Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 325(6): C1401-C1414, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37842750

RESUMO

Open heart surgery is often an unavoidable procedure for the treatment of coronary artery disease. The procedure-associated reperfusion injury affects postoperative cardiac performance and long-term outcomes. We addressed here whether cardioplegia essential for cardiopulmonary bypass surgery activates Nrf2, a transcription factor regulating the expression of antioxidant and detoxification genes. With commonly used cardioplegic solutions, high K+, low K+, Del Nido (DN), histidine-tryptophan-ketoglutarate (HTK), and Celsior (CS), we found that DN caused a significant increase of Nrf2 protein in AC16 human cardiomyocytes. Tracing the ingredients in DN led to the discovery of KCl at the concentration of 20-60 mM capable of significant Nrf2 protein induction. The antioxidant response element (ARE) luciferase reporter assays confirmed Nrf2 activation by DN or KCl. Transcriptomic profiling using RNA-seq revealed that oxidation-reduction as a main gene ontology group affected by KCl. KCl indeed elevated the expression of classical Nrf2 downstream targets, including TXNRD1, AKR1C, AKR1B1, SRXN1, and G6PD. DN or KCl-induced Nrf2 elevation is Ca2+ concentration dependent. We found that KCl decreased Nrf2 protein ubiquitination and extended the half-life of Nrf2 from 17.8 to 25.1 mins. Knocking out Keap1 blocked Nrf2 induction by K+. Nrf2 induction by DN or KCl correlates with the protection against reactive oxygen species generation or loss of viability by H2O2 treatment. Our data support that high K+ concentration in DN cardioplegic solution can induce Nrf2 protein and protect cardiomyocytes against oxidative damage.NEW & NOTEWORTHY Open heart surgery is often an unavoidable procedure for the treatment of coronary artery disease. The procedure-associated reperfusion injury affects postoperative cardiac performance and long-term outcomes. We report here that Del Nido cardioplegic solution or potassium is an effective inducer of Nrf2 transcription factor, which controls the antioxidant and detoxification response. This indicates that Del Nido solution is not only essential for open heart surgery but also exhibits cardiac protective activity.


Assuntos
Doença da Artéria Coronariana , Traumatismo por Reperfusão , Humanos , Soluções Cardioplégicas/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch , Fator 2 Relacionado a NF-E2/genética , Miócitos Cardíacos , Potássio , Antioxidantes/farmacologia , Peróxido de Hidrogênio/farmacologia , Parada Cardíaca Induzida/métodos , Estresse Oxidativo , Aldeído Redutase
2.
Cells ; 12(2)2023 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-36672226

RESUMO

The Nrf2 gene encodes a transcription factor best known for regulating the expression of antioxidant and detoxification genes. A long list of small molecules has been reported to induce Nrf2 protein via Keap1 oxidation or alkylation. Many of these Nrf2 inducers exhibit off-target or toxic effects due to their nature as electrophiles. In searching for non-toxic Nrf2 inducers, we found that a culture medium change to fresh DMEM is capable of inducing Nrf2 protein in HeLa, HEK293, AC16 and MCF7 cells. Testing the components of DMEM led to the discovery of L-Cystine as an effective Nrf2 inducer. L-Cystine induces a dose-dependent increase of Nrf2 protein, from 0.1 to 1.6 mM. RNA-seq analyses and RT-PCR revealed an induction of multiple Nrf2 downstream genes, including NQO1, HMOX1, GCLC, GCLM, SRXN1, TXNRD1, AKR1C and OSGIN1 by 0.8 mM L-Cystine. The induction of Nrf2 protein was dependent on L-Cystine entering cells via the cystine/glutamate antiporter and the presence of Keap1. The half-life of Nrf2 protein increased from 19.4 min to 30.9 min with 0.8 mM L-Cystine treatment. L-Cystine was capable of eliciting cytoprotection by reducing ROS generation and protecting against oxidant- or doxorubicin-induced apoptosis. As an amino acid derivative, L-Cystine is considered a non-toxic Nrf2 inducer that exhibits the potential for protection against oxidative stress and tissue injury.


Assuntos
Cistina , Fator 2 Relacionado a NF-E2 , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Cistina/farmacologia , Cistina/metabolismo , Citoproteção , Células HEK293 , Técnicas de Cultura de Células
3.
Mol Oncol ; 16(8): 1714-1727, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35184380

RESUMO

The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is often highly expressed in non-small cell lung cancer (NSCLC). Through its target genes, NRF2 enhances cancer progression and chemo/radioresistance, leading to a poorer prognosis in patients with high NRF2 expression. In this study, we identified CHM-like Rab escort protein (CHML; encoding Rep2) as an NRF2 target gene with an antioxidant response element (ARE) in its promoter region (-1622 to -1612). Analysis of patient data curated by The Cancer Genome Atlas (TCGA) and Oncomine databases revealed that CHML mRNA expression was elevated in lung adenocarcinoma (LUAD) patient tumor tissues and correlated with decreased patient survival. Immunohistochemistry (IHC) analysis of normal versus lung cancer patient tissues revealed that Rep2 protein levels were higher in lung tumors compared with normal tissue, which also correlated with increased levels of NRF2. Importantly, siRNA-mediated knockdown of CHML/Rep2 in A549 NSCLC cells decreased their ability to proliferate. Mechanistically, Rep2 mediates mTOR function, as loss of Rep2 inhibited, whereas overexpression enhanced, mTOR translocation and activation at the lysosome. Our findings identify a novel NRF2-Rep2-dependent regulation of mTOR function.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Ácidos Graxos Insaturados , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
4.
Redox Biol ; 41: 101906, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33676361

RESUMO

Oxidative stress is ubiquitously involved in disease etiology or progression. While the damaging effects have been well characterized, how cells deal with oxidative stress for prevention or removal of damage remains to be fully elucidated. Works from our laboratory have revealed de novo Nrf2 protein translation when cells are encountering low to mild levels of oxidative stress. Nrf2 encodes a transcription factor controlling a myriad of genes important for antioxidation, detoxification, wound repair and tissue remodeling. Here we report a role of FUBP1 in regulating de novo Nrf2 protein translation. An increase of FUBP1 binding to Nrf2 5'UTR due to H2O2 treatment has been found by LC-MS/MS, Far Western blot and ribonucleoprotein immunoprecipitation assays. Blocking FUBP1 expression using siRNA abolished H2O2 from inducing Nrf2 protein elevation or Nrf2 5'UTR activity. While no nuclear to cytoplasmic translocation was detected, cytosolic redistribution to the ribosomal fractions was observed due to oxidant treatment. The presence of FUBP1 in 40/43S ribosomal fractions confirm its involvement in translation initiation of Nrf2 protein. When tested by co-immunoprecipitation with eIF4E, eIF2a, eIF3η and eIF1, only eIF3η was found to gain physical interaction with FUBP1 due to H2O2 treatment. Our data support a role of FUBP1 for promoting the attachment of 40S ribosomal subunit to Nrf2 mRNA and formation of 43S pre-initiation complex for translation initiation of Nrf2 protein under oxidative stress.


Assuntos
Peróxido de Hidrogênio , Fator 2 Relacionado a NF-E2 , Proteínas de Transporte , Cromatografia Líquida , Proteínas de Ligação a DNA , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Proteínas de Ligação a RNA , Espectrometria de Massas em Tandem
5.
Front Immunol ; 8: 414, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28428789

RESUMO

With the rapid development of industry and farm, fungi contamination widely exists in occupational environment. Inhalation of fungi-contaminated organic dust results in hypersensitivity pneumonitis. 1,3-ß-Glucan is a major cell wall component of fungus and is considered as a biomarker of fungi exposure. Current studies showed that 1,3-ß-glucan exposure induced lung inflammation, which involved uncontrolled T helper (Th) cell immune responses, such as Th1, Th2, Th17, and regulatory T cell (Treg). A recently identified IL-10-producing B cells (B10) was reported in regulating immune homeostasis. However, its regulatory role in hypersensitivity pneumonitis is still subject to debate. In our study, we comprehensively investigated the role of B10 and the relationship between B10 and Treg in 1,3-ß-glucan-induced lung inflammation. Mice with insufficient B10 exhibited more inflammatory cells accumulation and severer pathological inflammatory changes. Insufficient B10 led to increasing Th1, Th2, and Th17 responses and restricted Treg function. Depletion of Treg before the onset of inflammation could suppress B10. Whereas, Treg depletion only at the late stage of inflammation failed to affect B10. Our study demonstrated that insufficient B10 aggravated the lung inflammation mediated by dynamic shifts in Th immune responses after 1,3-ß-glucan exposure. The regulatory function of B10 on Th immune responses might be associated with Treg and IL-10. Treg could only interact with B10 at an early stage.

6.
Sci Rep ; 6: 28911, 2016 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-27354007

RESUMO

Silicosis is characterized by chronic lung inflammation and fibrosis, which are seriously harmful to human health. Previous research demonstrated that uncontrolled T-helper (Th) cell immune responses were involved in the pathogenesis of silicosis. Lymphocytes also are reported to have important roles. Existing studies on lymphocyte regulation of Th immune responses were limited to T cells, such as the regulatory T (Treg) cell, which could negatively regulate inflammation and promote the process of silicosis. However, other regulatory subsets in silicosis have not been investigated in detail, and the mechanism of immune homeostasis modulation needs further exploration. Another regulatory lymphocyte, the regulatory B cell, has recently drawn increasing attention. In this study, we comprehensively showed the role of IL-10-producing regulatory B cell (B10) in a silicosis model of mice. B10 was inducible by silica instillation. Insufficient B10 amplified inflammation and attenuated lung fibrosis by promoting the Th1 immune response. Insufficient B10 clearly inhibited Treg and decreased the level of IL-10. Our study indicated that B10 could control lung inflammation and exacerbate lung fibrosis by inhibiting Th1 response and modulating the Th balance. The regulatory function of B10 could be associated with Treg induction and IL-10 secretion.


Assuntos
Linfócitos B Reguladores/fisiologia , Interleucina-10/metabolismo , Fibrose Pulmonar/imunologia , Células Th1/fisiologia , Animais , Feminino , Imunidade Celular , Linfonodos/patologia , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Dióxido de Silício , Células Th17/fisiologia
7.
Mediators Inflamm ; 2016: 6235614, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27069316

RESUMO

CD4(+) T cells play an important role in regulating silica-induced inflammation and fibrosis. Recent studies showed that Wnt/ß-catenin pathway could modulate the function and the differentiation of CD4(+) T cells. Therefore, Wnt/ß-catenin pathway may participate in the development and progress of silicosis. To investigate the role of Wnt/ß-catenin pathway, we used lentivirus expressing ß-catenin shRNA to block the Wnt/ß-catenin pathway by intratracheal instillation to the mice model of silicosis. Treatment of lentivirus could significantly aggravate the silica-induced lung inflammation and attenuated the fibrosis at the late stage. By analyzing CD4(+) T cells, we found that blockade of Wnt/ß-catenin pathway suppressed regulatory T cells (Tregs). Reciprocally, enhanced Th17 response was responsible for the further accumulation of neutrophils and production of proinflammatory cytokines. In addition, blockade of Wnt/ß-catenin pathway delayed the Th1/Th2 polarization by inhibiting Tregs and Th2 response. These results indicated that Wnt/ß-catenin pathway could regulate Tregs to modulate Th immune response, which finally altered the pathological character of silicosis. Our study suggested that Wnt/ß-catenin pathway might be a potential target to treat the silica-induced inflammation and fibrosis.


Assuntos
Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Linfócitos T Reguladores/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Dióxido de Silício , Linfócitos T Reguladores/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th1/metabolismo , Células Th17/efeitos dos fármacos , Células Th17/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/genética
8.
J Nat Prod ; 78(12): 3049-57, 2015 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-26605988

RESUMO

Silicosis is an inflammatory and fibrotic lung disease caused by inhalation of silica. Th17 cells play a key role in causing silica-induced lung inflammation and fibrosis. Baicalin, a compound isolated from the Chinese herb Huangqin, could suppress the differentiation of Th17 cells and alleviate inflammation. However, there are very few reports of the immunoregulatory mechanisms of baicalin in experimental silica-induced lung inflammation and fibrosis. In our study, mice were exposed to silica by intratracheal instillation, and in this way we established an experimental silicosis model. To elucidate the effects and mechanisms of baicalin in silica-induced inflammation and fibrosis, we used baicalin to treat the developed mouse model of silicosis. Treatment with baicalin attenuated the accumulation of inflammatory cells and led to milder pathological inflammatory and fibrotic changes in lung tissues. Baicalin affected the immunological balance between Th17 and Treg responses. Therefore, baicalin caused a decrease in Th17 cells by stimulating Treg cells and by inhibiting IL-6 and IL-23. We further demonstrated that silica-induced Th1 and Th2 immune responses were both inhibited by increased Treg activation, which was promoted by baicalin. Our findings confirmed the potential functions of baicalin in inhibiting the Th17 response and reducing silica-induced inflammation and fibrosis.


Assuntos
Flavonoides/farmacologia , Pulmão/fisiopatologia , Dióxido de Silício/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Flavonoides/química , Inflamação/induzido quimicamente , Interleucina-17/imunologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-23/antagonistas & inibidores , Interleucina-6/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Pneumonia , Reação em Cadeia da Polimerase , Scutellaria baicalensis/química , Dióxido de Silício/toxicidade , Silicose/fisiopatologia , Linfócitos T Reguladores/metabolismo , Células Th17/efeitos dos fármacos
9.
Int J Environ Res Public Health ; 12(9): 10739-54, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26340635

RESUMO

Silicosis is a form of occupational lung disease caused by inhalation of crystalline silica dust. While the pathogenesis of silicosis is not clearly understood, the Wnt/ß-catenin signaling pathway is thought to play a major role in lung fibrosis. To explore the role of Wnt/ß-catenin pathway in silicosis, we blocked Wnt/ß-catenin pathway both in silica-treated MLE-12 cells (a mouse pulmonary epithelial cell line) and in a mouse silicosis model by using a lentiviral vector expressing a short hairpin RNA silencing ß-catenin (Lv-shß-catenin). In vitro, Lv-shß-catenin significantly decreased the expression of ß-catenin, MMP2 and MMP9, and secretion of TGF-ß1. In vivo, intratracheal treatment with Lv-shß-catenin significantly reduced expression of ß-catenin in the lung and levels of TGF-ß1 in bronchoalveolar lavage fluid, and notably attenuated pulmonary fibrosis as evidenced by hydroxyproline content and collagen I\III synthesis in silica-administered mice. These results indicate that blockade of the Wnt/ß-catenin pathway can prevent the development of silica-induced lung fibrosis. Thus Wnt/ß-catenin pathway may be a target in prevention and treatment of silicosis.


Assuntos
Modelos Animais de Doenças , Lentivirus/genética , Fibrose Pulmonar/prevenção & controle , RNA Interferente Pequeno/genética , Silicose/prevenção & controle , Via de Sinalização Wnt/fisiologia , Animais , Líquido da Lavagem Broncoalveolar , Linhagem Celular , Inativação Gênica/fisiologia , Vetores Genéticos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Fibrose Pulmonar/metabolismo , Dióxido de Silício/toxicidade , Silicose/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , beta Catenina/metabolismo
10.
J Cell Mol Med ; 18(9): 1773-84, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25091058

RESUMO

Silicosis is an occupational lung disease caused by the inhalation of silica dust and characterized by lung inflammation and fibrosis. Interleukin (IL)-1ß is induced by silica and functions as the key pro-inflammatory cytokine in this process. The Th17 response, which is induced by IL-1ß, has been reported very important in chronic human lung inflammatory diseases. To elucidate the underlying mechanisms of IL-1ß and IL-17 in silicosis, we used anakinra and an anti-IL-17 monoclonal antibody (mAb) to block the receptor of IL-1ß (IL-RI) and IL-17, respectively, in a mouse model of silicosis. We observed increased IL-1ß expression and an enhanced Th17 response after silica instillation. Treatment with an IL-1 type I receptor (IL-1RI) antagonist anakinra substantially decreased silica-induced lung inflammation and the Th17 response. Lung inflammation and the accumulation of inflammatory cells were attenuated in the IL-17-neutralized silicosis group. IL-17 may promote lung inflammation by modulating the differentiation of Th1 and regulatory T cells (Tregs) and by regulating the production of IL-22 and IL-1ß during the lung inflammation of silicosis. Silica may induce IL-1ß production from alveolar macrophages and promote inflammation by initiating a Th17 response via an IL-1ß/IL-1RI-dependent mechanism. The Th17 response could induce lung inflammation during the pathogenesis of silicosis by regulating the homoeostasis of the Th immune responses and affecting the production of IL-22 and IL-1ß. This study describes a potentially important inflammatory mechanism of silicosis that may bring about novel therapies for this inflammatory and fibrotic disease.


Assuntos
Interleucina-1beta/fisiologia , Silicose/imunologia , Células Th17/imunologia , Animais , Células Cultivadas , Feminino , Humanos , Interleucina-17/fisiologia , Interleucinas/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos C57BL , Receptores Tipo I de Interleucina-1/metabolismo , Dióxido de Silício , Interleucina 22
11.
Mediators Inflamm ; 2014: 570894, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24692850

RESUMO

Silica inhalation can induce chronic lung inflammation and fibrosis. Upon silica stimulation, activated macrophages trigger the T-lymphocyte which can differentiate into many different types of Th cells, including the recently discovered Th17 cells. IL-17A, the typical Th17 cytokine, is reported in some inflammatory diseases. However, the role of IL-17A in silica-induced inflammatory response is still not clear. The regulatory mechanism of silica-induced Th17 response also needs to be investigated. So we established a mice primary cell coculture system (macrophage and lymphocyte) to investigate the role of IL-17A in silica-induced inflammatory response in vitro, by using anti-IL-17A mAb and IL-1Ra. Both anti-IL-17A mAb and IL-1Ra decreased the level of IL-17A and increased the function of Treg cells. The Th1 response was suppressed and the Th2 response was promoted by the addition of anti-IL-17A mAb or IL-1Ra. IL-1Ra treatment decreased the level of IL-6, whereas the levels of IL-23 and ROR- γ t were increased. Our study demonstrated that IL-17A reduction altered the pattern of silica-induced Th responses by boosting the function of Treg cells in vitro. Blocking the function of IL-1 signal pathway could suppress the level of IL-17A, which played the major role in modulating silica-induced Th responses in vitro.


Assuntos
Regulação da Expressão Gênica , Interleucina-17/fisiologia , Dióxido de Silício/química , Linfócitos T Reguladores/citologia , Células Th1/citologia , Células Th2/citologia , Animais , Anticorpos Monoclonais/imunologia , Líquido da Lavagem Broncoalveolar , Diferenciação Celular , Técnicas de Cocultura , Feminino , Inflamação/induzido quimicamente , Inflamação/imunologia , Interleucina-23/metabolismo , Linfócitos/citologia , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Transdução de Sinais
12.
Toxicol Appl Pharmacol ; 275(1): 62-72, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24291675

RESUMO

Silica exposure can cause lung inflammation and fibrosis, known as silicosis. Interleukin-17A (IL-17A) and Th17 cells play a pivotal role in controlling inflammatory diseases. However, the roles of IL-17A and Th17 cells in the progress of silica-induced inflammation and fibrosis are poorly understood. This study explored the effects of IL-17A on silica-induced inflammation and fibrosis. We used an anti-mouse IL-17A antibody to establish an IL-17A-neutralized mice model, and mice were exposed to silica to establish an experimental silicosis model. We showed that IL-17A neutralization delayed neutrophil accumulation and progression of silica-induced lung inflammation and fibrosis. IL-17A neutralization reduced the percentage of Th17 in CD4+ T cells, decreased IL-6 and IL-1ß expression, and increased Tregs at an early phase of silica-induced inflammation. Neutralization of IL-17A delayed silica-induced Th1/Th2 immune and autoimmune responses. These results suggest that IL-17A neutralization alleviates early stage silica-induced lung inflammation and delays progression of silica-induced lung inflammation and fibrosis. Neutralization of IL-17A suppressed Th17 cell development by decreasing IL-6 and/or IL-1ß and increased Tregs at an early phase of silica-induced inflammation. Neutralization of IL-17A also delayed the Th1/Th2 immune response during silica-induced lung inflammation and fibrosis. IL-17A may play a pivotal role in the early phase of silica-induced inflammation and may mediate the Th immune response to influence silica-induced lung inflammation and fibrosis in mice.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Modelos Animais de Doenças , Interleucina-17/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Pneumonia/prevenção & controle , Fibrose Pulmonar/prevenção & controle , Silicose/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Feminino , Interleucina-17/metabolismo , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/antagonistas & inibidores , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/efeitos dos fármacos , Pneumonia/etiologia , Fibrose Pulmonar/etiologia , Distribuição Aleatória , Silicose/imunologia , Silicose/patologia , Silicose/fisiopatologia , Organismos Livres de Patógenos Específicos , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia
13.
J Immunotoxicol ; 10(3): 253-61, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22963429

RESUMO

1,3-ß-glucan is considered a fungal biomarker and exposure to this agent can induce lung inflammation. Complement activation plays an important role in early immune responses to ß-glucan. Previous studies showed that T-regulatory cells (Tregs) regulated 1,3-ß-glucan-induced lung inflammation by modulating the maintenance of immune homeostasis in the lung. Both interleukin (IL)-17 and TH17 cells play pivotal roles in inflammation associated with lung disease and share reciprocal developmental pathways with Tregs. However, the effect of Tregs on IL-17 and TH17 responses in 1,3-ß-glucan-induced lung inflammation remains unclear. In this study, mice were exposed to 1,3-ß-glucan by intratracheal instillation. To investigate the effects of Tregs on IL-17 and TH17 cells in the induced lung inflammation, a Treg-depleted mice model was generated by administration of anti-CD25 mAb. The results indicated that Treg-depleted mice showed more severe pathological inflammatory changes in lung tissues. Tregs depletion reduced IL-17 expression in these tissues, and increased those of TH1 cytokines. The expression of IL-17 increased at the early phase of the inflammation response. There were no significant effects of the Tregs on expression of RORγt and IL-6 or the amount of CD4(+)IL-17(+) cells in the lungs. When taken together, the late phase of the 1,3-ß-glucan-induced inflammatory response in the mice was primarily mediated by TH1 cytokines rather than IL-17. In contrast, the early phase of the inflammatory response might be mediated in part by IL-17 along with activated complement. Tregs might be required for IL-17 expression during the late phase inflammatory response in mice. The increased IL-17 mRNA observed during the 1,3-ß-glucan induced inflammatory response were attributed to cells other than TH17 cells.


Assuntos
Regulação da Expressão Gênica/imunologia , Interleucina-17/imunologia , Pneumonia/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Zimosan/toxicidade , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-17/biossíntese , Depleção Linfocítica , Camundongos , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Pneumonia/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Células Th1/metabolismo , Células Th1/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA