Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Respir Res ; 23(1): 136, 2022 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-35643499

RESUMO

BACKGROUND: Pulmonary hypertension is a common and serious complication of chronic obstructive pulmonary disease (COPD). Studies suggest that cigarette smoke can initiate pulmonary vascular remodelling by stimulating cell proliferation; however, the underlying cause, particularly the role of vasoactive prostanoids, is unclear. We hypothesize that cigarette smoke extract (CSE) can induce imbalanced vasoactive prostanoid release by differentially modulating the expression of respective synthase genes in human pulmonary artery smooth muscle cells (PASMCs) and endothelial cells (PAECs), thereby contributing to cell proliferation. METHODS: Aqueous CSE was prepared from 3R4F research-grade cigarettes. Human PASMCs and PAECs were treated with or without CSE. Quantitative real-time RT-PCR and Western blotting were used to analyse the mRNA and protein expression of vasoactive prostanoid syhthases. Prostanoid concentration in the medium was measured using ELISA kits. Cell proliferation was assessed using the cell proliferation reagent WST-1. RESULTS: We demonstrated that CSE induced the expression of cyclooxygenase-2 (COX-2), the rate-limiting enzyme in prostanoid synthesis, in both cell types. In PASMCs, CSE reduced the downstream prostaglandin (PG) I synthase (PGIS) mRNA and protein expression and PGI2 production, whereas in PAECs, CSE downregulated PGIS mRNA expression, but PGIS protein was undetectable and CSE had no effect on PGI2 production. CSE increased thromboxane (TX) A synthase (TXAS) mRNA expression and TXA2 production, despite undetectable TXAS protein in both cell types. CSE also reduced microsomal PGE synthase-1 (mPGES-1) protein expression and PGE2 production in PASMCs, but increased PGE2 production despite unchanged mPGES-1 protein expression in PAECs. Furthermore, CSE stimulated proliferation of both cell types, which was significantly inhibited by the selective COX-2 inhibitor celecoxib, the PGI2 analogue beraprost and the TXA2 receptor antagonist daltroban. CONCLUSIONS: These findings provide the first evidence that cigarette smoke can induce imbalanced prostanoid mediator release characterized by the reduced PGI2/TXA2 ratio and contribute to pulmonary vascular remodelling and suggest that TXA2 may represent a novel therapeutic target for pulmonary hypertension in COPD.


Assuntos
Fumar Cigarros , Hipertensão Pulmonar , Doença Pulmonar Obstrutiva Crônica , Proliferação de Células , Células Endoteliais , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Prostaglandinas/metabolismo , Prostaglandinas E/metabolismo , Prostaglandinas E/farmacologia , Artéria Pulmonar/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , RNA Mensageiro/metabolismo , Nicotiana , Remodelação Vascular
2.
Am J Respir Cell Mol Biol ; 58(4): 449-460, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29053336

RESUMO

Selective repression of the antifibrotic gene CXCL10 contributes to tissue remodeling in idiopathic pulmonary fibrosis (IPF). We have previously reported that histone deacetylation and histone H3 lysine 9 (H3K9) methylation are involved in CXCL10 repression. In this study, we explored the role of H3K27 methylation and the interplay between the two histone lysine methyltransferases enhancer of zest homolog 2 (EZH2) and G9a in CXCL10 repression in IPF. By applying chromatin immunoprecipitation, Re-ChIP, and proximity ligation assays, we demonstrated that, like G9a-mediated H3K9 methylation, EZH2-mediated histone H3 lysine 27 trimethylation (H3K27me3) was significantly enriched at the CXCL10 promoter in fibroblasts from IPF lungs (F-IPF) compared with fibroblasts from nonfibrotic lungs, and we also found that EZH2 and G9a physically interacted with each other. EZH2 knockdown reduced not only EZH2 and H3K27me3 but also G9a and H3K9me3, and G9a knockdown reduced not only G9 and H3K9me3 but also EZH2 and H3K27me3. Depletion and inhibition of EZH2 and G9a also reversed histone deacetylation and restored CXCL10 expression in F-IPF. Furthermore, treatment of fibroblasts from nonfibrotic lungs with the profibrotic cytokine transforming growth factor-ß1 increased EZH2, G9a, H3K27me3, H3K9me3, and histone deacetylation at the CXCL10 promoter, similar to that observed in F-IPF, which was correlated with CXCL10 repression and was prevented by EZH2 and G9a knockdown. These findings suggest that a novel and functionally interdependent interplay between EZH2 and G9a regulates histone methylation-mediated epigenetic repression of the antifibrotic CXCL10 gene in IPF. This interdependent interplay may prove to be a target for epigenetic intervention to restore the expression of CXCL10 and other antifibrotic genes in IPF.


Assuntos
Quimiocina CXCL10/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Fibroblastos/enzimologia , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Fibrose Pulmonar Idiopática/enzimologia , Pulmão/enzimologia , Estudos de Casos e Controles , Células Cultivadas , Quimiocina CXCL10/genética , Metilação de DNA , Regulação para Baixo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Repressão Epigenética , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/genética , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Regiões Promotoras Genéticas , Transdução de Sinais , Fator de Crescimento Transformador beta1/farmacologia
3.
FASEB J ; 28(7): 3183-96, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24652950

RESUMO

Selective silencing of the cyclooxygenase-2 (COX-2) gene with the loss of the antifibrotic mediator prostaglandin E2 contributes to the fibrotic process in idiopathic pulmonary fibrosis (IPF). This study explored the role of G9a- and enhancer of zeste homolog 2 (EZH2)-mediated methylation of histone H3 lysine 9 (H3K9me3) and histone H3 lysine 27 (H3K27me3) in COX-2 silencing in IPF. Chromatin immunoprecipitation (ChIP) and re-ChIP assays demonstrated marked increases in H3K9me3, H3K27me3, and DNA methylation, together with their respective modifying enzymes G9a, EZH2, and DNA methyltransferases (Dnmts) and respective binding proteins heterochromatin protein 1 (HP1), polycomb protein complex 1 (PRC1) and methyl CpG binding protein 2 (MeCP2), at the COX-2 promoter in lung fibroblasts from patients with IPF (F-IPFs) compared with fibroblasts from nonfibrotic lungs. HP1, EZH2, and MeCP2 in turn were associated with additional repressive chromatin modifiers in F-IPFs. G9a and EZH2 inhibitors and small interfering RNAs and the Dnmt1 inhibitor markedly reduced H3K9me3 (49-79%), H3K27me3 (44-81%), and DNA methylation (61-97%) at the COX-2 promoter. These reductions were correlated with increased histone H3 and H4 acetylation, resulting in COX-2 mRNA and protein reexpression in F-IPFs. Our results support a central role for G9a- and EZH2-mediated histone hypermethylation and a model of bidirectional, mutually reinforcing, and interdependent crosstalk between histone hypermethylation and DNA methylation in COX-2 epigenetic silencing in IPF.-Coward, W. R., Feghali-Bostwick, C. A., Jenkins, G., Knox, A. J., Pang, L. A central role for G9a and EZH2 in the epigenetic silencing of cyclooxygenase-2 in idiopathic pulmonary fibrosis.


Assuntos
Ciclo-Oxigenase 2/genética , Epigênese Genética/genética , Inativação Gênica/fisiologia , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Acetilação , Adulto , Idoso , Células Cultivadas , Imunoprecipitação da Cromatina/métodos , Ciclo-Oxigenase 2/metabolismo , Metilação de DNA/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Fibroblastos/metabolismo , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Complexo Repressor Polycomb 2/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Adulto Jovem
4.
RNA ; 18(12): 2236-50, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23118416

RESUMO

Cordycepin (3' deoxyadenosine) has long been used in the study of in vitro assembled polyadenylation complexes, because it terminates the poly(A) tail and arrests the cleavage complex. It is derived from caterpillar fungi, which are highly prized in Chinese traditional medicine. Here we show that cordycepin specifically inhibits the induction of inflammatory mRNAs by cytokines in human airway smooth muscle cells without affecting the expression of control mRNAs. Cordycepin treatment results in shorter poly(A) tails, and a reduction in the efficiency of mRNA cleavage and transcription termination is observed, indicating that the effects of cordycepin on 3' processing in cells are similar to those described in in vitro reactions. For the CCL2 and CXCL1 mRNAs, the effects of cordycepin are post-transcriptional, with the mRNA disappearing during or immediately after nuclear export. In contrast, although the recruitment of RNA polymerase II to the IL8 promoter is also unaffected, the levels of nascent transcript are reduced, indicating a defect in transcription elongation. We show that a reporter construct with 3' sequences from a histone gene is unaffected by cordycepin, while CXCL1 sequences confer cordycepin sensitivity to the reporter, demonstrating that polyadenylation is indeed required for the effect of cordycepin on gene expression. In addition, treatment with another polyadenyation inhibitor and knockdown of poly(A) polymerase α also specifically reduced the induction of inflammatory mRNAs. These data demonstrate that there are differences in the 3' processing of inflammatory and housekeeping genes and identify polyadenylation as a novel target for anti-inflammatory drugs.


Assuntos
Desoxiadenosinas/farmacologia , Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Inflamação/prevenção & controle , Poliadenilação/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular , Quimiocina CCL2/genética , Quimiocina CXCL1/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Células HeLa , Humanos , Inflamação/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Interleucina-8/genética , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Células NIH 3T3 , Regiões Promotoras Genéticas , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Músculos Respiratórios/efeitos dos fármacos , Músculos Respiratórios/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
5.
J Immunol ; 187(11): 6094-107, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22025551

RESUMO

Severe asthma is associated with airway remodeling, characterized by structural changes including increased smooth muscle mass and matrix deposition in the airway, leading to deteriorating lung function. TGF-ß is a pleiotropic cytokine leading to increased synthesis of matrix molecules by human airway smooth muscle (HASM) cells and is implicated in asthmatic airway remodeling. TGF-ß is synthesized as a latent complex, sequestered in the extracellular matrix, and requires activation for functionality. Activation of latent TGF-ß is the rate-limiting step in its bioavailability. This study investigated the effect of the contraction agonists LPA and methacholine on TGF-ß activation by HASM cells and its role in the development of asthmatic airway remodeling. The data presented show that LPA and methacholine induced TGF-ß activation by HASM cells via the integrin αvß5. Our findings highlight the importance of the ß5 cytoplasmic domain because a polymorphism in the ß5 subunit rendered the integrin unable to activate TGF-ß. To our knowledge, this is the first description of a biologically relevant integrin that is unable to activate TGF-ß. These data demonstrate that murine airway smooth muscle cells express αvß5 integrins and activate TGF-ß. Finally, these data show that inhibition, or genetic loss, of αvß5 reduces allergen-induced increases in airway smooth muscle thickness in two models of asthma. These data highlight a mechanism of TGF-ß activation in asthma and support the hypothesis that bronchoconstriction promotes airway remodeling via integrin mediated TGF-ß activation.


Assuntos
Remodelação das Vias Aéreas/imunologia , Asma/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores de Vitronectina/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Asma/imunologia , Asma/patologia , Western Blotting , Linhagem Celular , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/patologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Vitronectina/imunologia , Sistema Respiratório , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Fator de Crescimento Transformador beta/imunologia
6.
Front Med (Lausanne) ; 10: 1275684, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37881627

RESUMO

Pulmonary hypertension (PH) due to chronic obstructive pulmonary disease (COPD) is classified as Group 3 PH, with no current proven targeted therapies. Studies suggest that cigarette smoke, the most risk factor for COPD can cause vascular remodelling and eventually PH as a result of dysfunction and proliferation of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs). In addition, hypoxia is a known driver of pulmonary vascular remodelling in COPD, and it is also thought that the presence of hypoxia in patients with COPD may further exaggerate cigarette smoke-induced vascular remodelling; however, the underlying cause is not fully understood. Three main pathways (prostanoids, nitric oxide and endothelin) are currently used as a therapeutic target for the treatment of patients with different groups of PH. However, drugs targeting these three pathways are not approved for patients with COPD-associated PH due to lack of evidence. Thus, this review aims to shed light on the role of impaired prostanoids, nitric oxide and endothelin pathways in cigarette smoke- and hypoxia-induced pulmonary vascular remodelling and also discusses the potential of using these pathways as therapeutic target for patients with PH secondary to COPD.

7.
J Biol Chem ; 285(38): 29101-10, 2010 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-20833730

RESUMO

Asthmatic airway smooth muscle (ASM) expresses interferon-γ-inducible protein-10 (CXCL10), a chemokine known to mediate mast cell migration into ASM bundles that has been reported in the airways of asthmatic patients. CXCL10 is elevated in patients suffering from viral exacerbations of asthma and in patients with chronic obstructive pulmonary disease (COPD), diseases in which corticosteroids are largely ineffective. IFNγ and TNFα synergistically induce CXCL10 release from human ASM cells in a steroid-insensitive manner, via an as yet undefined mechanism. We report that TNFα activates the classical NF-κB (nuclear factor κB) pathway, whereas IFNγ activates JAK2/STAT-1α and that inhibition of the JAK/STAT pathway is more effective in abrogating CXCL10 release than the steroid fluticasone. The synergy observed with TNFα and IFNγ together, however, did not lie at the level of NF-κB activation, STAT-1α phosphorylation, or in vivo binding of these transcription factors to the CXCL10 promoter. Stimulation of human ASM cells with TNFα and IFNγ induced histone H4 but not histone H3 acetylation at the CXCL10 promoter, although no synergism was observed when both cytokines were combined. We show, however, that TNFα and IFNγ exert a synergistic effect on the recruitment of CREB-binding protein (CBP) to the CXCL10, which is accompanied by increased RNA polymerase II. Our results provide evidence that synergism between TNFα and IFNγ lies at the level of coactivator recruitment in human ASM and suggest that inhibition of JAK/STAT signaling may be of therapeutic benefit in steroid-resistant airway disease.


Assuntos
Proteína de Ligação a CREB/metabolismo , Quimiocina CXCL10/metabolismo , Interferon gama/farmacologia , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Western Blotting , Proteína de Ligação a CREB/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Quimiocina CXCL10/genética , Imunoprecipitação da Cromatina , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos , NF-kappa B/genética , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética
8.
J Immunol ; 183(7): 4682-92, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19734226

RESUMO

CXCL8 is a neutrophil and mast cell chemoattractant that is involved in regulating inflammatory cell influx in asthma. Here, we investigated the transcriptional mechanism involved in CXCL8 induction by TNF-alpha in cultured human airway smooth muscle (HASM) cells and compared these in cells from nonasthmatic and asthmatic individuals. Transfection studies with mutated CXCL8 promoter constructs identified NF-kappaB, activating protein-1, and CAAT/enhancer binding protein (C/EBP)beta as key transcription factors, and binding of these three transcription factors to the CXCL8 promoter after TNF-alpha stimulation was confirmed by chromatin immunoprecipitation analysis. Cells derived from asthmatic individuals produced significantly higher levels of CXCL8 than nonasthmatic cells both basally and following 24 h of stimulation with TNF-alpha (p < 0.001). Furthermore, chromatin immunoprecipitation studies detected increased binding of NF-kappaB p65 and RNA polymerase II to the CXCL8 promoter of asthmatic HASM cells both in the presence and absence of TNF-alpha stimulation. This was not due to either an increased activation or phosphorylation of NF-kappaB per se or to an increase in its translocation to the nucleus. Increased binding of C/EBPbeta to the CXCL8 promoter of unstimulated cells was also detected in the asthmatic HASM cells. Collectively these studies show that HASM cells from asthmatic individuals have increased CXCL8 production due to the presence of a transcription complex on the CXCL8 promoter, which contains NF-kappaB, C/EBPbeta, and RNA polymerase II. This is the first description of an abnormality in transcription factor binding altering chemokine expression in airway structural cells in asthma.


Assuntos
Asma/imunologia , Brônquios/imunologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Miócitos de Músculo Liso/imunologia , RNA Polimerase II/metabolismo , Fator de Transcrição RelA/metabolismo , Regulação para Cima/imunologia , Asma/metabolismo , Asma/patologia , Brônquios/metabolismo , Brônquios/patologia , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Regulação para Cima/genética
9.
Am J Respir Cell Mol Biol ; 38(5): 600-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18079491

RESUMO

Mast cells are central in the development of several allergic diseases and contain a number of pre-formed mediators. beta-tryptase, the most abundant mast cell product, is increasingly recognized as a key inflammatory mediator, as it causes the release of cytokines, particularly the chemokine IL-8, from both inflammatory and structural cells. The molecular mechanisms, however, remain largely unknown. In this study we sought to investigate whether beta-tryptase could induce IL-8 expression in human airway smooth muscle (ASM) cells and to explore the molecular mechanisms involved. We found that purified human beta-tryptase stimulated IL-8 production in a time- and concentration-dependent manner, which was inhibited by protease inhibitors and mimicked by recombinant human beta-tryptase, but not by the protease-activated receptor-2 (PAR-2) agonist SLIGKV-NH(2), consistent with the low-level expression of PAR-2 protein in these cells. beta-tryptase also up-regulated IL-8 mRNA expression, as analyzed by RT-PCR and real-time PCR, which was abolished by the transcription inhibitor actinomycin D. Reporter gene assay showed that beta-tryptase-induced IL-8 transcription was mediated by the transcription factors activator protein-1, CCAAT/enhancer binding protein, and NF-kappaB, and chromatin immunoprecipitation assay demonstrated that beta-tryptase induced in vivo binding of these transcription factors to the IL-8 gene promoter. Furthermore, beta-tryptase stabilized IL-8 mRNA, suggesting additional post-transcriptional regulation. Collectively these findings show that beta-tryptase up-regulates IL-8 expression in ASM cells through a PAR-2-independent proteolytic mechanism and coordinated transcriptional and post-transcriptional regulation, which may be of particular importance in understanding the role and the mechanisms of action of beta-tryptase in regulating chemokine expression in mast cell-related disorders.


Assuntos
Regulação da Expressão Gênica/fisiologia , Interleucina-8/biossíntese , Interleucina-8/genética , Miócitos de Músculo Liso/enzimologia , Receptor PAR-2/fisiologia , Traqueia/enzimologia , Triptases/fisiologia , Células Cultivadas , Humanos , Hidrólise , Interleucina-8/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Receptor PAR-2/agonistas , Receptor PAR-2/metabolismo , Traqueia/citologia , Traqueia/metabolismo , Transcrição Gênica/fisiologia , Regulação para Cima/fisiologia
10.
Biochem Biophys Res Commun ; 370(2): 239-42, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18359288

RESUMO

Transforming growth factor beta (TGFbeta) is a key remodelling factor in asthma. It is produced as a latent complex and the main limiting step in TGFbeta bioavailability is its activation. Mast cell tryptase has been shown to stimulate the release of functionally active TGFbeta from human airway smooth muscle (ASM) cells [P. Berger, P.O. Girodet, H. Begueret, O. Ousova, D.W. Perng, R. Marthan, A.F. Walls, J.M. Tunon de Lara, Tryptase-stimulated human airway smooth muscle cells induce cytokine synthesis and mast cell chemotaxis, FASEB J. 17 (2003) 2139-2141]. The aim of this study was to determine if tryptase could cause TGFbeta activation as well as expression in ASM cells via its receptor, proteinase-activated receptor 2 (PAR2). Tryptase caused TGFbeta activation without affecting levels of total TGFbeta. This effect was inhibited by the selective tryptase inhibitor FUT175 and leupeptin but not mimicked by the PAR2 activating peptide SLIGKV-NH(2). Furthermore, the ASM cells used in the study did not express PAR2. The results indicate that tryptase activates TGFbeta via a PAR2-independent proteolytic mechanism in human ASM cells and may help understanding the role of tryptase in asthma.


Assuntos
Brônquios/enzimologia , Miócitos de Músculo Liso/enzimologia , Fator de Crescimento Transformador beta/metabolismo , Triptases/metabolismo , Benzamidinas , Brônquios/citologia , Guanidinas/farmacologia , Humanos , Leupeptinas/farmacologia , Oligopeptídeos/farmacologia , Receptor PAR-2/metabolismo , Inibidores da Tripsina/farmacologia , Triptases/antagonistas & inibidores
11.
Biochim Biophys Acta Gene Regul Mech ; 1861(5): 463-472, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29555582

RESUMO

Cyclooxygenase-2 (COX-2), with its main antifibrotic metabolite PGE2, is regarded as an antifibrotic gene. Repressed COX-2 expression and deficient PGE2 have been shown to contribute to the activation of lung fibroblasts and excessive deposition of collagen in pulmonary fibrosis. We have previously demonstrated that COX-2 expression in lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) is epigenetically silenced and can be restored by epigenetic inhibitors. This study aimed to investigate whether COX-2 downregulation induced by the profibrotic cytokine transforming growth factor-ß1 (TGF-ß1) in normal lung fibroblasts could be prevented by epigenetic inhibitors. We found that COX-2 protein expression and PGE2 production were markedly reduced by TGF-ß1 and this was prevented by the pan-histone deacetylase inhibitor suberanilohydroxamic acid (SAHA) and to a lesser extent by the DNA demethylating agent Decitabine (DAC), but not by the G9a histone methyltransferase (HMT) inhibitor BIX01294 or the EZH2 HMT inhibitor 3-deazaneplanocin A (DZNep). However, chromatin immunoprecipitation assay revealed that the effect of SAHA was unlikely mediated by histone modifications. Instead 3'-untranslated region (3'-UTR) luciferase reporter assay indicated the involvement of post-transcriptional mechanisms. This was supported by the downregulation by SAHA of the 3'-UTR mRNA binding protein TIA-1 (T-cell intracellular antigen-1), a negative regulator of COX-2 translation. Furthermore, TIA-1 knockdown by siRNA mimicked the effect of SAHA on COX-2 expression. These findings suggest SAHA can prevent TGF-ß1-induced COX-2 repression in lung fibroblasts post-transcriptionally through a novel TIA-1-dependent mechanism and provide new insights into the mechanisms underlying its potential antifibrotic activity.


Assuntos
Ciclo-Oxigenase 2/genética , Inibidores de Histona Desacetilases/administração & dosagem , Antígeno-1 Intracelular de Células T/genética , Fator de Crescimento Transformador beta1/genética , Adenosina/administração & dosagem , Adenosina/análogos & derivados , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Linhagem Celular , Ciclo-Oxigenase 1/genética , Metilação de DNA/genética , Decitabina , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Regiões Promotoras Genéticas , Vorinostat
12.
Mol Cell Biol ; 23(24): 9233-44, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14645533

RESUMO

Bradykinin and interleukin-1beta (IL-1beta) induce cyclooxygenase 2 (COX-2) in human airway smooth muscle cells. Here we extended our study to explore the gene transcriptional regulation. By transfection with various COX-2 promoter reporter constructs, we found that the bp -327-to-+59 promoter region was essential for COX-2 gene transcription by bradykinin and IL-1beta and that the cyclic AMP response element (CRE) was critical in bradykinin-induced transcription, whereas nuclear factor IL-6 and CRE and, to a lesser extent, nuclear factor-kappaB (NF-kappaB) were involved in IL-1beta-induced transcription. An electrophoretic mobility shift assay revealed that both bradykinin and IL-1beta elicited CRE-binding protein-1 (CREB-1) binding, and IL-1beta also elicited CCAAT/enhancer-binding protein beta and NF-kappaB binding to their respective elements in the COX-2 promoter. These transcription factors were associated with the COX-2 promoter, which was dynamically linked to different patterns of histone H4 acetylation by bradykinin and IL-1beta, as demonstrated by chromatin immunoprecipitation. We also revealed that endogenous prostaglandin E(2) was critical in bradykinin-induced COX-2 transcription initiation and involved in IL-1beta-induced COX-2 transcription at a latter stage. Our result provide the first evidence that COX-2 transcriptional regulation by different stimuli involves different promoter elements and transcription factors and is associated with chromatin remodeling after selective histone H4 acetylation in a stimulus-specific manner.


Assuntos
Bradicinina/farmacologia , Interleucina-1/farmacologia , Isoenzimas/genética , Prostaglandina-Endoperóxido Sintases/genética , Acetilação , Transporte Ativo do Núcleo Celular , Sequência de Bases , Sítios de Ligação/genética , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Ciclo-Oxigenase 2 , DNA/genética , DNA/metabolismo , Dinoprostona/metabolismo , Histonas/metabolismo , Humanos , Técnicas In Vitro , Proteínas de Membrana , Modelos Biológicos , Dados de Sequência Molecular , Músculo Liso/efeitos dos fármacos , Músculo Liso/enzimologia , Regiões Promotoras Genéticas , Músculos Respiratórios/efeitos dos fármacos , Músculos Respiratórios/enzimologia , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transfecção
13.
FEBS Lett ; 579(12): 2551-6, 2005 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-15862289

RESUMO

Here, we report that vascular endothelial growth factor (VEGF)-A secretion by human airway smooth muscle cells was increased by interleukin 1 beta (IL-1beta) and transforming growth factor beta (TGFbeta). IL-1beta and TGFbeta induced cyclo-oxygenase (COX)-2 protein and increased prostaglandin E(2) (PGE(2)). Both IL-1beta and TGFbeta increased VEGF-A(165) mRNA and VEGF promoter luciferase construct activity, in addition VEGF-A protein was inhibited by actinomycin D suggesting transcriptional regulation. The COX inhibitors indomethacin and NS398 inhibited IL-1beta but not TGFbeta mediated VEGF-A production. Furthermore, the effect of the COX inhibitors was overcome by adding exogenous PGE(2). In conclusion, IL-1beta increases VEGF-A secretion by COX-2 derived PGE(2) production whereas TGFbeta uses COX-independent pathways.


Assuntos
Citocinas/antagonistas & inibidores , Dinoprostona/biossíntese , Músculo Liso/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/biossíntese , Traqueia/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Citocinas/farmacologia , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Indução Enzimática , Humanos , Indometacina/farmacologia , Interleucina-1/antagonistas & inibidores , Interleucina-1/farmacologia , Cinética , Luciferases/metabolismo , Proteínas de Membrana , Músculo Liso/citologia , Músculo Liso/enzimologia , Nitrobenzenos/farmacologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Sulfonamidas/farmacologia , Traqueia/citologia , Traqueia/enzimologia , Fator de Crescimento Transformador beta/farmacologia
14.
FASEB J ; 16(11): 1435-7, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12205039

RESUMO

We previously reported that proinflammatory mediator bradykinin (BK) induces cyclooxygenase (COX)-2 expression in human airway smooth muscle (HASM), but the mechanism is unknown in any biological system. Here, we studied the role of specific protein kinase C (PKC) isozyme(s) in COX-2 expression. Among the eight PKC isozymes present in HASM cells, the Ca2+-independent PKC-delta and -epsilon and the Ca2+-dependent PKC-alpha and -betaI were translocated to the nucleus upon BK stimulation. BK-induced COX-2 expression and prostaglandin E2 (PGE2) accumulation were mimicked by the direct PKC activator phorbol 12-myristate 13-acetate (PMA) and inhibited by the broad spectrum PKC inhibitor bisindolylmaleimide I. However, the selective Ca2+-dependent PKC isozyme inhibitor Go 6976 had no effect. Furthermore, the membrane-permeable calcium chelator BAPTA-AM had no effect on BK-induced COX-2 expression and COX activity despite its inhibition of PGE2 accumulation, suggesting the involvement of Ca2+-independent PKC isozymes. Rottlerin, a PKC-delta inhibitor, also had no effect, likely implicating PKC-epsilon. BK-stimulated transcriptional activation of a COX-2 promoter reporter construct was enhanced by overexpression of wild-type PKC-epsilon and abolished by a dominant negative PKC-epsilon, but it was not affected by wild-type or dominant negative PKC-alpha or -delta. Collectively, our results demonstrate that PKC-e mediates BK-induced COX-2 expression in HASM cells.


Assuntos
Bradicinina/farmacologia , Isoenzimas/biossíntese , Isoenzimas/fisiologia , Pulmão/enzimologia , Músculo Liso/enzimologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Proteína Quinase C/fisiologia , Células Cultivadas , Ciclo-Oxigenase 2 , Dinoprostona/biossíntese , Humanos , Isoenzimas/genética , Pulmão/citologia , Proteínas de Membrana , Modelos Biológicos , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Mutação , Prostaglandina-Endoperóxido Sintases/genética , Proteína Quinase C/genética , Proteína Quinase C-épsilon , RNA Mensageiro/biossíntese , Ativação Transcricional
15.
Lung Cancer ; 40(1): 33-44, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12660005

RESUMO

Cyclooxygenase (COX) inhibitors are chemopreventive in many tumours but the role of COX inhibition in their effects is contentious. Here we determined if COX inhibitors influenced apoptosis in two non-small cell lung cancer cells one which over expresses COX-2 (MOR-P) and one which expresses neither isoform (H-460). NS398, a selective COX inhibitor, and indomethacin, a non-selective COX inhibitor, were cytotoxic in both cell lines, independently of their COX-2 expression. Furthermore, the cytotoxic concentrations were far greater than the concentrations required to inhibit COX. As indomethacin was more effective we used it in mechanistic studies. Indomethacin induced apoptotic cell death assessed as cytochrome c and apoptotic inducing factor (AIF) release, caspase activation, PARP, lamin B and gelsolin cleavage, chromatin condensation and nuclear fragmentation. The pan-caspase inhibitor, z-VAD, attenuated cell death, and blocked caspase activation, PARP cleavage and nuclear fragmentation without preventing cytochrome c release, suggesting that cytochrome c release is upstream of caspase activation. These observations suggest that COX inhibitors induce apoptosis in non-small lung cancer cells through cytochrome c and AIF release, and subsequent caspase activation, independently of COX-2 expression and prostaglandin production.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Inibidores de Ciclo-Oxigenase/farmacologia , Grupo dos Citocromos c/metabolismo , Indometacina/farmacologia , Isoenzimas/antagonistas & inibidores , Neoplasias Pulmonares/patologia , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Fator de Indução de Apoptose , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Dinoprostona/metabolismo , Flavoproteínas/metabolismo , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/enzimologia , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Prostaglandina-Endoperóxido Sintases , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
16.
Int J Biochem Cell Biol ; 45(7): 1366-76, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23583295

RESUMO

Chemokine (CC motif) ligand-2 (CCL2) is a member of C-C chemokine superfamily that contributes to inflammatory and fibrotic process. Studies in patients and experimental animals provide compelling evidence that increased CCL2 expression plays an important role in the development of fibroproliferative lung disease. The up-regulated CCL2 expression in pulmonary fibrosis is also involved in the potent profibrotic effects that thrombin exerts during lung injury. Here, we investigated the transcriptional mechanism involved in CCL2 production by thrombin in human primary lung fibroblasts and explored the transcriptional mechanism of increased CCL2 expression in pulmonary fibrosis. Thrombin increased CCL2 mRNA levels but not mRNA stability, suggesting it was acting transcriptionally. The increased binding of transcription factors to nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) elements in the CCL2 promoter contributed to active transcription following thrombin stimulation. Primary human lung fibroblasts isolated from patients with idiopathic pulmonary fibrosis (IPF) produced significantly higher levels of CCL2 than nonfibrotic lung fibroblasts. Furthermore, chromatin immunoprecipitation assays detected increased binding of NF-κB p65 and AP-1 subunit c-Jun to the CCL2 promoter of IPF cells both in the presence and absence of thrombin stimulation. The significantly increased binding of p65 and c-Jun to the CCL2 promoter was also observed in the lung tissue of bleomycin-induced pulmonary fibrosis murine model. Collectively, these findings strongly suggest that the increased binding of transcription factors to NF-κB and AP-1 elements in the CCL2 promoter is responsible for the active transcription expression of CCL2 in pulmonary fibrosis.


Assuntos
Quimiocina CCL2/metabolismo , NF-kappa B/metabolismo , Fibrose Pulmonar/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Bleomicina , Células Cultivadas , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Regulação da Expressão Gênica , Humanos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/genética , Transdução de Sinais , Trombina/metabolismo , Fator de Transcrição RelA/metabolismo , Transcrição Gênica , Ativação Transcricional
17.
Mol Cell Biol ; 30(12): 2874-86, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20404089

RESUMO

Targeted repression of a subset of key genes involved in tissue remodeling is a cardinal feature of idiopathic pulmonary fibrosis (IPF). The mechanism is unclear but is potentially important in disease pathogenesis and therapeutic targeting. We have previously reported that defective histone acetylation is responsible for the repression of the antifibrotic cyclooxygenase-2 gene. Here we extended our study to the repression of another antifibrotic gene, the potent angiostatic chemokine gamma interferon (IFN-gamma)-inducible protein of 10 kDa (IP-10), in lung fibroblasts from patients with IPF. We revealed that this involved not only histone deacetylation, as with cyclooxygenase-2 repression, but also histone H3 hypermethylation, as a result of decreased recruitment of histone acetyltransferases and increased presence of histone deacetylase (HDAC)-containing repressor complexes, histone methyltransferases G9a and SUV39H1, and heterochromatin protein 1 at the IP-10 promoter, leading to reduced transcription factor binding. More importantly, treatment of diseased cells with HDAC or G9a inhibitors similarly reversed the repressive histone deacetylation and hypermethylation and restored IP-10 expression. These findings strongly suggest that epigenetic dysregulation involving interactions between histone deacetylation and hypermethylation is responsible for targeted repression of IP-10 and potentially other antifibrotic genes in fibrotic lung disease and that this is amenable to therapeutic targeting.


Assuntos
Quimiocina CXCL10/metabolismo , Histonas/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Proteínas Repressoras/metabolismo , Acetilação/efeitos dos fármacos , Linhagem Celular , Quimiocina CXCL10/genética , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Citocinas/farmacologia , Histona Acetiltransferases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Fibrose Pulmonar Idiopática/genética , Metilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
18.
Mol Cell Biol ; 29(15): 4325-39, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19487460

RESUMO

Diminished cyclooxygenase 2 (COX-2) expression in fibroblasts, with a resultant defect in the production of the antifibrotic mediator prostaglandin E(2), plays a key role in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Here, we have characterized the molecular mechanism. We found that COX-2 mRNA levels in fibroblasts from patients with IPF (F-IPF) were significantly lower than those in fibroblasts from nonfibrotic lungs (F-NL) after transforming growth factor beta1 and interleukin-1beta treatment but that COX-2 mRNA degradation rates were similar, suggesting defective transcription. A reporter gene assay showed that there were no clear differences between F-IPF and F-NL in transcription factor involvement and activation in COX-2 gene transcription. However, a chromatin immunoprecipitation assay revealed that transcription factor binding to the COX-2 promoter in F-IPF was reduced compared to that in F-NL, an effect that was dynamically linked to reduced histone H3 and H4 acetylation due to decreased recruitment of histone acetyltransferases (HATs) and increased recruitment of transcriptional corepressor complexes to the COX-2 promoter. The treatment of F-IPF with histone deacetylase (HDAC) inhibitors together with cytokines increased histone H3 and H4 acetylation. Both HDAC inhibitors and the overexpression of HATs restored cytokine-induced COX-2 mRNA and protein expression in F-IPF. The results demonstrate that epigenetic abnormality in the form of histone hypoacetylation is responsible for diminished COX-2 expression in IPF.


Assuntos
Ciclo-Oxigenase 2/genética , Regulação Enzimológica da Expressão Gênica , Histonas/metabolismo , Fibrose Pulmonar Idiopática/genética , Acetilação/efeitos dos fármacos , Western Blotting , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Fibrose Pulmonar Idiopática/enzimologia , Fibrose Pulmonar Idiopática/patologia , Interleucina-1beta/farmacologia , Luciferases/genética , Luciferases/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo , Transfecção , Fator de Crescimento Transformador beta1/farmacologia
19.
J Immunol ; 176(6): 3788-95, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16517749

RESUMO

Recent studies have shown that a lack of eosinophils in asthmatic airway smooth muscle (ASM) bundles in contrast to the large number of mast cells is a key feature of asthma. We hypothesized that this is caused by beta-tryptase, the predominant mast cell-specific protease, abrogating the eosinophil chemotactic activities of ASM cell-derived eosinophil chemoattractants such as eotaxin and RANTES. We studied the effect of beta-tryptase on the immunoreactivities of human ASM cell-derived and recombinant eotaxin and other recombinant chemokines that are known to be produced by human ASM cells. We report in this study that purified beta-tryptase markedly reduced the immunoreactivity of human ASM cell-derived and recombinant eotaxin, but had no effect on eotaxin mRNA expression. The effect was mimicked by recombinant human beta-tryptase in the presence of heparin and was reversed by heat inactivation and the protease inhibitor leupeptin, suggesting that the proteolytic activity of tryptase is required. beta-Tryptase also exerted similar effects on recombinant RANTES, but not on the other chemokines and cytokines that were screened. Furthermore, a chemotaxis assay revealed that recombinant eotaxin and RANTES induced eosinophil migration concentration-dependently, which was abrogated by pretreatment of these chemokines with beta-tryptase. Another mast cell protease chymase also markedly reduced the immunoreactivity of eotaxin, but had no effect on RANTES and other chemokines and did not affect the influence of beta-tryptase on RANTES. These findings suggest that mast cell beta-tryptase selectively cleaves ASM-derived eotaxin and RANTES and abrogates their chemotactic activities, thus providing an explanation for the eosinophil paucity in asthmatic ASM bundles.


Assuntos
Quimiocina CCL5/metabolismo , Quimiocinas CC/metabolismo , Quimiotaxia , Eosinófilos/citologia , Mastócitos/enzimologia , Serina Endopeptidases/metabolismo , Asma/imunologia , Asma/metabolismo , Asma/patologia , Células Cultivadas , Quimiocina CCL11 , Quimiocina CCL5/imunologia , Quimiocinas CC/genética , Eosinófilos/imunologia , Eosinófilos/metabolismo , Regulação da Expressão Gênica , Humanos , Leupeptinas/metabolismo , RNA Mensageiro/genética , Proteínas Recombinantes/metabolismo , Triptases
20.
J Biol Chem ; 280(25): 23451-63, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15833737

RESUMO

Here we tested the effect of interleukin-1beta, a pro-inflammatory cytokine, on cAMP accumulation and chloride efflux in Calu-3 airway epithelial cells in response to ligands binding to adenylyl cyclase-coupled receptors such as the beta2 adrenoreceptor and EP prostanoid receptors. Interleukin-1beta significantly increased isoprenaline-induced cAMP accumulation by increasing beta2 adrenoreceptor numbers via a protein kinase A-dependent mechanism. In contrast, interleukin-1beta significantly impaired prostaglandin E2-induced cAMP accumulation by induction of cyclo-oxygenase-2, prostaglandin E2 production, and a resulting down-regulation of adenylyl cyclase. The cAMP changes were all mirrored by alterations in chloride efflux assessed using the fluorescent chloride probe N-(ethoxycarbonylmethyl)-6-methoxyquinolinium bromide with interleukin-1beta increasing chloride efflux in response to isoprenaline and reducing the response to prostaglandin E2. Studies with glibenclamide confirmed that chloride efflux was via the cystic fibrosis transmembrane conductance regulator. Calu-3 expresses EP4 receptors, but not EP2, and receptor expression is reduced by interleukin-1beta. Collectively, these results provide mechanistic insight into how interleukin-1beta can differentially regulate cAMP generation and chloride efflux in response to different adenylyl cyclase-coupled ligands in the same cell. These findings have important implications for diseases involving inflammation and abnormal ion flux such as cystic fibrosis.


Assuntos
Adenilil Ciclases/metabolismo , Brônquios/metabolismo , Cloretos/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Dinoprostona/fisiologia , Interleucina-1/fisiologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptores Adrenérgicos beta 2/fisiologia , Adulto , Sequência de Bases , Brônquios/citologia , Brônquios/efeitos dos fármacos , Brônquios/enzimologia , Linhagem Celular Transformada , Ciclo-Oxigenase 2 , Primers do DNA , Dinoprostona/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Humanos , Transporte de Íons , Masculino , Proteínas de Membrana
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa