Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 131(22)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34779418

RESUMO

Metabolic pathways regulate immune responses and disrupted metabolism leads to immune dysfunction and disease. Coronavirus disease 2019 (COVID-19) is driven by imbalanced immune responses, yet the role of immunometabolism in COVID-19 pathogenesis remains unclear. By investigating 87 patients with confirmed SARS-CoV-2 infection, 6 critically ill non-COVID-19 patients, and 47 uninfected controls, we found an immunometabolic dysregulation in patients with progressed COVID-19. Specifically, T cells, monocytes, and granulocytes exhibited increased mitochondrial mass, yet only T cells accumulated intracellular reactive oxygen species (ROS), were metabolically quiescent, and showed a disrupted mitochondrial architecture. During recovery, T cell ROS decreased to match the uninfected controls. Transcriptionally, T cells from severe/critical COVID-19 patients showed an induction of ROS-responsive genes as well as genes related to mitochondrial function and the basigin network. Basigin (CD147) ligands cyclophilin A and the SARS-CoV-2 spike protein triggered ROS production in T cells in vitro. In line with this, only PCR-positive patients showed increased ROS levels. Dexamethasone treatment resulted in a downregulation of ROS in vitro and T cells from dexamethasone-treated patients exhibited low ROS and basigin levels. This was reflected by changes in the transcriptional landscape. Our findings provide evidence of an immunometabolic dysregulation in COVID-19 that can be mitigated by dexamethasone treatment.


Assuntos
Basigina/fisiologia , COVID-19/imunologia , Dexametasona/farmacologia , SARS-CoV-2 , Linfócitos T/metabolismo , Adulto , COVID-19/metabolismo , Ciclofilina A/fisiologia , Ácidos Graxos/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mitocôndrias/patologia , Espécies Reativas de Oxigênio/metabolismo
2.
FASEB J ; 35(11): e21958, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34606626

RESUMO

Cyclophilin A (CypA), a member of the cyclophilin family, plays a vital role in microorganismal infections, inflammatory diseases, and cancers. Interleukin-6 (IL-6) is a pleiotropic cytokine, exerting variety of effects on inflammation, immune response, hematopoiesis, and tumor proliferation. Binding of IL-6 to soluble IL-6 receptor (sIL-6R) induces pro-inflammatory trans-signaling, which has been described to be stronger than anti-inflammatory classic signaling triggered by the binding of IL-6 to membrane-bound IL-6 receptor. Here we found that upon the treatment of IL-6 and sIL-6R, CypA inhibited the ubiquitination-mediated degradation of IL-6 membrane receptor gp130 and enhanced its dimerization, thereby positively regulated the IL-6 trans-signaling and increased the expression of downstream iNOS, IL-6, and CypA. Furthermore, CypA expression could be negatively regulated by suppressor of cytokine signaling 1 (SOCS1). The SH2 and Box domains of SOCS1 interacted with CypA and promoted its K48-linked ubiquitination-mediated degradation, which inhibited the IL-6 trans-signaling pathway. Collectively, our findings reveal an important role of CypA in the positive and negative feedback regulation of the IL-6 trans-signaling pathway.


Assuntos
Ciclofilina A/fisiologia , Interleucina-6/metabolismo , Receptores de Interleucina-6/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Células A549 , Células HEK293 , Humanos , Transdução de Sinais
3.
J Periodontal Res ; 55(5): 705-712, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32406527

RESUMO

OBJECTIVE: This in vitro study aimed to evaluate the expression of cyclophilin A (CyPA) in U937 monocytic cells after coculturing with the human gingival fibroblasts (HGFs) and the effect of CyPA on the augmentation of MMP-2 expression in the coculture environment. BACKGROUND: Leukocyte infiltration in gingival connective tissue is one of the major findings in the lesions of inflammatory periodontal diseases. A crosstalk between the resident gingival fibroblasts and the recruited inflammatory cells that promote the expression of matrix metalloproteinases (MMPs) was proposed based on recent findings, whereas the cluster of differentiation 147 (CD147)-CyPA pathway was suggested to be involved with the crosstalk. MATERIAL AND METHODS: CyPA was released into media, in the independent or transwell coculture of HGF and U937 cells, as determined by enzyme-linked immunosorbent assay, whereas intracellular mRNA expressions for CyPA and MMP-2 were examined by quantitative real-time polymerase chain reaction, in the transwell coculture or conditional medium models. Zymography was conducted to analyze the activities of pro-MMP-2/MMP-2 released into the media. RESULTS: (a) A significantly increased CyPA protein level was observed in the transwell coculture media compared with that in the independent culture. (b) The transwell coculture-enhanced mRNA expression for CyPA was noticed in U937 cells but not in HGFs. After adding with HGF-conditioned medium, the mRNA enhancement in U937 cells occurred in a dose-dependent manner. (c) Although the MMP-2 activities significantly increased after transwell coculturing, the MMP-2 mRNA enhancement was observed only in HGFs. (d) Exogenous CyPA could enhance MMP-2 activities in HGFs in a dose-dependent manner. However, the CyPA antagonist reduced the MMP-2 activities in the transwell cocultures. (e) Moreover, the CyPA-enhanced MMP-2 activity in HGF was decreased significantly by the pathway inhibitor for c-Jun amino-terminal kinase (JNK). CONCLUSION: Based on the present findings, we suggest that gingival fibroblasts could enhance the CyPA release from U937 cells, via the JNK pathway, resulting in MMP-2 enhancement in fibroblasts. The finding shed light on a new mechanism of cellular interaction involving MMP-2 and CyPA, in two cells.


Assuntos
Ciclofilina A , Gengiva , Metaloproteinase 2 da Matriz , Células Cultivadas , Ciclofilina A/fisiologia , Fibroblastos/metabolismo , Gengiva/metabolismo , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Células U937
4.
Curr Issues Mol Biol ; 21: 1-20, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27033630

RESUMO

Cyclophilin A (CypA) is a key member of immunophilins that has peptidyl-prolyl cis/trans isomerase (PPIase) activity. Besides acting as a cellular receptor for immunosuppressive drug cyclosporine A (CsA), CypA is involved in various cellular activities. CypA has an important role in viral infection which either facilitates or inhibits their replication. Inhibition of CypA via inhibitors is useful for overcoming several viral infections, indicating that CypA is an attractive target for anti-viral therapy. Collectively, these facts demonstrate the critical roles of CypA in mediating or inhibiting viral infections, suggesting that CypA can be an attractive cellular target for the development of anti-viral therapy.


Assuntos
Antivirais/farmacologia , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Replicação Viral/fisiologia , Humanos , Terapia de Alvo Molecular/métodos , Viroses/metabolismo , Viroses/virologia , Replicação Viral/efeitos dos fármacos
7.
Crit Care Med ; 43(9): e369-81, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26132882

RESUMO

OBJECTIVES: Cyclophilin A has been found to be involved in many inflammatory diseases via its receptor, cluster of differentiation 147 (CD147). This study was designed to estimate the potential role of cyclophilin A/CD147 in subarachnoid hemorrhage-induced early brain injury. DESIGN: Controlled in vivo laboratory study. SETTING: Animal research laboratory. SUBJECTS: Two hundred ninety adult male Sprague-Dawley rats weighing 300-350 g. INTERVENTIONS: A prechiasmatic cistern single-injection model was used to produce experimental subarachnoid hemorrhage in Sprague-Dawley rats. The expressions of cyclophilin A and CD147, the interaction between cyclophilin A and CD147, and the secretion of cyclophilin A were assessed using immunofluorescence staining, Western blot analysis, and coimmunoprecipitation analysis. Down-regulation of cyclophilin A expression by small interfering RNA was performed, and recombinant human cyclophilin A and monoclonal antibody of CD147 were exploited to study the role of cyclophilin A/CD147 in subarachnoid hemorrhage-induced early brain injury. MEASUREMENTS AND MAIN RESULTS: The expressions of cyclophilin A and CD147 in neurons were higher than that of the sham group and peaked at 24 hours after subarachnoid hemorrhage. Compared with sham group, subarachnoid hemorrhage was found to increase the secretion of cyclophilin A and the interaction between cyclophilin A and CD147. Cyclophilin A small interfering RNA and anti-CD147 treatments were found to ameliorate subarachnoid hemorrhage-induced early brain injury, including cortical apoptosis and necrosis, brain edema, blood-brain barrier damage, and neurobehavioral deficits. Cyclophilin A small interfering RNA and anti-CD147 treatments also decreased the phosphorylation of extracellular signal-regulated protein kinase 1/2, the protein levels of p53 and caspase-3, and the level of active nuclear factor-κB. Finally, recombinant human cyclophilin A treatment resulted in an opposite effect, which was inhibited by anti-CD147 treatment. CONCLUSIONS: Cyclophilin A/CD147 interactions may participate in subarachnoid hemorrhage-induced early brain injury via increasing neuronal apoptosis pathway, at least partly through the ERK1/2-nuclear factor-κB pathway. Cyclophilin A/CD147 may be a suitable therapeutic target for subarachnoid hemorrhage.


Assuntos
Basigina/fisiologia , Lesões Encefálicas/fisiopatologia , Ciclofilina A/fisiologia , Fármacos Neuroprotetores , Hemorragia Subaracnóidea/fisiopatologia , Animais , Apoptose/fisiologia , Barreira Hematoencefálica/metabolismo , Western Blotting , Edema Encefálico/fisiopatologia , Modelos Animais de Doenças , Masculino , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley
8.
PLoS One ; 10(3): e0121666, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25822622

RESUMO

TRIMCyp is a fusion protein consisting of the TRIM5 gene product and retrotransposed Cyclophilin A (CypA). Two primate TRIMCyp fusion proteins with varying anti-HIV-1 activities independently evolved in owl monkeys and Old World monkeys. In addition, Old World monkey TRIMCyps lack exon7, which encodes amino acids in the Linker2 region. Previous studies on TRIM5α indicated that this region affects anti-retroviral activity, cytoplasmic body formation, and multimerization. The effects of exon7 deletion on the functions of the TRIMCyp are unclear. In this study, we found that the cytoplasmic bodies and multimers of owl monkey TRIMCyp (omTRIMCyp) are different from those of northern pig-tailed macaque TRIMCyp (npmTRIMCyp). In addition, we demonstrated that exon7 deletion affected cytoplasmic body formation and multimerization. Moreover, we unexpectedly found two chimeric proteins of omTRIMCyp and npmTRIMCyp that failed to block HIV-1 replication, despite the presence of CypA in omTRIMCyp. Further studies indicated that the cytoplasmic bodies and spontaneous multimerization were not responsible for TRIMCyp anti-HIV-1 activity. Moreover, potent viral restriction is associated with higher amounts of monomeric TRIMCyp when the CypA domain is able to recognize and bind to the HIV-1 capsid. Our results suggested that the deletion of exon7 during the evolution of TRIMCyp affected its function.


Assuntos
Proteínas de Transporte/genética , Ciclofilina A/genética , HIV-1/fisiologia , Animais , Aotidae/genética , Aotidae/virologia , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Gatos , Linhagem Celular , Ciclofilina A/química , Ciclofilina A/fisiologia , Citoplasma/genética , Citoplasma/virologia , Evolução Molecular , Éxons , Células HEK293 , HIV-1/patogenicidade , Especificidade de Hospedeiro , Humanos , Macaca nemestrina/genética , Macaca nemestrina/virologia , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Deleção de Sequência , Virulência , Montagem de Vírus , Replicação Viral
10.
Cell Death Dis ; 4: e888, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24176846

RESUMO

Cyclophilin A (CyPA) is a ubiquitously distributed protein belonging to the immunophilin family. CyPA has peptidyl prolyl cis-trans isomerase (PPIase) activity, which regulates protein folding and trafficking. Although CyPA was initially believed to function primarily as an intracellular protein, recent studies have revealed that it can be secreted by cells in response to inflammatory stimuli. Current research in animal models and humans has provided compelling evidences supporting the critical function of CyPA in several human diseases. This review discusses recently available data about CyPA in cardiovascular diseases, viral infections, neurodegeneration, cancer, rheumatoid arthritis, sepsis, asthma, periodontitis and aging. It is believed that further elucidations of the role of CyPA will provide a better understanding of the molecular mechanisms underlying these diseases and will help develop novel pharmacological therapies.


Assuntos
Ciclofilina A/metabolismo , Ciclofilina A/fisiologia , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Ciclofilina A/genética , Humanos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo
11.
Biochem Biophys Res Commun ; 424(4): 647-50, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22814107

RESUMO

Cyclophilin A (CyPA) is a peptidyl-prolyl cis/trans isomerase originally identified as the target of the immunosuppressive drug cyclosporine A. A number of reports have demonstrated that CyPA plays a critical role in the successful replication of viruses such as human immunodeficiency virus (HIV), hepatitis C virus (HCV), hepatitis B virus (HBV), etc. However, recent studies demonstrated that CyPA also possesses a repressive effect on the replication of some viruses like Influenza A virus and rotavirus. Moreover, CyPA could also regulate host IFN-I response to viral infections. Together, these evidences showed diverse roles of CyPA in viral infection.


Assuntos
Ciclofilina A/fisiologia , Viroses/enzimologia , Viroses/virologia , Replicação Viral , Ciclofilina A/genética , Infecções por HIV/enzimologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Hepacivirus/fisiologia , Hepatite B/enzimologia , Hepatite B/imunologia , Vírus da Hepatite B/fisiologia , Hepatite C/imunologia , Humanos , Vírus da Influenza A/fisiologia , Influenza Humana/enzimologia , Influenza Humana/imunologia , Interferon Tipo I/imunologia , Viroses/imunologia
12.
J Virol ; 86(18): 9875-87, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761365

RESUMO

Human papillomaviruses (HPV) are composed of the major and minor capsid proteins, L1 and L2, that encapsidate a chromatinized, circular double-stranded DNA genome. At the outset of infection, the interaction of HPV type 16 (HPV16) (pseudo)virions with heparan sulfate proteoglycans triggers a conformational change in L2 that is facilitated by the host cell chaperone cyclophilin B (CyPB). This conformational change results in exposure of the L2 N terminus, which is required for infectious internalization. Following internalization, L2 facilitates egress of the viral genome from acidified endosomes, and the L2/DNA complex accumulates at PML nuclear bodies. We recently described a mutant virus that bypasses the requirement for cell surface CyPB but remains sensitive to cyclosporine for infection, indicating an additional role for CyP following endocytic uptake of virions. We now report that the L1 protein dissociates from the L2/DNA complex following infectious internalization. Inhibition and small interfering RNA (siRNA)-mediated knockdown of CyPs blocked dissociation of L1 from the L2/DNA complex. In vitro, purified CyPs facilitated the dissociation of L1 pentamers from recombinant HPV11 L1/L2 complexes in a pH-dependent manner. Furthermore, CyPs released L1 capsomeres from partially disassembled HPV16 pseudovirions at slightly acidic pH. Taken together, these data suggest that CyPs mediate the dissociation of HPV L1 and L2 capsid proteins following acidification of endocytic vesicles.


Assuntos
Proteínas do Capsídeo/fisiologia , Ciclofilinas/fisiologia , Papillomavirus Humano 16/fisiologia , Proteínas Oncogênicas Virais/fisiologia , Substituição de Aminoácidos , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/genética , Ciclofilina A/fisiologia , Ciclofilinas/antagonistas & inibidores , Ciclofilinas/genética , DNA Viral/química , DNA Viral/genética , DNA Viral/metabolismo , Endossomos/fisiologia , Endossomos/virologia , Técnicas de Silenciamento de Genes , Genoma Viral , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidade , Humanos , Concentração de Íons de Hidrogênio , Substâncias Macromoleculares , Mutagênese Sítio-Dirigida , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/genética , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Internalização do Vírus
13.
Blood ; 120(6): 1317-26, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22740452

RESUMO

Platelet adhesion and aggregation play a critical role in primary hemostasis. Uncontrolled platelet activation leads to pathologic thrombus formation and organ failure. The decisive central step for different processes of platelet activation is the increase in cytosolic Ca(2+) activity ([Ca(2+)](i)). Activation-dependent depletion of intracellular Ca(2+) stores triggers Ca(2+) entry from the extracellular space. Stromal interaction molecule 1 (STIM1) has been identified as a Ca(2+) sensor that regulates store-operated Ca(2+) entry through activation of the pore-forming subunit Orai1, the major store-operated Ca(2+) entry channel in platelets. In the present study, we show for the first time that the chaperone protein cyclophilin A (CyPA) acts as a Ca(2+) modulator in platelets. CyPA deficiency strongly blunted activation-induced Ca(2+) mobilization from intracellular stores and Ca(2+) influx from the extracellular compartment and thus impaired platelet activation substantially. Furthermore, the phosphorylation of the Ca(2+) sensor STIM1 was abrogated upon CyPA deficiency, as shown by immunoprecipitation studies. In a mouse model of arterial thrombosis, CyPA-deficient mice were protected against arterial thrombosis, whereas bleeding time was not affected. The results of the present study identified CyPA as an important Ca(2+) regulator in platelets, a critical mechanism for arterial thrombosis.


Assuntos
Plaquetas/metabolismo , Cálcio/metabolismo , Ciclofilina A/fisiologia , Trombose/genética , Animais , Células CHO , Sinalização do Cálcio/genética , Degranulação Celular/genética , Degranulação Celular/fisiologia , Cricetinae , Cricetulus , Ciclofilina A/genética , Ciclofilina A/metabolismo , Integrina beta3/metabolismo , Espaço Intracelular/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Doença Arterial Periférica/genética , Doença Arterial Periférica/metabolismo , Ativação Plaquetária/genética , Trombose/metabolismo
14.
Can J Physiol Pharmacol ; 90(8): 1005-15, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22647032

RESUMO

The pathophysiology of vascular disease in diabetes involves abnormalities in endothelial cells, vascular smooth muscle cells, and monocytes. The metabolic abnormalities that characterize diabetes, such as hyperglycemia, increased free fatty acids, and insulin resistance, each provoke molecular mechanisms that contribute to vascular dysfunction. Several molecules have been identified as risk markers, and have been studied to prevent progression of disease and long-term complications. Markers such as C-reactive protein and monocyte chemoattractant protein-1 are used to assess risk for adverse cardiac events, but elevated levels are possible due to the presence of other risk factors as part of the natural physiological defense mechanism. In this review we discuss potential of cyclophilin-A, a secreted oxidative-stress-induced immunophilin with diverse functions. We present evidence for a significant role of cyclophilin-A in the pathogenesis of atherosclerosis in diabetes, and its potential as a marker for vascular disease in type-2 diabetes.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Ciclofilina A/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Biomarcadores/metabolismo , Proteína C-Reativa/metabolismo , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/complicações , Ciclofilina A/fisiologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Humanos , Inflamação/fisiopatologia , Mediadores da Inflamação/sangue , Modelos Cardiovasculares , Monócitos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Risco
15.
Am J Transplant ; 12(9): 2348-62, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22642569

RESUMO

Immunosuppressants have impacts on the development of polyomavirus-associated nephropathy. We previously demonstrated that cyclosporin A (CsA) suppressed polyomavirus BK (BKV) replication. The role of cyclophilin A (CypA) and nuclear factor of activated T cells (NFAT) in CsA-imposed suppression of BKV replication was determined in this study. Results demonstrated that knockdown of CypA but not CypB significantly reduced BKV large T antigen (TAg) expression and BKV titer. Overexpression of CypA reversed CypA siRNA-induced inhibition in BKV TAg expression. In addition, CypA overexpression attenuated the suppressive effect of CsA on TAg expression, suggesting CypA implicated in CsA-mediated anti-BKV effect. Knockdown of NFATc3 abrogated TAg expression, while overexpression of NFATc3 promoted TAg expression and augmented BKV promoter activity. NFATc3 binding to the BKV promoter was verified by chromatin immunoprecipitation assay and electrophoretic mobility shift assay. Renal histology also displayed an increase in NFATc3 expression in tubulointerstitium of BKV-associated nephropathy. Furthermore, overexpression of NFATc3 rescued CsA-mediated inhibition of BKV load and TAg expression. A CsA analog, NIM811, which cannot block NFAT functionality, failed to suppress TAg expression. In conclusion, CypA and NFAT are indispensable in BKV replication. CsA inhibits BKV replication through CypA and NFAT, which may be potential targets of anti-BKV treatment.


Assuntos
Vírus BK/fisiologia , Ciclofilina A/fisiologia , Ciclosporina/farmacologia , Fatores de Transcrição NFATC/fisiologia , Replicação Viral/efeitos dos fármacos , Vírus BK/isolamento & purificação , Linhagem Celular Transformada , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Inativação Gênica , Humanos , Regiões Promotoras Genéticas , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Carga Viral
16.
PLoS One ; 7(5): e37277, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615963

RESUMO

The immunosuppressive drug cyclosporin A (CsA) has inhibitory effects on the replication of several viruses. The antiviral effects are through targeting the interaction between viral proteins and host factor cyclophilin A (CypA). CypA has been identified to interact with influenza A virus M1 protein and impair the early stage of the viral life cycle. In order to identify the effect of CsA on influenza virus replication, a CypA-depleted 293T cell line, which was named as 293T/CypA-, was constructed. The cytopathic effect (CPE) assay and the growth curve results indicated that CsA specifically suppressed the influenza A virus replication in a dose-dependent manner. CsA treatment had no effect on the viral genome replication and transcription but selectively suppressed the viral proteins expression. Further studies indicated that CsA could impair the nuclear export of viral mRNA in the absence of CypA. In addition, the antiviral activity of CsA was independent of calcineurin signaling. Finally, CsA could enhance the binding between CypA and M1. The above results suggested that CsA inhibited the replication of influenza A virus through CypA-dependent and -independent pathways.


Assuntos
Ciclofilina A/farmacologia , Ciclosporina/farmacologia , Vírus da Influenza A/fisiologia , Replicação Viral/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Calcineurina/fisiologia , Linhagem Celular , Núcleo Celular/metabolismo , Ciclofilina A/fisiologia , Cães , Vírus da Influenza A/efeitos dos fármacos , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Transdução de Sinais/fisiologia , Proteínas da Matriz Viral/metabolismo
17.
Biochem Biophys Res Commun ; 422(4): 664-9, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22609402

RESUMO

Rotavirus (RV) infection causes serious dehydrating diarrhoea in infants and newborn animals. Our previous study revealed that cyclophilin A (CYPA), a peptidyl-prolyl cis-trans isomerase (PPIase), could be temporarily upregulated in RV-infected MA104 cells in early stage of infection (unpublished data). To find out the possible roles of CYPA in RV infection, we overexpressed and silenced CYPA in various cell lines by gene transfection and shRNA. We found that transfection of CYPA significantly inhibited RV replication, while silencing the expression of CYPA significantly increased RV replication. Accordingly, overexpression of CYPA significantly increased IFN-ß production; while silencing CYPA significantly reduced IFN-ß production. This effect of CYPA on IFN-ß production was independent of its PPIase activity. Moreover, IFN-ß secreted by host cells in RV infection had a critical repressive effect on viral replication. Finally, we found that inhibiting JNK pathway by SP600125 and JNK siRNA abrogated the effect of CYPA on IFN-ß transcription in RV-infected MA104 cells. Together, our data suggested that CYPA inhibited RV replication by facilitating host IFN-ß production, which was independent on the PPIase activity of CYPA but dependent on the activation of JNK signaling pathway.


Assuntos
Ciclofilina A/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Interferon beta/biossíntese , Infecções por Rotavirus/metabolismo , Rotavirus/fisiologia , Replicação Viral , Animais , Células CACO-2 , Linhagem Celular , Ciclofilina A/genética , Células HEK293 , Humanos , Interferon beta/genética , MAP Quinase Quinase 4/metabolismo , Infecções por Rotavirus/virologia , Transcrição Gênica
18.
Neurobiol Dis ; 46(3): 745-58, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22426396

RESUMO

Neuronal programmed cell death (PCD) contributes to delayed tissue damage after traumatic brain injury (TBI). Both caspase-dependent and caspase-independent mechanisms have been implicated, with the latter including apoptosis inducing factor (AIF). The peptidyl-proplyl isomerase Cyclophilin A (CypA) transports AIF from the cytosol to the nucleus, a key step for AIF-dependent cell death. We compared the effects of single versus combined inhibition of caspase and AIF pathways in a mouse controlled cortical impact (CCI) model, by examining the effects of CypA gene knockout (CypA(-/-)), caspase inhibition with a pan-caspase inhibitor (boc-aspartyl(OMe)-fluoromethylketone, BAF), or combined modulation. TBI caused caspase activation as well as translocation of AIF to the nucleus. Markers of caspase activation including caspase-specific fodrin cleavage fragments and number of FLIVO-positive cells were reduced in BAF-treated CypA(+/+) mice, whereas markers of AIF activation including AIF/H2AX interaction and AIF translocation to the nucleus were attenuated in CypA(-/-) mice. Each single intervention, (CypA(-/-) or BAF-treated CypA(+/+)) reduced the number of apoptotic cells (TUNEL-positive) in the cortex and improved long-term sensorimotor function; CypA(-/-) also attenuated microglial activation after injury. Importantly, BAF-treated CypA(-/-) mice, showed greater effects than either intervention alone on multiple outcomes including: reduction in TUNEL-positive cells, decrease in neuroinflammation, improved motor and cognitive recovery, and attenuation of lesion volume and neuronal loss in the hippocampus. Using two in vitro neuronal cell death models known to induce AIF-mediated PCD, we also showed that neurons from CypA(-/-) animals were protected and that effects were unrelated to caspase activation. These data indicate that AIF-mediated and caspase-dependent pathways contribute independently and in parallel to secondary injury after TBI, and suggest that combined therapeutic strategies directed at multiple PCD pathways may provide superior neuroprotection than those directed at single mechanisms.


Assuntos
Fator de Indução de Apoptose/farmacologia , Lesões Encefálicas/tratamento farmacológico , Caspases/farmacologia , Morte Celular/fisiologia , Fármacos Neuroprotetores , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Western Blotting , Lesões Encefálicas/patologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cognição/efeitos dos fármacos , Ciclofilina A/genética , Ciclofilina A/fisiologia , Inibidores de Cisteína Proteinase/farmacologia , Hipocampo/patologia , Imuno-Histoquímica , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Injeções Intraventriculares , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , Movimento/efeitos dos fármacos , Neurônios/patologia , Transdução de Sinais/efeitos dos fármacos
19.
J Gen Virol ; 93(Pt 4): 722-732, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22238232

RESUMO

Human cytomegalovirus (HCMV) is a large DNA virus belonging to the subfamily Betaherpesvirinae. Haematopoietic cells of the myeloid lineage have been shown to harbour latent HCMV. However, following terminal differentiation of these cells, virus is reactivated, and in an immunocompromised host acute infection can occur. It is currently unknown which viral and cellular factors are involved in regulating the switch between lytic and latent infections. Cyclophilin A (CyPA) is a cellular protein that acts as a major factor in virus replication and/or virion maturation for a number of different viruses, including human immunodeficiency virus, hepatitis C virus, murine cytomegalovirus, influenza A virus and vaccinia virus. This study investigated the role of CyPA during HCMV infection. CyPA expression was silenced in human foreskin fibroblast (HF) and THP-1 cells using small interfering RNA (siRNA) technology, or the cells were treated with cyclosporin A (CsA) to inhibit CyPA activity. Silencing CyPA in HF cells with siRNA resulted in an overall reduction in virus production characterized by delayed expression of immediate-early (IE) proteins, decreased viral DNA loads and reduced titres. Furthermore, silencing of CyPA in THP-1 cells pre- and post-differentiation prevented IE protein expression and virus reactivation from a non-productive state. Interestingly, it was observed that treatment of THP-1 cells with CsA prevented the cells from establishing a fully latent infection. In summary, these results demonstrate that CyPA expression is an important factor in HCMV IE protein expression and virus production in lytically infected HF cells, and is a major component in virus reactivation from infected THP-1 cells.


Assuntos
Ciclofilina A/fisiologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Ativação Viral/fisiologia , Replicação Viral/fisiologia , Western Blotting , Infecções por Citomegalovirus/metabolismo , Replicação do DNA/fisiologia , DNA Viral/metabolismo , Fibroblastos/virologia , Inativação Gênica , Genes Virais/fisiologia , Humanos , Proteínas Imediatamente Precoces/metabolismo , Proteínas Imediatamente Precoces/fisiologia , Reação em Cadeia da Polimerase em Tempo Real
20.
Mol Cancer ; 10: 102, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21871105

RESUMO

BACKGROUND: Cyclophilin A (CypA) expression is associated with malignant phenotypes in many cancers. However, the role and mechanisms of CypA in liver fluke-associated cholangiocarcinoma (CCA) are not presently known. In this study, we investigated the expression of CypA in CCA tumor tissues and CCA cell lines as well as regulation mechanisms of CypA in tumor growth using CCA cell lines. METHODS: CypA expression was determined by real time RT-PCR, Western blot or immunohistochemistry. CypA silence or overexpression in CCA cells was achieved using gene delivery techniques. Cell proliferation was assessed using MTS assay or Ki-67 staining. The effect of silencing CypA on CCA tumor growth was determined in nude mice. The effect of CypA knockdown on ERK1/2 activation was assessed by Western blot. RESULTS: CypA was upregulated in 68% of CCA tumor tissues. Silencing CypA significantly suppressed cell proliferation in several CCA cell lines. Likewise, inhibition of CypA peptidyl-prolyl cis-trans isomerase (PPIase) activity using cyclosporin A (CsA) decreased cell proliferation. In contrast, overexpression of CypA resulted in 30% to 35% increases in proliferation of CCA cell lines. Interestingly, neither silence nor overexpression of CypA affected cell proliferation of a non-tumor human cholangiocyte cell line, MMNK1. Suppression of CypA expression attenuated ERK1/2 activity in CCA M139 cells by using both transient and stable knockdown methods. In the in vivo study, there was a 43% reduction in weight of tumors derived from CypA-silenced CCA cell lines compared with control vector CCA tumors in mice; these tumors with stable CypA silencing showed a reduced cell proliferation. CONCLUSIONS: CypA is upregulated in majority of CCA patients' tissues and confers a significant growth advantage in CCA cells. Suppression of CypA expression decreases proliferation of CCA cell lines in vitro and reduces tumor growth in the nude mouse model. Inhibition of CypA activity also reduces CCA cell proliferation. The ERK1/2 pathway may be involved in the CypA-mediated CCA cell proliferation. Thus, CypA may represent an important new therapeutic target for liver fluke-associated CCA.


Assuntos
Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos/patologia , Proliferação de Células , Colangiocarcinoma/genética , Ciclofilina A/fisiologia , Fasciola hepatica/fisiologia , Animais , Neoplasias dos Ductos Biliares/etiologia , Neoplasias dos Ductos Biliares/parasitologia , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/parasitologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colangiocarcinoma/etiologia , Colangiocarcinoma/parasitologia , Colangiocarcinoma/patologia , Ciclofilina A/antagonistas & inibidores , Ciclofilina A/genética , Ciclofilina A/metabolismo , Fasciolíase/complicações , Fasciolíase/genética , Fasciolíase/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Nus , RNA Interferente Pequeno/farmacologia , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...