Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Respir Crit Care Med ; 204(4): 421-430, 2021 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-33848447

RESUMO

Rationale: Mechanical ventilation is a mainstay of intensive care but contributes to the mortality of patients through ventilator-induced lung injury. eCypA (extracellular CypA [cyclophilin A]) is an emerging inflammatory mediator and metalloproteinase inducer, and the gene responsible for its expression has recently been linked to coronavirus disease (COVID-19). Objectives: To explore the involvement of eCypA in the pathophysiology of ventilator-induced lung injury. Methods: Mice were ventilated with a low or high Vt for up to 3 hours, with or without blockade of eCypA signaling, and lung injury and inflammation were evaluated. Human primary alveolar epithelial cells were exposed to in vitro stretching to explore the cellular source of eCypA, and CypA concentrations were measured in BAL fluid from patients with acute respiratory distress syndrome to evaluate the clinical relevance. Measurements and Main Results: High-Vt ventilation in mice provoked a rapid increase in soluble CypA concentration in the alveolar space but not in plasma. In vivo ventilation and in vitro stretching experiments indicated the alveolar epithelium as the likely major source. In vivo blockade of eCypA signaling substantially attenuated physiological dysfunction, macrophage activation, and MMPs (matrix metalloproteinases). Finally, we found that patients with acute respiratory distress syndrome showed markedly elevated concentrations of eCypA within BAL fluid. Conclusions: CypA is upregulated within the lungs of injuriously ventilated mice (and critically ill patients), where it plays a significant role in lung injury. eCypA represents an exciting novel target for pharmacological intervention.


Assuntos
Anti-Inflamatórios/imunologia , Ciclofilina A/imunologia , Inflamação/imunologia , Respiração Artificial/efeitos adversos , Síndrome do Desconforto Respiratório/imunologia , Mucosa Respiratória/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia , Animais , COVID-19/genética , COVID-19/fisiopatologia , Células Cultivadas/efeitos dos fármacos , Ciclofilina A/farmacologia , Humanos , Inflamação/fisiopatologia , Masculino , Camundongos , Modelos Animais , Síndrome do Desconforto Respiratório/fisiopatologia , SARS-CoV-2 , Lesão Pulmonar Induzida por Ventilação Mecânica/genética
3.
Elife ; 92020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32539931

RESUMO

Counteracting innate immunity is essential for successful viral replication. Host cyclophilins (Cyps) have been implicated in viral evasion of host antiviral responses, although the mechanisms are still unclear. Here, we show that hepatitis C virus (HCV) co-opts the host protein CypA to aid evasion of antiviral responses dependent on the effector protein kinase R (PKR). Pharmacological inhibition of CypA rescues PKR from antagonism by HCV NS5A, leading to activation of an interferon regulatory factor-1 (IRF1)-driven cell intrinsic antiviral program that inhibits viral replication. These findings further the understanding of the complexity of Cyp-virus interactions, provide mechanistic insight into the remarkably broad antiviral spectrum of Cyp inhibitors, and uncover novel aspects of PKR activity and regulation. Collectively, our study identifies a novel antiviral mechanism that harnesses cellular antiviral immunity to suppress viral replication.


Assuntos
Ciclofilina A/antagonistas & inibidores , Hepacivirus/fisiologia , Fator Regulador 1 de Interferon/imunologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , eIF-2 Quinase/genética , Linhagem Celular Tumoral , Ciclofilina A/imunologia , Humanos , eIF-2 Quinase/imunologia
4.
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
5.
Fish Shellfish Immunol ; 98: 728-737, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31740398

RESUMO

As a ubiquitously expressed protein, cyclophilin A (CypA) is involved in a variety of pathological process, including immune suppression, inflammation, cell apoptosis, viral infection and stress response. However, the functional roles of CypA were largely unknown in economic marine animals. In this report, a novel CypA gene from sea cucumber Apostichopus japonicus (designated as AjCypA) was cloned and its function roles in immune responses were explored. The full-length cDNA of AjCypA was 1297 bp containing an open reading frame of 489 bp encoding a putative protein of 162 amino acids (aa). A conserved cyclophilin-like domain (CLD) with PPIase signature was located from 5 to 155 aa sequences in AjCypA, in which five necessary aa residues was totally conserved. In healthy sea cucumbers, AjCypA was expressed in all detected tissues, with highly expressed in muscles and weakly expressed in coelomocytes. AjCypA transcripts was significantly induced 8.08-fold and 5.65-fold in coelomocytes when sea cucumbers challenged with Vibrio splendidus in vivo and LPS in vitro, respectively. The expression pattern is similar with the expression of AjRel in the same condition. Moreover, GST pull-down and immunofluorescence analysis both revealed that AjCypA might be interacted with AjRel. Furthermore, AjCypA knockdown not only inhibited the expression of inflammation cytokines, but also suppressed the translocation of AjRel in nucleus induced by LPS. Taken together, our results suggested that AjCypA play key roles in V. splendidus mediated immune responses via suppressing the nuclear translocation of AjRel activity in sea cucumber.


Assuntos
Ciclofilina A/genética , Ciclofilina A/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , NF-kappa B/metabolismo , Sistemas de Translocação de Proteínas/metabolismo , Stichopus/genética , Stichopus/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Ciclofilina A/química , Perfilação da Expressão Gênica , Lipopolissacarídeos/farmacologia , Filogenia , Alinhamento de Sequência , Stichopus/enzimologia , Vibrio/fisiologia
6.
Sci Rep ; 9(1): 15027, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31636292

RESUMO

Olive pollen is a major cause of immunoglobulin E (IgE)-mediated allergy in Mediterranean countries. It is expected to become a worldwide leading allergenic source because olive cultivation is increasing in many countries. Ole e 15 belongs to the cyclophilin pan-allergen family, which includes highly cross-reactive allergens from non-related plant, animal and mold species. Here, the amino acid differences between Ole e 15 and its weak cross-reactive human homolog PPIA were grafted onto Ole e 15 to assess the contribution of specific surface areas to the IgE-binding. Eight Ole e 15-PPIA chimeras were produced in E. coli, purified and tested with 20 sera from Ole e 15-sensitized patients with olive pollen allergy by ELISA experiments. The contribution of linear epitopes was analyzed using twelve overlapping peptides spanning the entire Ole e 15 sequence. All the patients displayed a diverse reduction of the IgE-reactivity to the chimeras, revealing a highly polyclonal and patient-specific response to Ole e 15. IgE-epitopes are distributed across the entire Ole e 15 surface. Two main surface areas containing relevant conformational epitopes have been characterized. This is the first study to identify important IgE-binding regions on the surface of an allergenic cyclophilin.


Assuntos
Antígenos de Plantas/imunologia , Ciclofilina A/imunologia , Imunoglobulina E/imunologia , Olea/imunologia , Proteínas Recombinantes/imunologia , Rinite Alérgica Sazonal/imunologia , Sequência de Aminoácidos , Aminoácidos/genética , Antígenos de Plantas/química , Reações Cruzadas , Ciclofilina A/química , Humanos , Imunoglobulina E/sangue , Imunoglobulina G/metabolismo , Modelos Moleculares , Mutação/genética , Peptídeos/metabolismo , Ligação Proteica , Proteínas Recombinantes/genética , Rinite Alérgica Sazonal/sangue
7.
Sci Rep ; 9(1): 11263, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31375773

RESUMO

Evolution of cellular innate immune genes in response to viral threats represents a rich area of study for understanding complex events that shape mammalian genomes. One of these genes, TRIM5, is a retroviral restriction factor that mediates a post-entry block to infection. Previous studies on the genomic cluster that contains TRIM5 identified different patterns of gene amplification and the independent birth of CypA gene fusions in various primate species. However, the evolution of Trim5 in the largest order of mammals, Rodentia, remains poorly characterized. Here, we present an expansive phylogenetic and genomic analysis of the Trim5 cluster in rodents. Our findings reveal substantial evolutionary changes including gene amplifications, rearrangements, loss and fusion. We describe the first independent evolution of TrimCyp fusion genes in rodents. We show that the TrimCyp gene found in some Peromyscus species was acquired about 2 million years ago. When ectopically expressed, the P. maniculatus TRIMCyp shows anti-retroviral activity that is reversed by cyclosporine, but it does not activate Nf-κB or AP-1 promoters, unlike the primate TRIMCyps. These results describe a complex pattern of differential gene amplification in the Trim5 cluster of rodents and identify the first functional TrimCyp fusion gene outside of primates and tree shrews.


Assuntos
Ciclofilina A/genética , Evolução Molecular , Fusão Gênica/imunologia , Família Multigênica , Peromyscus/genética , Proteínas com Motivo Tripartido/genética , Animais , Linhagem Celular , Ciclofilina A/imunologia , Amplificação de Genes/imunologia , Genômica , HIV-1/imunologia , Humanos , Imunidade Inata/genética , Peromyscus/imunologia , Filogenia , Alinhamento de Sequência , Proteínas com Motivo Tripartido/imunologia
8.
Fish Shellfish Immunol ; 92: 172-180, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31176008

RESUMO

Cyclophilin A (CypA) is a ubiquitously expressed cellular protein and involves in diverse pathological conditions, including infection and inflammation. CypA acts as a key factor in the replication of several viruses. However, little is known about the role of CypA in the replication of the red-spotted grouper nervous necrosis virus (RGNNV). In the present report, grouper CypA (GF-CypA) was cloned from the grouper fin cell line (GF-1) derived from orange-spotted grouper (Epinephelus coioides). Sequence analysis found that GF-CypA open reading frame (ORF) of 495 bp encodes a polypeptide of 164 amino acids residues with a molecular weight of 17.4 kDa. The deduced amino acid sequence shared highly conserved regions with CypA of other animal species, showing that GF-CypA is a new member of Cyclophilin A family. We observed that GF-CypA was up-regulated in the GF-1 cells infected with RGNNV. Additionally, overexpression of CypA could significantly inhibit the replication of RGNNV in GF-1 cells. By contrast, when the GF-CypA was knock-downed by siRNA in GF-1 cells, the replication of RGNNV was enhanced. Furthermore, the expressions of pro-inflammatory factors, such as TNF-2, TNF-α, IL-1b, and ISG-15, were increased in GF-CypA transfected GF-1 cells challenged with RGNNV, indicating that GF-CypA might be involved in the regulation of the host pro-inflammatory factors. Altogether, we conclude that GF-CypA plays a vital role in the inhibitory effect of RGNNV replication that might be modulating the cytokines secretion in GF-1 cells during RGNNV infection. These results will shed new light on the function of CypA in the replication of RGNNV and will pave a new way for the prevention of the infection of RGNNV in fish.


Assuntos
Bass/genética , Bass/imunologia , Ciclofilina A/genética , Ciclofilina A/imunologia , Doenças dos Peixes/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Ciclofilina A/química , Proteínas de Peixes/química , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Perfilação da Expressão Gênica/veterinária , Nodaviridae/fisiologia , Filogenia , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/veterinária , Alinhamento de Sequência/veterinária , Replicação Viral
9.
Mol Immunol ; 111: 172-181, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31063938

RESUMO

Great efforts have been made to identify promising antigens and vaccine formulations against schistosomiasis. Among the previously described Schistosoma vaccine candidates, cyclophilins comprise an interesting antigen that could be used for vaccine formulations. Cyclophilin A is the target for the cyclosporine A, a drug with schistosomicide activity, and its orthologue from Schistosoma japonicum induces a protective immune response in mice. Although Schistosoma mansoni cyclophilin A also represents a promising target for anti-schistosome vaccines, its potential to induce protection has not been evaluated. In this study, we characterized the cyclophilin A (SmCyp), initially described as Smp17.7, analyzed its allergenic potential using in vitro functional assays, and evaluated its ability to induce protection in mice when administered as an antigen using different vaccine formulations and strategies. Results indicated that SmCyp could be successfully expressed by mammalian cells and bacteria. The recombinant protein did not promote IgE-reporter system activation in vitro, demonstrating its probable safety for use in vaccine formulations. T and B-cell epitopes were predicted in the SmCyp sequence, with two of them located within the active isomerase site. The most immunogenic antigen, SmCyp (107-121), was then used for immunization protocols. Immunization with the SmCyp gene or protein failed to reduce parasite burden but induced an immune response that modulated the granuloma area. In contrast, immunization with the synthetic peptide SmCyp (107-121) significantly reduced worm burden (48-50%) in comparison to control group, but did not regulate liver pathology. Moreover, the protection observed in mice immunized with the synthetic peptide was associated with the significant production of antibodies against the SmCyp (107-121) epitope. Therefore, in this study, we identified an epitope within the SmCyp sequence that induces a protective immune response against the parasite, thus representing a promising antigen that could be used for vaccine formulation against schistosomiasis.


Assuntos
Ciclofilina A/imunologia , Epitopos/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/imunologia , Feminino , Proteínas de Helminto/imunologia , Imunização/métodos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/imunologia , Vacinação/métodos , Vacinas/imunologia
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(8): 746-751, 2018 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-30384873

RESUMO

Objective To prepare monoclonal antibodies against human cyclophilin A (CypA) and explore the role of CypA in the occurrence and development of colon cancer. Methods According to the CypA gene sequence, the recombinant protein of CypA was expressed by the prokaryotic expression vector and purified by a nickel affinity chromatography column. The monoclonal antibody was prepared by the antigen which is purified protein immunizing mice and hybridoma technique. Utilizing CypA monoclonal antibody , immunohistochemistry was used to detect the difference expression between colorectal cancer and adjacent tissues, meanwhile, analyse clinicopathological parameters. Results The prokaryotic expression vector of CypA was successfully constructed, so we obtain the purified protein; One hybridoma cell line secreting anti-CypA antibody was obtained. Immunohistochemical results showed that CypA expressed high level in colorectal cancer and relate with differentiation degree and lymph node metastasis. Conclusion The monoclonal antibody against human CypA was successfully produced in mice, and CypA was overexpressed in colorectal cancer tissues, which was positively correlated with low differentiation and lymph node metastasis.


Assuntos
Ciclofilina A/imunologia , Animais , Anticorpos Monoclonais , Humanos , Hibridomas , Metástase Linfática , Camundongos , Proteínas Recombinantes
11.
Antivir Chem Chemother ; 26: 2040206618811413, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30449131

RESUMO

BACKGROUND: Viruses are obligate parasites that depend on the cellular machinery of the host to regenerate and manufacture their proteins. Most antiviral drugs on the market today target viral proteins. However, the more recent strategies involve targeting the host cell proteins or pathways that mediate viral replication. This new approach would be effective for most viruses while minimizing drug resistance and toxicity. METHODS: Cytomegalovirus replication, latency, and immune response are mediated by the intermediate early protein 2, the main protein that determines the effectiveness of drugs in cytomegalovirus inhibition. This review explains how intermediate early protein 2 can modify the action of cyclosporin A, an immunosuppressive, and antiviral drug. It also links all the pathways mediated by cyclosporin A, cytomegalovirus replication, and its encoded proteins. RESULTS: Intermediate early protein 2 can influence the cellular cyclophilin A pathway, affecting cyclosporin A as a mediator of viral replication or anti-cytomegalovirus drug. CONCLUSION: Cyclosporin A has a dual function in cytomegalovirus pathogenesis. It has the immunosuppressive effect that establishes virus replication through the inhibition of T-cell function. It also has an anti-cytomegalovirus effect mediated by intermediate early protein 2. Both of these functions involve cyclophilin A pathway.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Ciclofilina A/imunologia , Ciclofilina A/metabolismo , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Antivirais/química , Ciclofilina A/antagonistas & inibidores , Infecções por Citomegalovirus/virologia , Relação Dose-Resposta a Droga , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade
12.
Int Immunopharmacol ; 59: 347-353, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29680745

RESUMO

Cyclophilin A (CyPA), ubiquitously existing in cytoplasm of all eukaryotes, can be secreted in response to inflammatory stimuli. Extracellular CyPA plays a prominent role in the pathological processes of inflammatory diseases, acting as a proinflammatory mediator, exerting chemotactic effects, promoting apoptosis of endothelial cells and amplifying ROS generation, thus being considered as a potential treatment target of sepsis, a systemic inflammatory response syndrome. Our previous study found that antibodies against cyclophilin A of Clonorchis sinensis (CsCyPA) could neutralize mouse cyclophilin A (MuCyPA). In this study, we explored whether CsCyPA immunization could prevent or ameliorate mice sepsis induced by cecum ligation puncture (CLP). The results showed that CsCyPA immunization could improve the 72 h survival rate of mice after CLP. Moreover, the protective effect presented in a titer-dependent manner. The levels of cytokine IL-1ß, IL-6, TNF-α, MCP-1 and AST in serum were remarkably decreased compared to CLP control group mice. Pathological damages of liver, lung and kidney were ameliorated accompanied by less inflammatory cell infiltration. CFU per whole peripheral blood at 12 h and 24 h after CLP surgery was significantly lower than that of CLP control group. In vitro, intracellular ROS generation and cytokine mRNA expression in peritoneal macrophages stimulated by LPS were reduced obviously with anti-CsCyPA antibodies (anti-CsCyPAs) preincubation. All these results demonstrated that CsCyPA immunization protected mice from CLP induced sepsis.


Assuntos
Clonorchis sinensis , Ciclofilina A/imunologia , Sepse/terapia , Alanina Transaminase/sangue , Animais , Anticorpos/farmacologia , Aspartato Aminotransferases/sangue , Carga Bacteriana , Ceco/cirurgia , Citocinas/sangue , Citocinas/genética , Imunização , Rim/patologia , Ligadura , Fígado/patologia , Pulmão/patologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Sepse/sangue , Sepse/microbiologia , Sepse/patologia
13.
Sci China Life Sci ; 61(8): 954-965, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29705873

RESUMO

TRIM5α restricts retroviruses in a species-specific manner. Cyclophilin A was independently retrotransposed into the TRIM5 loci in different species, leading to the generation of antiviral TRIM5-cyclophilin A (TRIMCyp) proteins. Previously, we found that assam macaques express a TRIMCyp chimera (amTRIMCyp), along with a TRIM5α allelic protein (amTRIM5α). Herein, we investigated the antiviral activity of amTRIMCyp and amTRIM5α individually, as well as their interaction and joint effects. amTRIMCyp showed a divergent restriction pattern from amTRIM5α. Although both proteins potently restricted the replication of HIV-1, only amTRIM5α inhibited N-MLV. Remarkably, cellular anti-HIV-1 activity increased when amTRIMCyp and amTRIM5α were coexpressed, indicating a synergistic block of HIV-1 replication. Consistently, PMBCs from heterozygous amTRIM5α/TRIMCyp showed stronger resistance to HIV-1 infection than those from amTRIM5α/TRIM5α homozygotes. The anti-HIV-1 synergistic effect was dependent on the amTRIMCyp-amTRIM5α interaction. In contrast, amTRIMCyp completely abrogated the anti-N-MLV activity mediated by amTRIM5α, showing a dominant-negative effect, indicating that the generation of amTRIMCyp was involved in the trade-off between divergent restriction activities. Our results provide a new paradigm to study functional trade-offs mediated by allelic proteins, a theoretical basis for utilizing animal models with various TRIM5 alleles, as well as novel HIV-1 gene therapy strategies.


Assuntos
HIV-1/imunologia , Vírus da Leucemia Murina/imunologia , Macaca/imunologia , Proteínas Mutantes Quiméricas/imunologia , Infecções por Retroviridae/imunologia , Animais , Gatos , Linhagem Celular , Ciclofilina A/genética , Ciclofilina A/imunologia , Ciclofilina A/metabolismo , Expressão Gênica/imunologia , Células HEK293 , HIV-1/fisiologia , Humanos , Vírus da Leucemia Murina/fisiologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Macaca/virologia , Camundongos , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Proteínas/genética , Proteínas/imunologia , Proteínas/metabolismo , Interferência de RNA/imunologia , Infecções por Retroviridae/virologia , Ubiquitina-Proteína Ligases
14.
Sheng Wu Gong Cheng Xue Bao ; 34(1): 90-101, 2018 Jan 25.
Artigo em Chinês | MEDLINE | ID: mdl-29380574

RESUMO

Cyclophilin A (CypA) is a member of peptidyl prolylisomerases (PPIase) family. CypA is best known as a ubiquitously distributed intracellular protein. It has also been shown to be secreted by cells in response to inflammatory stimuli and oxidative stress. Extracellular CypA (eCypA) interacts with CD147 to initiate inflammatory responses via recruiting leucocytes into inflamed tissue. Recombinant CypA was expressed in Escherichia coli and then purified using Superdex 75™ 16/60. The results of Real-time PCR and ELISA showed that the expression levels of proinflammatory cytokines, such as IL-1ß, secreted by eCypA stimulated BMDM were significantly up-regulated, indicating that eCypA played an important role in promoting inflammatory responses. In addition, anti-CypA antibody was prepared using purified CypA protein for therapeutic evaluation in a mouse model of LPS-induced acute lung inflammation. Antibody-treated mice showed reduced lung injury and the expression levels of IL-1ß in the lung tissue and blood were decreased significantly, indicating that anti-CypA antibody exerted a potent anti-inflammatory activity. Our findings provide a potential therapeutic antibody for inflammation-mediated diseases.


Assuntos
Anti-Inflamatórios/farmacologia , Anticorpos/imunologia , Ciclofilina A/farmacologia , Inflamação/tratamento farmacológico , Pulmão/efeitos dos fármacos , Animais , Ciclofilina A/imunologia , Citocinas/imunologia , Leucócitos/imunologia , Pulmão/imunologia , Camundongos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia
15.
Fish Shellfish Immunol ; 72: 383-388, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29097323

RESUMO

Cyclophilin A (Cyp A) is the main intracellular receptor of cyclosporin A (CsA) belonging to the immunophilin family, which is known as an effective immunosuppressive drug. This study aimed to gain insights into the structure and biological function of cyclophilin A in the red swamp crayfish, Procambarus clarkii (PcCypA). We cloned PcCypA by homology cloning and anchored polymerase chain reaction (PCR), and assessed its mRNA and protein expression levels in different tissues using quantitative real-time PCR and western blot analysis, respectively. The full-length DNA contained a 5' untranslated region (UTR) comprising 108 base pairs (bp), an open reading frame of 495 bp encoding a polypeptide of 164 amino acids with an estimated molecular mass of 17.3 kDa, and a 3' UTR of 281 bp including a significant poly(A) plus tail sequence. The predicted amino acid sequence of PcCypA shared high identity with CypA in other organisms. PcCypA transcripts were detected in the hepatopancreas, gill, heart, muscle, testis, and ovary of crayfish, with the highest expression levels in the heart. Western blot analysis found one 17-kDa band in all of the tissues examined, except for the ovary. Molecular identification and expression analysis of PcCypA will facilitate further studies of the immune defense mechanisms in red swamp crayfish, and provide new insights into freshwater invertebrate immunology.


Assuntos
Astacoidea/genética , Astacoidea/imunologia , Ciclofilina A/genética , Ciclofilina A/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes/química , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/imunologia , Sequência de Bases , Ciclofilina A/química , Perfilação da Expressão Gênica , Filogenia , Alinhamento de Sequência
16.
PLoS Negl Trop Dis ; 11(10): e0006012, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29073139

RESUMO

The helminth Schistosoma mansoni modulates the infected host's immune system to facilitate its own survival, by producing excretory/secretory molecules that interact with a variety of the host's cell types including those of the immune system. Herein, we characterise the S. mansoni adult male worm secretome and identify 111 proteins, including 7 vaccine candidates and several molecules with potential immunomodulatory activity. Amongst the molecules present in the secretome, a 17-19kDa protein analogous to human cyclophilin A was identified. Given the ability of cyclophilin A to modulate the immune system by regulating antigen presenting cell activity, we sought to determine whether recombinant S. mansoni Cyclophilin A (rSmCypA) is capable of modulating bone-marrow derived dendritic cell (BMDC) and T cell responses under in vitro conditions. rSmCypA was enzymatically active and able to alter the pro-inflammatory cytokine profile of LPS-activated dendritic cells. rSmCypA also modulated DC function in the induction of CD4+ T cell proliferation with a preferential expansion of Treg cells. This work demonstrates the unique protein composition of the S. mansoni male worm secretome and immunomodulatory activity of S. mansoni Cyclophilin A.


Assuntos
Ciclofilina A/imunologia , Proteínas de Helminto/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/parasitologia , Animais , Ciclofilina A/genética , Células Dendríticas/imunologia , Feminino , Proteínas de Helminto/genética , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico , Schistosoma mansoni/genética , Esquistossomose mansoni/imunologia , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia
17.
J Leukoc Biol ; 102(4): 989-992, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28729360

RESUMO

Cyclophilin A (CypA), a peptidylprolyl cis-trans isomerase, is a ubiquitous and multifunctional protein. In addition to its role as a host-cell receptor for cyclosporine A, CypA has diverse functions in inflammatory conditions and diseases. CypA secreted in response to inflammatory stimuli binds to the cell surface via its receptor CD147 and induces secretion of various inflammatory cytokines. However, silencing and inhibition of either CypA or CD147 inhibits inflammatory cytokine expression and inflammation. This report reviews the literature related to the mechanism of CypA-dependent cytokine secretion and discusses this factor as a possible therapeutic target in inflammatory diseases.


Assuntos
Basigina/imunologia , Ciclofilina A/imunologia , Citocinas/imunologia , Animais , Humanos , Inflamação/imunologia , Inflamação/patologia
18.
Semin Thromb Hemost ; 42(3): 205-14, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26886394

RESUMO

Platelet-derived mediators, either in an autocrine or paracrine mode of action, regulate systemic and vascular inflammation as well as contribute to innate immune defense and also to regenerative mechanisms. This review reevaluates the impact of inflammatory mediators such as CXC-chemokine-Ligands, their receptors in modulating platelet functions and platelet survival, thereby influencing inflammatory or regenerative processes. We further explore the contribution of cyclophilin A and C-reactive protein in regulating thrombotic and hemostatic attributes of platelets. Moreover, we emphasize on the role of platelets as active components bridging the innate and adaptive immune responses, toll-like receptors on platelets, platelet interactions with the complement system, and platelet-derived thrombocidins exhibiting direct antimicrobial properties. As highlighted in this review, the multifaceted aspects of platelets and platelet-derived factors encourage further investigations in this intriguing and expansive but largely uncharted area of research in platelet biology.


Assuntos
Plaquetas/imunologia , Mediadores da Inflamação/imunologia , Inflamação/imunologia , Trombose/imunologia , Plaquetas/metabolismo , Proteína C-Reativa/imunologia , Proteína C-Reativa/metabolismo , Ciclofilina A/imunologia , Ciclofilina A/metabolismo , Hemostasia/imunologia , Humanos , Imunidade/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Modelos Imunológicos , Trombose/metabolismo
19.
Hum Vaccin Immunother ; 12(2): 545-53, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26305669

RESUMO

Previous research has shown that host Cyclophilin A (CyPA) can promote dendritic cell maturation and the subsequent innate immune response when incorporated into an HIV-1 Gag protein to circumvent the resistance of dendritic cells to HIV-1 infection. This led us to hypothesize that CyPA may improve HIV-1 Gag-specific vaccine immunogenicity via binding with Gag antigen. The adjuvant effect of CyPA was evaluated using a DNA vaccine with single or dual expression cassettes. Mouse studies indicated that CyPA specifically and markedly promoted HIV-1 Gag-specific cellular immunity but not an HIV-1 Env-specific cellular response. The Gag/CyPA dual expression cassettes stimulated a greater Gag-specific cellular immune response, than Gag immunization alone. Furthermore, CyPA induced a broad Gag-specific T cell response and strong cellular immunity that lasted up to 5 months. In addition, CyPA skewed to cellular rather than humoral immunity. To investigate the mechanisms of the adjuvant effect, site-directed mutagenesis in CyPA, including active site residues H54Q and F60A resulted in mutants that were co-expressed with Gag in dual cassettes. The immune response to this vaccine was analyzed in vivo. Interestingly, the wild type CyPA markedly increased Gag cellular immunity, but the H54Q and F60A mutants drastically reduced CyPA adjuvant activation. Therefore, we suggest that the adjuvant effect of CyPA was based on Gag-CyPA-specific interactions. Herein, we report that Cyclophilin A can augment HIV-1 Gag-specific cellular immunity as a genetic adjuvant in multiplex DNA immunization strategies, and that activity of this adjuvant is specific, broad, long-term, and based on Gag-CyPA interaction.


Assuntos
Vacinas contra a AIDS/imunologia , Adjuvantes Imunológicos , Ciclofilina A/imunologia , Imunidade Celular/imunologia , Vacinas de DNA/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C
20.
Tuberculosis (Edinb) ; 95(6): 736-743, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26546406

RESUMO

One third of the world's population is estimated to harbour latent tuberculosis infection (LTBI). Around 10% of them have the life time risk of developing active tuberculosis (PTB). Currently there is no gold standard test for identifying LTBI. Therefore identification of specific markers for LTBI will help as to develop a test specific for LTBI. Earlier, in our immunoproteomic analysis, we found that peptidyl-prolyl cis-trans isomerase A (PpiA) protein-containing fractions induced significantly higher interferon-gamma (IFN-γ) response in LTBI than in PTB. Immunological characterisation of recombinant PpiA protein was carried out in the current study. We have studied 10 cytokines and 2 chemokine responses against PpiA and standard antigens such as early secretory antigenic target-6 (ESAT-6) and culture filtrate antigen-10 (CFP-10). In healthy household contacts (HHC), all the tested antigens induced significantly higher levels of IFN-γ and Interlukin-8 (IL-8) compared with those in PTB. PpiA-specific IL-12p40 response was significantly increased in HHC compared with that in PTB. PpiA antigen-specific IFN-γ and IL-12p40 both showed 86% positivity in HHC, whereas in PTB, they showed 20% and 38% positivity, respectively. In terms of IFN-γ/TNF-α ratio, PpiA displayed 86% (30/35) positivity in HHC and 18% (7/39) positivity in PTB. In summary we found that PpiA-specific IFN-γ and IFN-γ/TNF-α ratio response were specific biomarkers for LTBI identification.


Assuntos
Proteínas de Bactérias/imunologia , Ciclofilina A/imunologia , Tuberculose Latente/diagnóstico , Tuberculose Latente/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose Pulmonar/diagnóstico , Tuberculose Pulmonar/imunologia , Biomarcadores/sangue , Estudos de Casos e Controles , Quimiocina CCL8/sangue , Quimiocina CCL8/imunologia , Ciclofilina A/genética , Humanos , Epitopos Imunodominantes , Interferon gama/sangue , Interferon gama/imunologia , Testes de Liberação de Interferon-gama , Interleucina-10/sangue , Interleucina-10/imunologia , Subunidade p40 da Interleucina-12/sangue , Subunidade p40 da Interleucina-12/imunologia , Interleucina-6/sangue , Interleucina-6/imunologia , Tuberculose Latente/sangue , Tuberculose Latente/microbiologia , Mycobacterium tuberculosis/enzimologia , Valor Preditivo dos Testes , Prognóstico , Tuberculose Pulmonar/sangue , Tuberculose Pulmonar/microbiologia , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...