisiologia , Selectina-P/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-hck/fisiologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Quinases da Família src/fisiologia
8.
Eur J Cancer ; 45(3): 321-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19114024

RESUMO

Hck is a phagocyte specific proto-oncogene of the Src family expressed as two isoforms, p59Hck and p61Hck. It plays a critical role in Bcr/Abl-chronic myeloid leukaemia and is able to transform fibroblasts in vitro. However, the tumourigenic activity of Hck and the respective oncogenic functions of Hck isoforms have not been examined. Tet-Off fibroblasts expressing constitutively active mutants of p59Hck and p61Hck together or individually were used. In contrast to cells expressing p59Hck(ca) or p61Hck(ca) alone, cells expressing both isoforms were transformed in vitro and induced tumour formation in 90% of nude mice within 2 weeks. This is the first demonstration of (i) the tumourigenic activity of Hck in mice, (ii) the cooperative action of the two Hck isoforms in vitro and in vivo. To our knowledge, this is the first example of a transforming activity 'split' in two requisite isoforms.


Assuntos
Membrana Celular/metabolismo , Transformação Celular Neoplásica , Fibroblastos/metabolismo , Mutagênese/genética , Proteínas Proto-Oncogênicas c-hck/fisiologia , Animais , Membrana Celular/genética , Proliferação de Células , Células Cultivadas , Humanos , Lisossomos/genética , Masculino , Camundongos , Camundongos Nus , Isoformas de Proteínas/fisiologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-hck/metabolismo , Transdução de Sinais/genética
9.
J Leukoc Biol ; 84(4): 1082-91, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18632989

RESUMO

Chemokines play pivotal roles in regulating a wide variety of biological processes by modulating cell migration and recruitment. Deregulation of chemokine signaling can alter cell recruitment, contributing to the pathogenic states associated with autoimmune disease, inflammatory disorders, and sepsis. During chemotaxis, lipid rafts and their resident signaling molecules have been demonstrated to partition to different parts of the cell. Herein, we investigated the role of lipid raft resident Src-family kinases (SFK) in stromal cell-derived factor 1/CXCL12-mediated chemotaxis. We have shown that Lck-deficient J.CaM 1.6 cells are defective in CXCL12-mediated chemotaxis in contrast to their parental counterpart, Jurkat cells. Ectopic expression of the SFK hematopoietic cell kinase (Hck) in J.CaM 1.6 cells reconstituted CXCL12 responsiveness. The requirement of lipid raft association of SFK was assessed using both isoforms of Hck: the dually acylated p59(Hck) isoform that is targeted to lipid rafts and the monoacylated p61(Hck) isoform that is nonraft-associated. We have shown using several gain and loss of acylation alleles that dual acylation of Hck was required for CXCL12-mediated chemotaxis in J.CaM 1.6 cells. These results highlight the importance of the unique microenvironment provided by lipid rafts and their specific contribution in providing specificity to CXCL12 signaling.


Assuntos
Quimiocina CXCL12/fisiologia , Quimiotaxia/fisiologia , Células Jurkat/fisiologia , Quinases da Família src/metabolismo , Acilação , Citometria de Fluxo , Humanos , Indóis/farmacologia , Microdomínios da Membrana/fisiologia , Plasmídeos , Proteínas Proto-Oncogênicas c-hck/genética , Proteínas Proto-Oncogênicas c-hck/fisiologia , Sulfonamidas/farmacologia , Quinases da Família src/antagonistas & inibidores
10.
J Clin Invest ; 118(3): 924-34, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18246197

RESUMO

The proliferation and differentiation of hematopoietic stem cells (HSCs) is finely regulated by extrinsic and intrinsic factors via various signaling pathways. Here we have shown that, similar to mice deficient in the lipid phosphatase SHIP, loss of 2 Src family kinases, Lyn and Hck, profoundly affects HSC differentiation, producing hematopoietic progenitors with increased proliferation, reduced apoptosis, growth factor-independent survival, and skewed differentiation toward M2 macrophages. This phenotype culminates in a Stat5-dependent myeloproliferative disease that is accompanied by M2 macrophage infiltration of the lung. Expression of a membrane-bound form of SHIP in HSCs lacking both Lyn and Hck restored normal hematopoiesis and prevented myeloproliferation. In vitro and in vivo studies suggested the involvement of autocrine and/or paracrine production of IL-3 and GM-CSF in the increased proliferation and myeloid differentiation of HSCs. Thus, this study has defined a myeloproliferative transformation-sensitive signaling pathway, composed of Lyn/Hck, SHIP, autocrine/paracrine cytokines, and Stat5, that regulates HSC differentiation and M2 macrophage programming.


Assuntos
Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Proto-Oncogênicas c-hck/fisiologia , Fator de Transcrição STAT5/fisiologia , Quinases da Família src/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Inositol Polifosfato 5-Fosfatases , Interleucina-3/fisiologia , Pulmão/patologia , Pneumopatias/etiologia , Camundongos , Camundongos Knockout , Transtornos Mieloproliferativos/etiologia , Transdução de Sinais
11.
Oncogene ; 27(27): 3831-44, 2008 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-18246120

RESUMO

Bcr-Abl oncogene is responsible for the initial phase of chronic myelogenous leukemia (CML), which is effectively treated by the Bcr-Abl inhibitor imatinib. Over time patients become resistant to treatment and progress to blast crisis, an event that is driven by additional genetic and epigenetic aberrations. Recently, we showed that Riz1 expression decreases in blast crisis and that re-expression of Riz1 inhibits IGF-1 expression. IGF-1 signaling is required in many stages of hematopoiesis and inappropriate activation of autocrine IGF-1 signaling may facilitate transformation to blast crisis. We observed that in 8 out of 11 matched CML patient biopsies the IGF-1 expression is elevated in blast crisis. We examined mechanisms used by CML blast crisis cell lines to activate IGF-1 expression. We found that Bcr-Abl activates autocrine IGF-1 signaling using Hck and Stat5b. Inhibition of these signaling components using small molecule drugs or shRNA decreases proliferation and enhances apoptosis. Together, our study suggests that aberrant IGF-1 signaling is an important event in blast crisis transformation and it provides a mechanism to explain the activity of IGF-1R and Hck inhibitors in blocking CML blast crisis phenotypes.


Assuntos
Proteínas de Fusão bcr-abl/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Antineoplásicos/uso terapêutico , Benzamidas , Crise Blástica , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Humanos , Mesilato de Imatinib , Fator de Crescimento Insulin-Like I/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/fisiopatologia , Piperazinas/uso terapêutico , Proteínas Proto-Oncogênicas c-hck/fisiologia , Pirimidinas/uso terapêutico , RNA Mensageiro/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
12.
Curr Drug Targets ; 7(12): 1583-93, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17168833

RESUMO

Vif is an HIV accessory protein whose primary function is to negate the action of APOBEC3G, a naturally occurring cellular inhibitor of HIV replication. Vif acts by binding to APOBEC3G, inducing its protein degradation within infected cells and reducing its levels in progeny virions. Interventions that interfere with the Vif-APOBEC3G interaction, raise intracellular or virion associated levels of APOBEC3G, or reduce intracellular levels of Vif, all could hold promise as potential therapeutic approaches aimed at enhancing the cells innate antiviral activity. Levels of APOBEC3G might be increased or Vif levels decreased, by strategies targeting protein synthesis, protein degradation or cellular localisation and function, and properties of APOBEC3G and Vif relevant to these strategies are discussed. Recent data have suggested that Vif may have other mechanisms of action apart from the above activities against APOBEC3G, including effects against other anti-viral mechanisms independent of APOBEC3G cytidine deaminase activity. In addition to interaction with APOBEC3G, Vif may have other accessory functions, which are discussed in relation to potential therapies that may affect multiple stages of the HIV life cycle. Future development of strategies that combine enhancement of APBOEC3G functional with inhibition of multiple Vif functions may become useful tools for HIV therapy.


Assuntos
Fármacos Anti-HIV/farmacologia , Produtos do Gene vif/antagonistas & inibidores , Nucleosídeo Desaminases/fisiologia , Proteínas Repressoras/fisiologia , Desaminase APOBEC-3G , Acetiltransferases/fisiologia , Citidina Desaminase , Farmacorresistência Viral , Produtos do Gene vif/metabolismo , Protease de HIV/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Fenótipo , Ligação Proteica , Proteínas Proto-Oncogênicas c-hck/fisiologia , Ubiquitina-Proteína Ligases , Montagem de Vírus , Replicação Viral
13.
J Biol Chem ; 281(37): 27029-38, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849330

RESUMO

Nef is an HIV-1 virulence factor that promotes viral pathogenicity by altering host cell signaling pathways. Nef binds several members of the Src kinase family, and these interactions have been implicated in the pathogenesis of HIV/AIDS. However, the direct effect of Nef interaction on Src family kinase (SFK) regulation and activity has not been systematically addressed. We explored this issue using Saccharomyces cerevisiae, a well defined model system for the study of SFK regulation. Previous studies have shown that ectopic expression of c-Src arrests yeast cell growth in a kinase-dependent manner. We expressed Fgr, Fyn, Hck, Lck, Lyn, and Yes as well as c-Src in yeast and found that each kinase was active and induced growth suppression. Co-expression of the negative regulatory kinase Csk suppressed SFK activity and reversed the growth-inhibitory effect. We then co-expressed each SFK with HIV-1 Nef in the presence of Csk. Nef strongly activated Hck, Lyn, and c-Src but did not detectably affect Fgr, Fyn, Lck, or Yes. Mutagenesis of the Nef PXXP motif essential for SH3 domain binding greatly reduced the effect of Nef on Hck, Lyn, and c-Src, suggesting that Nef activates these Src family members through allosteric displacement of intramolecular SH3-linker interactions. These data show that Nef selectively activates Hck, Lyn, and c-Src among SFKs, identifying these kinases as proximal effectors of Nef signaling and potential targets for anti-HIV drug discovery.


Assuntos
Produtos do Gene nef/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-hck/fisiologia , Quinases da Família src/fisiologia , Sítio Alostérico , Animais , Proteína Tirosina Quinase CSK , Produtos do Gene nef/química , Humanos , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiologia , Domínios de Homologia de src
14.
J Immunol ; 177(1): 604-11, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16785558

RESUMO

Neutrophil beta(2) integrins are activated by inside-out signaling regulating integrin affinity and valency; following ligand binding, beta(2) integrins trigger outside-in signals regulating cell functions. Addressing inside-out and outside-in signaling in hck(-/-)fgr(-/-) neutrophils, we found that Hck and Fgr do not regulate chemoattractant-induced activation of beta(2) integrin affinity. In fact, beta(2) integrin-mediated rapid adhesion, in static condition assays, and neutrophil adhesion to glass capillary tubes cocoated with ICAM-1, P-selectin, and a chemoattractant, under flow, were unaffected in hck(-/-)fgr(-/-) neutrophils. Additionally, examination of integrin affinity by soluble ICAM-1 binding assays and of beta(2) integrin clustering on the cell surface, showed that integrin activation did not require Hck and Fgr expression. However, after binding, hck(-/-)fgr(-/-) neutrophil spreading over beta(2) integrin ligands was reduced and they rapidly detached from the adhesive surface. Whether alterations in outside-in signaling affect sustained adhesion to the vascular endothelium in vivo was addressed by examining neutrophil adhesiveness to inflamed muscle venules. Intravital microscopy analysis allowed us to conclude that Hck and Fgr regulate neither the number of rolling cells nor rolling velocity in neutrophils. However, arrest of hck(-/-)fgr(-/-) neutrophils to >60 microm in diameter venules was reduced. Thus, Hck and Fgr play no role in chemoattractant-induced inside-out beta(2) integrin activation but regulate outside-in signaling-dependent sustained adhesion.


Assuntos
Antígenos CD18/fisiologia , Neutrófilos/enzimologia , Neutrófilos/imunologia , Proteínas Proto-Oncogênicas c-hck/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais/imunologia , Quinases da Família src/fisiologia , Animais , Antígenos CD18/metabolismo , Adesão Celular/genética , Adesão Celular/imunologia , Inibição de Migração Celular , Células Cultivadas , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/patologia , N-Formilmetionina Leucil-Fenilalanina/imunologia , Ativação de Neutrófilo/genética , Ativação de Neutrófilo/imunologia , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-hck/deficiência , Proteínas Proto-Oncogênicas c-hck/genética , Transdução de Sinais/genética , Vênulas/imunologia , Vênulas/patologia , Quinases da Família src/deficiência , Quinases da Família src/genética
15.
Protein Sci ; 15(1): 65-73, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16322569

RESUMO

The ability of proteins to regulate their own enzymatic activity can be facilitated by changes in structure or protein dynamics in response to external regulators. Because many proteins contain SH2 and SH3 domains, transmission of information between the domains is a potential method of allosteric regulation. To determine if ligand binding to one modular domain may alter structural dynamics in an adjacent domain, allowing potential transmission of information through the protein, we used hydrogen exchange and mass spectrometry to measure changes in protein dynamics in the SH3 and SH2 domains of hematopoietic cell kinase (Hck). Ligand binding to either domain had little or no effect on hydrogen exchange in the adjacent domain, suggesting that changes in protein structure or dynamics are not a means of SH2/SH3 crosstalk. Furthermore, ligands of varying affinity covalently attached to SH3/SH2 altered dynamics only in the domain to which they bind. Such results demonstrate that ligand binding may not structurally alter adjacent SH3/SH2 domains and implies that other aspects of protein architecture contribute to the multiple levels of regulation in proteins containing SH3 and SH2 domains.


Assuntos
Medição da Troca de Deutério , Fragmentos de Peptídeos/química , Domínios de Homologia de src/fisiologia , Sítio Alostérico , Humanos , Ligantes , Espectrometria de Massas , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/fisiologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-hck/química , Proteínas Proto-Oncogênicas c-hck/metabolismo , Proteínas Proto-Oncogênicas c-hck/fisiologia , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...