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.
Brain ; 144(12): 3710-3726, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-34972208

RESUMO

Aggregation and cytoplasmic mislocalization of TDP-43 are pathological hallmarks of amyotrophic lateral sclerosis and frontotemporal dementia spectrum. However, the molecular mechanism by which TDP-43 aggregates form and cause neurodegeneration remains poorly understood. Cyclophilin A, also known as peptidyl-prolyl cis-trans isomerase A (PPIA), is a foldase and molecular chaperone. We previously found that PPIA interacts with TDP-43 and governs some of its functions, and its deficiency accelerates disease in a mouse model of amyotrophic lateral sclerosis. Here we characterized PPIA knock-out mice throughout their lifespan and found that they develop a neurodegenerative disease with key behavioural features of frontotemporal dementia, marked TDP-43 pathology and late-onset motor dysfunction. In the mouse brain, deficient PPIA induces mislocalization and aggregation of the GTP-binding nuclear protein Ran, a PPIA interactor and a master regulator of nucleocytoplasmic transport, also for TDP-43. Moreover, in absence of PPIA, TDP-43 autoregulation is perturbed and TDP-43 and proteins involved in synaptic function are downregulated, leading to impairment of synaptic plasticity. Finally, we found that PPIA was downregulated in several patients with amyotrophic lateral sclerosis and amyotrophic lateral sclerosis-frontotemporal dementia, and identified a PPIA loss-of-function mutation in a patient with sporadic amyotrophic lateral sclerosis . The mutant PPIA has low stability, altered structure and impaired interaction with TDP-43. These findings strongly implicate that defective PPIA function causes TDP-43 mislocalization and dysfunction and should be considered in future therapeutic approaches.


Assuntos
Esclerose Lateral Amiotrófica/genética , Ciclofilina A/genética , Demência Frontotemporal/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Ciclofilina A/deficiência , Proteínas de Ligação a DNA/metabolismo , Demência Frontotemporal/patologia , Humanos , Camundongos , Camundongos Knockout
2.
J Neuroimmunol ; 339: 577118, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31790981

RESUMO

Cyclophilin A has multiple known functions in inflammation. Intracellular cyclophilin A modulates T helper 2 response (Th2) and extracellular cyclophilin A functions as a leukocyte chemotactic factor. The contribution of cyclophilin A to central nervous system (CNS) inflammation has not been reported. To test the hypothesis that inhibition of cyclophilin A would ameliorate immune-mediated CNS inflammation, we compared the course and neuroimmunology of experimental allergic encephalomyelitis (EAE) in cyclophilin A knockout mice and wild type littermates. There was a trend towards lower incidence of EAE in cyclophilin A knockout mice, but the clinical course of EAE among animals that manifested clinical signs of EAE was similar in cyclophilin A knockout and wild type littermates. Antigen recall response to myelin oligodendrocyte glycoprotein (MOG) peptide showed that interferon-γ release was lower in cyclophilin A knockout mice. Analysis of CNS inflammatory cells showed that CD3+ T cell infiltration into the CNS was lower in cyclophilin A knockout mice. These results showed that the loss of cyclophilin A results in altered peripheral immune activation and CNS leukocyte infiltration, but these changes did not result in a substantial change in the clinical course of EAE.


Assuntos
Ciclofilina A/deficiência , Encefalomielite Autoimune Experimental/metabolismo , Imunidade Celular/fisiologia , Animais , Ciclofilina A/genética , Ciclofilina A/imunologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Feminino , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Cell Mol Life Sci ; 76(20): 4165-4178, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31076805

RESUMO

Efficient cell-to-cell transfer of Listeria monocytogenes (L. monocytogenes) requires the proper formation of actin-rich membrane protrusions. To date, only the host proteins ezrin, the binding partner of ezrin, CD44, as well as cyclophilin A (CypA) have been identified as crucial components for L. monocytogenes membrane protrusion stabilization and, thus, efficient cell-to-cell movement of the microbes. Here, we examine the classical binding partner of CypA, CD147, and find that this membrane protein is also hijacked by the bacteria for their cellular dissemination. CD147 is enriched at the plasma membrane surrounding the membrane protrusions as well as the resulting invaginations generated in neighboring cells. In cells depleted of CD147, these actin-rich structures appear similar to those generated in CypA depleted cells as they are significantly shorter and more contorted as compared to their straighter counterparts formed in wild-type control cells. The presence of malformed membrane protrusions hampers the ability of L. monocytogenes to efficiently disseminate from CD147-depleted cells. Our findings uncover another important host protein needed for L. monocytogenes membrane protrusion formation and efficient microbial dissemination.


Assuntos
Basigina/genética , Membrana Celular/microbiologia , Interações Hospedeiro-Patógeno/genética , Listeria monocytogenes/fisiologia , Shigella flexneri/fisiologia , Células A549 , Actinas/genética , Actinas/metabolismo , Animais , Basigina/antagonistas & inibidores , Basigina/metabolismo , Células CACO-2 , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Ciclofilina A/deficiência , Ciclofilina A/genética , Endocitose , Fibroblastos/microbiologia , Fibroblastos/ultraestrutura , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Listeria monocytogenes/patogenicidade , Listeria monocytogenes/ultraestrutura , Camundongos , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Shigella flexneri/patogenicidade , Shigella flexneri/ultraestrutura , Transdução de Sinais
4.
Curr Opin Virol ; 14: 56-61, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26318518

RESUMO

Replication of coronaviruses is inhibited in vitro by cyclosporin A, a well-known immunosuppressive drug which binds to cellular cyclophilins thus inactivating their enzymatic cis-trans peptidyl-prolyl isomerase function. Latter is required for proper folding of cellular proteins and of proteins of several viruses. Here, we summarize present knowledge on the role of cyclophilin A during coronavirus replication. We present data on the effect of cyclophilin A single nucleotide polymorphism mutants on the replication of human CoV-229E demonstrating the requirement of proper cyclophilin A function for virus propagation. Results define cellular cyclophilin A as a host target for inhibition of coronaviruses ranging from relatively mild common cold to highly pathogenic SARS-CoV and MERS-CoV viruses with the perspective of disclosing non-immunosuppressive cyclosporin A analogs to broadly inactivate the coronavirus family.


Assuntos
Coronavirus Humano 229E/fisiologia , Ciclofilina A/deficiência , Ciclofilina A/genética , Interações Hospedeiro-Patógeno , Replicação Viral , Ciclofilina A/metabolismo , Humanos , Polimorfismo de Nucleotídeo Único
5.
Nature ; 485(7399): 512-6, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22622580

RESUMO

Human apolipoprotein E has three isoforms: APOE2, APOE3 and APOE4. APOE4 is a major genetic risk factor for Alzheimer's disease and is associated with Down's syndrome dementia and poor neurological outcome after traumatic brain injury and haemorrhage. Neurovascular dysfunction is present in normal APOE4 carriers and individuals with APOE4-associated disorders. In mice, lack of Apoe leads to blood-brain barrier (BBB) breakdown, whereas APOE4 increases BBB susceptibility to injury. How APOE genotype affects brain microcirculation remains elusive. Using different APOE transgenic mice, including mice with ablation and/or inhibition of cyclophilin A (CypA), here we show that expression of APOE4 and lack of murine Apoe, but not APOE2 and APOE3, leads to BBB breakdown by activating a proinflammatory CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes. This, in turn, leads to neuronal uptake of multiple blood-derived neurotoxic proteins, and microvascular and cerebral blood flow reductions. We show that the vascular defects in Apoe-deficient and APOE4-expressing mice precede neuronal dysfunction and can initiate neurodegenerative changes. Astrocyte-secreted APOE3, but not APOE4, suppressed the CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes through a lipoprotein receptor. Our data suggest that CypA is a key target for treating APOE4-mediated neurovascular injury and the resulting neuronal dysfunction and degeneration.


Assuntos
Apolipoproteínas E/metabolismo , Barreira Hematoencefálica/fisiologia , Circulação Cerebrovascular/fisiologia , Ciclofilina A/metabolismo , Animais , Apolipoproteína E2/deficiência , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E3/deficiência , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/deficiência , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/deficiência , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Microcirculação , NF-kappa B/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Pericitos/metabolismo
6.
J Mol Cell Cardiol ; 53(1): 6-14, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22446162

RESUMO

Extracellular cyclophilin A (CyPA) and its receptor Extracellular Matrix Metalloproteinase Inducer (EMMPRIN, CD147) modulate inflammatory processes beyond metalloproteinase (MMP) activity. Recently, we have shown that CyPA and CD147 are upregulated in patients with inflammatory cardiomyopathy. Here we investigate the role of CyPA and CD147 in murine coxsackievirus B3 (CVB3)-induced myocarditis. CVB3-infected CyPA(-/-) mice (129S6/SvEv) revealed a significantly reduced T-cell and macrophage recruitment at 8 days p.i. compared to wild-type mice. In A.BY/SnJ mice, treatment with the cyclophilin-inhibitor NIM811 was associated with a reduction of inflammatory lesions and MMP-9 expression but with enhanced virus replication 8 days p.i. At 28 days p.i. the extent of lesion areas was not affected bei NIM811, whereas the collagen content was reduced. Initiation of NIM811-treatment on day 12 (after an effective virus defense) resulted in an even more pronounced reduction of myocardial fibrosis. In conclusion, in CVB3-induced myocarditis CyPA is important for macrophage and T cell recruitment and effective virus defense and may represent a pharmacological target to modulate myocardial remodeling in myocarditis.


Assuntos
Infecções por Coxsackievirus/imunologia , Ciclofilina A/metabolismo , Fibrose Endomiocárdica/imunologia , Enterovirus Humano B/imunologia , Miocardite/imunologia , Animais , Basigina/metabolismo , Linhagem Celular , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/deficiência , Ciclosporina/farmacologia , Fibrose Endomiocárdica/etiologia , Fibrose Endomiocárdica/patologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Miocardite/complicações , Miocardite/virologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Remodelação Ventricular/efeitos dos fármacos
7.
Arterioscler Thromb Vasc Biol ; 31(5): 1116-23, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21330604

RESUMO

OBJECTIVE: Cyclophilin A (CyPA, encoded by Ppia) is a proinflammatory protein secreted in response to oxidative stress in mice and humans. We recently demonstrated that CyPA increased angiotensin II (Ang II)-induced reactive oxygen species (ROS) production in the aortas of apolipoprotein E (Apoe)-/- mice. In this study, we sought to evaluate the role of CyPA in Ang II-induced cardiac hypertrophy. METHODS AND RESULTS: Cardiac hypertrophy was not significantly different between Ppia+/+ and Ppia-/- mice infused with Ang II (1000 ng/min per kg for 4 weeks). Therefore, we investigated the effect of CyPA under conditions of high ROS and inflammation using the Apoe-/- mice. In contrast to Apoe-/- mice, Apoe-/-Ppia-/- mice exhibited significantly less Ang II-induced cardiac hypertrophy. Bone marrow cell transplantation showed that CyPA in cells intrinsic to the heart plays an important role in the cardiac hypertrophic response. Ang II-induced ROS production, cardiac fibroblast proliferation, and cardiac fibroblast migration were markedly decreased in Apoe-/-Ppia-/- cardiac fibroblasts. Furthermore, CyPA directly induced the hypertrophy of cultured neonatal cardiac myocytes. CONCLUSIONS: CyPA is required for Ang II-mediated cardiac hypertrophy by directly potentiating ROS production, stimulating the proliferation and migration of cardiac fibroblasts, and promoting cardiac myocyte hypertrophy.


Assuntos
Apolipoproteínas E/deficiência , Cardiomegalia/enzimologia , Ciclofilina A/metabolismo , Miocárdio/enzimologia , Angiotensina II , Animais , Animais Recém-Nascidos , Apolipoproteínas E/genética , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/imunologia , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Comunicação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Ciclofilina A/deficiência , Ciclofilina A/genética , Modelos Animais de Doenças , Fibroblastos/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Tempo
8.
Cell Death Dis ; 1: e84, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21368857

RESUMO

Cranial radiotherapy in children often leads to progressive cognitive decline. We have established a rodent model of irradiation-induced injury to the young brain. A single dose of 8 Gy was administered to the left hemisphere of postnatal day 10 (P10) mice. Harlequin (Hq) mice, carrying the hypomorphic apoptosis-inducing factor AIF(Hq) mutation, express 60% less AIF at P10 and displayed significantly fewer dying cells in the subventricular zone (SVZ) 6 h after IR, compared with wild type (Wt) littermates. Irradiated cyclophilin A-deficient (CypA(-/-)) mice confirmed that CypA has an essential role in AIF-induced apoptosis after IR. Hq mice displayed no reduction in SVZ size 7 days after IR, whereas 48% of the SVZ was lost in Wt mice. The proliferation rate was lower in the SVZ of Hq mice. Cultured neural precursor cells from the SVZ of Hq mice displayed a slower proliferation rate and were more resistant to IR. IR preferentially kills proliferating cells, and the slower proliferation rate in the SVZ of Hq mice may, at least partly, explain the protective effect of the Hq mutation. Together, these results indicate that targeting AIF may provide a fruitful strategy for protection of normal brain tissue against the detrimental side effects of IR.


Assuntos
Fator de Indução de Apoptose/genética , Encéfalo/efeitos da radiação , Radiação Ionizante , Animais , Fator de Indução de Apoptose/deficiência , Fator de Indução de Apoptose/metabolismo , Encéfalo/citologia , Proliferação de Células , Células Cultivadas , Ciclofilina A/deficiência , Ciclofilina A/genética , Ciclofilina A/metabolismo , Ictiose Lamelar/radioterapia , Camundongos , Camundongos Knockout , Mutação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fatores de Tempo
10.
Nat Med ; 15(6): 649-56, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19430489

RESUMO

Inflammation and oxidative stress are pathogenic mediators of many diseases, but molecules that could be therapeutic targets remain elusive. Inflammation and matrix degradation in the vasculature are crucial for abdominal aortic aneurysm (AAA) formation. Cyclophilin A (CypA, encoded by Ppia) is highly expressed in vascular smooth muscle cells (VSMCs), is secreted in response to reactive oxygen species (ROS) and promotes inflammation. Using the angiotensin II (AngII)-induced AAA model in Apoe-/- mice, we show that Apoe-/-Ppia-/- mice are completely protected from AngII-induced AAA formation, in contrast to Apoe-/-Ppia+/+ mice. Apoe-/-Ppia-/- mice show decreased inflammatory cytokine expression, elastic lamina degradation and aortic expansion. These features were not altered by reconstitution of bone marrow cells from Ppia+/+ mice. Mechanistic studies showed that VSMC-derived intracellular and extracellular CypA are required for ROS generation and matrix metalloproteinase-2 activation. These data define a previously undescribed role for CypA in AAA formation and suggest CypA as a new target for treating cardiovascular disease.


Assuntos
Angiotensina II/farmacologia , Aneurisma Aórtico/metabolismo , Aneurisma Aórtico/patologia , Ciclofilina A/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Aneurisma Aórtico/genética , Ciclofilina A/deficiência , Ciclofilina A/genética , Camundongos , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo
11.
Circulation ; 117(24): 3088-98, 2008 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-18541741

RESUMO

BACKGROUND: Oxidative stress, generated by excessive reactive oxygen species, promotes cardiovascular disease. Cyclophilin A (CyPA) is a 20-kDa chaperone protein secreted from vascular smooth muscle cells (VSMCs) in response to reactive oxygen species that stimulates VSMC proliferation and inflammatory cell migration in vitro; however, the role CyPA plays in vascular function in vivo remains unknown. METHODS AND RESULTS: We tested the hypothesis that CyPA contributes to vascular remodeling by analyzing the response to complete carotid ligation in CyPA knockout mice, wild-type mice, and mice that overexpress CyPA in VSMC (VSMC-Tg). After carotid ligation, CyPA expression in vessels of wild-type mice increased dramatically and was significantly greater in VSMC-Tg mice. Reactive oxygen species-induced secretion of CyPA from mouse VSMCs correlated significantly with intracellular CyPA expression. Intimal and medial hyperplasia correlated significantly with CyPA expression after 2 weeks of carotid ligation, with marked decreases in CyPA knockout mice and increases in VSMC-Tg mice. Inflammatory cell migration into the intima was significantly reduced in CyPA knockout mice and increased in VSMC-Tg mice. Additionally, VSMC proliferation assessed by Ki67(+) cells was significantly less in CyPA knockout mice and was increased in VSMC-Tg mice. The importance of CyPA for intimal and medial thickening was shown by strong correlations between CyPA expression and the number of both inflammatory cells and proliferating VSMCs in vivo and in vitro. CONCLUSIONS: In response to low flow, CyPA plays a crucial role in VSMC migration and proliferation, as well as inflammatory cell accumulation, thereby regulating flow-mediated vascular remodeling and intima formation.


Assuntos
Ciclofilina A/deficiência , Inflamação/fisiopatologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Animais , Aorta , Artérias Carótidas/fisiologia , Divisão Celular , Meios de Cultivo Condicionados , Ciclofilina A/genética , Ciclofilina A/fisiologia , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Túnica Íntima/citologia , Túnica Íntima/fisiologia
12.
Mol Ther ; 14(4): 546-54, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16901758

RESUMO

Lentiviral vectors derived from the human immunodeficiency virus-1 (HIV-1) have a higher propensity to transduce nondividing cells compared to vectors based on oncoretroviruses. We report here that genistein, a previously known protein tyrosine kinase (PTK) inhibitor and G2 cell cycle arrest inducer, significantly enhanced lentiviral transduction in a dose-dependent manner. Increased transduction, as measured by vector expression, was seen in a variety of human cell lines, murine primary lymphocytes, and primary human CD34(+) peripheral blood progenitor cells as well. Increased vector expression was also associated with an increase in vector DNA copy number, as assessed by quantitative PCR. Genistein-mediated G2 cell cycle arrest, rather than PTK inhibition, appears to be the major factor responsible for increased gene transfer. Genistein also increases cyclophilin A (CypA) protein, a cellular protein important for efficient HIV-1 infection. While we show that CypA(-/-) Jurkat cells transduce poorly with lentiviral vectors, genistein does increase gene transfer in CypA-deficient cells. CypA and G2 cell cycle arrest appear to be two independent factors important for efficient lentiviral gene transfer. The role of genistein and other G2-arresting agents may be useful for improving the efficiency of lentiviral gene therapy.


Assuntos
Ciclofilina A/metabolismo , Fase G2 , Lentivirus/genética , Animais , Células Cultivadas , Ciclofilina A/deficiência , Ciclofilina A/genética , Fase G2/efeitos dos fármacos , Dosagem de Genes , Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Genisteína , Humanos , Camundongos , Fatores de Tempo
14.
J Immunol ; 174(10): 6030-8, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15879096

RESUMO

Cyclosporine is an immunosuppressive drug that is widely used to prevent organ transplant rejection. Known intracellular ligands for cyclosporine include the cyclophilins, a large family of phylogenetically conserved proteins that potentially regulate protein folding in cells. Immunosuppression by cyclosporine is thought to result from the formation of a drug-cyclophilin complex that binds to and inhibits calcineurin, a serine/threonine phosphatase that is activated by TCR engagement. Amino acids within the cyclophilins that are critical for binding to cyclosporine have been identified. Most of these residues are highly conserved within the 15 mammalian cyclophilins, suggesting that many are potential targets for the drug. We examined the effects of cyclosporine on immune cells and mice lacking Ppia, the gene encoding the prototypical cyclophilin protein cyclophilin A. TCR-induced proliferation and signal transduction by Ppia(-/-) CD4(+) T cells were resistant to cyclosporine, an effect that was attributable to diminished calcineurin inhibition. Immunosuppressive doses of cyclosporine failed to block the responses of Ppia(-/-) mice to allogeneic challenge. Rag2(-/-) mice reconstituted with Ppia(-/-) splenocytes were also cyclosporine resistant, indicating that this property is intrinsic to Ppia(-/-) immune cells. Thus, among multiple potential ligands, CypA is the primary mediator of immunosuppression by cyclosporine.


Assuntos
Ciclofilina A/deficiência , Ciclofilina A/genética , Ciclosporina/administração & dosagem , Resistência a Medicamentos/genética , Resistência a Medicamentos/imunologia , Imunossupressores/administração & dosagem , Animais , Calcineurina/fisiologia , Inibidores de Calcineurina , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclofilina A/fisiologia , Ciclosporina/farmacologia , Inativação Gênica/efeitos dos fármacos , Inativação Gênica/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/genética , Imunossupressores/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Sarcoma Experimental/enzimologia , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Baço/citologia , Baço/imunologia , Baço/transplante , Linfócitos T/efeitos dos fármacos , Linfócitos T/enzimologia , Linfócitos T/metabolismo
15.
J Virol ; 77(7): 4431-4, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12634401

RESUMO

We have previously shown that virions with nef deleted can be restored to wild-type infectivity by treatment to induce natural endogenous reverse transcription (NERT). Since Nef and cyclophilin A (CyPA) appear to act in similar ways on postentry events, we determined whether NERT treatment would restore infectivity to virions depleted of CyPA. Our results show that the infectivity of virions depleted of CyPA by treatment with cyclosporine A could be restored by NERT treatment, while mutants in the CyPA binding loop of capsid could only be partially restored. These results suggest that CyPA is involved in some aspect of the uncoating process.


Assuntos
Ciclofilina A/fisiologia , HIV-1/patogenicidade , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/fisiologia , Linhagem Celular , Ciclofilina A/deficiência , Ciclosporina/farmacologia , Genes Virais , Genes nef , HIV-1/efeitos dos fármacos , HIV-1/genética , HIV-1/fisiologia , Humanos , Imunossupressores/farmacologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Mutação , Transcrição Gênica , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...