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











Base de dados
Intervalo de ano de publicação
1.
J Biosci ; 462021.
Artigo em Inglês | MEDLINE | ID: mdl-34857676

RESUMO

S-nitrosothiols (SNOs) are small naturally occurring thiol and nitric oxide adducts that participate in many cell signaling pathways in living organisms. SNOs receive widespread attention in cell biology, biochemistry and chemistry because they can donate nitric oxide and/or nitrosonium ions in S-nitrosylation reactions, which are comparable to phosphorylation, acetylation, glutathionylation, and palmitoylation reactions. SNOs have advantageous effects in respiratory diseases and other systems in the body. S-nitrosylation signaling is a metabolically regulated physiological process that leads to specific post-translational protein modifications. S-nitrosylation signaling is faulty in cystic fibrosis (CF) and many other lung diseases. CF is an inherited, lethal autosomal recessive multisystem disease resulting from mutations in the gene encoding the CF transmembrane conductance regulatory (CFTR) protein. F508del CFTR is the most common mutation associated with CF, which results in CFTR misfolding because a phenylalanine is deleted from the primary structure of CFTR. The majority of wild-type CFTR and almost all F508del is degraded before reaching the cell surface. Ultimately, CF researchers have been looking to correct the mutated CFTR protein in the CF patients. Remarkably, researchers have found that SNOs levels are low in the CF lower airway compared to non-CF patients. We have been interested in determining whether SNOs increase CFTR maturation through S-nitrosylation. Maturation of both wild type and mutant F508del CFTR increases SNOs, which up-regulate CFTR maturation. In this review, we summarized our current knowledge of S-nitrosothiols signaling in cystic fibrosis airways.


Assuntos
Fibrose Cística , S-Nitrosotióis , Membrana Celular/metabolismo , Fibrose Cística/genética , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Humanos , Mutação , S-Nitrosotióis/metabolismo , Transdução de Sinais
2.
Int J Mol Sci ; 22(8)2021 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-33918821

RESUMO

Aptamers are single-stranded DNA or RNA sequences that bind target molecules with high specificity and affinity. Aptamers exhibit several notable advantages over protein-based therapeutics. Aptamers are non-immunogenic, easier to synthesize and modify, and can bind targets with greater affinity. Due to these benefits, aptamers are considered a promising therapeutic candidate to treat various conditions, including hematological disorders and cancer. An active area of research involves developing aptamers to target blood coagulation factors. These aptamers have the potential to treat cardiovascular diseases, blood disorders, and cancers. Although no aptamers targeting blood coagulation factors have been approved for clinical use, several aptamers have been evaluated in clinical trials and many more have demonstrated encouraging preclinical results. This review summarized our knowledge of the aptamers targeting proteins involved in coagulation, anticoagulation, fibrinolysis, their extensive applications as therapeutics and diagnostics tools, and the challenges they face for advancing to clinical use.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , Fatores de Coagulação Sanguínea/genética , Coagulação Sanguínea , Marcação de Genes , Animais , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Fatores de Coagulação Sanguínea/metabolismo , Proteínas de Transporte , Avaliação Pré-Clínica de Medicamentos , Fibrinólise , Marcação de Genes/métodos , Humanos , Ligação Proteica , Técnica de Seleção de Aptâmeros , Transdução de Sinais
3.
JCI Insight ; 6(1)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33232302

RESUMO

Existing animal models of cystic fibrosis (CF) have provided key insights into CF pathogenesis but have been limited by short lifespans, absence of key phenotypes, and/or high maintenance costs. Here, we report the CRISPR/Cas9-mediated generation of CF rabbits, a model with a relatively long lifespan and affordable maintenance and care costs. CF rabbits supplemented solely with oral osmotic laxative had a median survival of approximately 40 days and died of gastrointestinal disease, but therapeutic regimens directed toward restoring gastrointestinal transit extended median survival to approximately 80 days. Surrogate markers of exocrine pancreas disorders were found in CF rabbits with declining health. CFTR expression patterns in WT rabbit airways mimicked humans, with widespread distribution in nasal respiratory and olfactory epithelia, as well as proximal and distal lower airways. CF rabbits exhibited human CF-like abnormalities in the bioelectric properties of the nasal and tracheal epithelia. No spontaneous respiratory disease was detected in young CF rabbits. However, abnormal phenotypes were observed in surviving 1-year-old CF rabbits as compared with WT littermates, and these were especially evident in the nasal respiratory and olfactory epithelium. The CF rabbit model may serve as a useful tool for understanding gut and lung CF pathogenesis and for the practical development of CF therapeutics.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Animais , Sistemas CRISPR-Cas , Fibrose Cística/patologia , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Feminino , Trato Gastrointestinal/patologia , Trato Gastrointestinal/fisiopatologia , Técnicas de Inativação de Genes , Humanos , Masculino , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Sistema Respiratório/patologia , Sistema Respiratório/fisiopatologia , Distribuição Tecidual , Transcriptoma
4.
Sci Rep ; 10(1): 21088, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33273578

RESUMO

NADPH diaphorase is used as a histochemical marker of nitric oxide synthase (NOS) in aldehyde-treated tissues. It is thought that the catalytic activity of NOS promotes NADPH-dependent reduction of nitro-blue tetrazolium (NBT) to diformazan. However, it has been argued that a proteinaceous factor other than NOS is responsible for producing diformazan in aldehyde-treated tissues. We propose this is a NO-containing factor such as an S-nitrosothiol and/or a dinitrosyl-iron (II) cysteine complex or nitrosated proteins including NOS. We now report that (1) S-nitrosothiols covalently modify both NBT and TNBT, but only change the reduction potential of NBT after modification, (2) addition of S-nitrosothiols or ß- or α-NADPH to solutions of NBT did not elicit diformazan, (3) addition of S-nitrosothiols to solutions of NBT plus ß- or α-NADPH elicited rapid formation of diformazan in the absence or presence of paraformaldehyde, (4) addition of S-nitrosothiols to solutions of NBT plus ß- or α-NADP did not produce diformazan, (5) S-nitrosothiols did not promote NADPH-dependent reduction of tetra-nitro-blue tetrazolium (TNBT) in which all four phenolic rings are nitrated, (6) cytoplasmic vesicles in vascular endothelial cells known to stain for NADPH diaphorase were rich in S-nitrosothiols, and (7) procedures that accelerate decomposition of S-nitrosothiols, markedly reduced NADPH diaphorase staining in tissue sections subsequently subjected to paraformaldehyde fixation. Our results suggest that NADPH diaphorase in aldehyde-fixed tissues is not enzymatic but is due to the presence of NO-containing factors (free SNOs or nitrosated proteins such as NOS), which promote NADPH-dependent reduction of NBT to diformazan.


Assuntos
NADPH Desidrogenase/metabolismo , Óxido Nítrico Sintase/metabolismo , S-Nitrosotióis/metabolismo , Animais , Compostos Azo/metabolismo , Tronco Encefálico/química , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Cerebelo/química , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Formaldeído/farmacologia , Masculino , Nitroazul de Tetrazólio/metabolismo , Oxirredução , Polímeros/farmacologia , Ratos , Ratos Sprague-Dawley , Coloração e Rotulagem/métodos , Coloração e Rotulagem/normas
5.
Am J Respir Cell Mol Biol ; 61(6): 765-775, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31596601

RESUMO

S-nitrosothiols (SNOs) are endogenous signaling molecules that have numerous beneficial effects on the airway via cyclic guanosine monophosphate-dependent and -independent processes. Healthy human airways contain SNOs, but SNO levels are lower in the airways of patients with cystic fibrosis (CF). In this study, we examined the interaction between SNOs and the molecular cochaperone C-terminus Hsc70 interacting protein (CHIP), which is an E3 ubiquitin ligase that targets improperly folded CF transmembrane conductance regulator (CFTR) for subsequent degradation. Both CFBE41o- cells expressing either wild-type or F508del-CFTR and primary human bronchial epithelial cells express CHIP. Confocal microscopy and IP studies showed the cellular colocalization of CFTR and CHIP, and showed that S-nitrosoglutathione inhibits the CHIP-CFTR interaction. SNOs significantly reduced both the expression and activity of CHIP, leading to higher levels of both the mature and immature forms of F508del-CFTR. In fact, SNO inhibition of the function and expression of CHIP not only improved the maturation of CFTR but also increased CFTR's stability at the cell membrane. S-nitrosoglutathione-treated cells also had more S-nitrosylated CHIP and less ubiquitinated CFTR than cells that were not treated, suggesting that the S-nitrosylation of CHIP prevents the ubiquitination of CFTR by inhibiting CHIP's E3 ubiquitin ligase function. Furthermore, the exogenous SNOs S-nitrosoglutathione diethyl ester and S-nitro-N-acetylcysteine increased the expression of CFTR at the cell surface. After CHIP knockdown with siRNA duplexes specific for CHIP, F508del-CFTR expression increased at the cell surface. We conclude that SNOs effectively reduce CHIP-mediated degradation of CFTR, resulting in increased F508del-CFTR expression on airway epithelial cell surfaces. Together, these findings indicate that S-nitrosylation of CHIP is a novel mechanism of CFTR correction, and we anticipate that these insights will allow different SNOs to be optimized as agents for CF therapy.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Processamento de Proteína Pós-Traducional , S-Nitrosotióis/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Aprotinina/farmacologia , Células Cultivadas , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Humanos , Leupeptinas/farmacologia , Dobramento de Proteína , Estabilidade Proteica , Proteólise , Interferência de RNA , RNA Interferente Pequeno/farmacologia , S-Nitrosoglutationa/farmacologia , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
6.
FASEB J ; 33(11): 12602-12615, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31450978

RESUMO

Protein interactions that stabilize the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) at the apical membranes of epithelial cells have not yet been fully elucidated. We identified keratin 19 (CK19 or K19) as a novel CFTR-interacting protein. CK19 overexpression stabilized both wild-type (WT)-CFTR and Lumacaftor (VX-809)-rescued F508del-CFTR (where F508del is the deletion of the phenylalanine residue at position 508) at the plasma membrane (PM), promoting Cl- secretion across human bronchial epithelial (HBE) cells. CK19 prevention of Rab7A-mediated lysosomal degradation was a key mechanism in apical CFTR stabilization. Unexpectedly, CK19 expression was decreased by ∼40% in primary HBE cells from homogenous F508del patients with CF relative to non-CF controls. CK19 also positively regulated multidrug resistance-associated protein 4 expression at the PM, suggesting that this keratin may regulate the apical expression of other ATP-binding cassette proteins as well as CFTR.-Hou, X., Wu, Q., Rajagopalan, C., Zhang, C., Bouhamdan, M., Wei, H., Chen, X., Zaman, K., Li, C., Sun, X., Chen, S., Frizzell, R. A., Sun, F. CK19 stabilizes CFTR at the cell surface by limiting its endocytic pathway degradation.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Endocitose , Queratina-19/metabolismo , Proteólise , Fibrose Cística/genética , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células HEK293 , Células HeLa , Humanos , Queratina-19/genética , Lisossomos/genética , Lisossomos/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Mutação , Estabilidade Proteica
7.
Am J Physiol Lung Cell Mol Physiol ; 317(2): L247-L258, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31116581

RESUMO

The mechanisms by which transepithelial pressure changes observed during exercise and airway clearance can benefit lung health are challenging to study. Here, we have studied 117 mature, fully ciliated airway epithelial cell filters grown at air-liquid interface grown from 10 cystic fibrosis (CF) and 19 control subjects. These were exposed to cyclic increases in apical air pressure of 15 cmH2O for varying times. We measured the effect on proteins relevant to lung health, with a focus on the CF transmembrane regulator (CFTR). Immunoflourescence and immunoblot data were concordant in demonstrating that air pressure increased F508Del CFTR expression and maturation. This effect was in part dependent on the presence of cilia, on Ca2+ influx, and on formation of nitrogen oxides. These data provide a mechanosensory mechanism by which changes in luminal air pressure, like those observed during exercise and airway clearance, can affect epithelial protein expression and benefit patients with diseases of the airways.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Epitélio/metabolismo , Linhagem Celular , Humanos , Pulmão/metabolismo , Mucosa Respiratória/metabolismo
8.
Sci Rep ; 8(1): 4764, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29555962

RESUMO

Endoplasmic reticulum (ER)-associated protein degradation (ERAD) is an important quality control mechanism that eliminates misfolded proteins from the ER. The Derlin-1/VCP/VIMP protein complex plays an essential role in ERAD. Although the roles of Derlin-1 and VCP are relatively clear, the functional activity of VIMP in ERAD remains to be understood. Here we investigate the role of VIMP in the degradation of CFTRΔF508, a cystic fibrosis transmembrane conductance regulator (CFTR) mutant known to be a substrate of ERAD. Overexpression of VIMP markedly enhances the degradation of CFTRΔF508, whereas knockdown of VIMP increases its half-life. We demonstrate that VIMP is associated with CFTRΔF508 and the RNF5 E3 ubiquitin ligase (also known as RMA1). Thus, VIMP not only forms a complex with Derlin-1 and VCP, but may also participate in recruiting substrates and E3 ubiquitin ligases. We further show that blocking CFTRΔF508 degradation by knockdown of VIMP substantially augments the effect of VX809, a drug that allows a fraction of CFTRΔF508 to fold properly and mobilize from ER to cell surface for normal functioning. This study provides insight into the role of VIMP in ERAD and presents a potential target for the treatment of cystic fibrosis patients carrying the CFTRΔF508 mutation.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Degradação Associada com o Retículo Endoplasmático/genética , Proteínas de Membrana/metabolismo , Selenoproteínas/metabolismo , Deleção de Sequência , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Selenoproteínas/deficiência , Selenoproteínas/genética
9.
Am J Physiol Lung Cell Mol Physiol ; 310(3): L263-70, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26637637

RESUMO

S-nitrosoglutathione (GSNO) reductase regulates novel endogenous S-nitrosothiol signaling pathways, and mice deficient in GSNO reductase are protected from airways hyperreactivity. S-nitrosothiols are present in the airway, and patients with cystic fibrosis (CF) tend to have low S-nitrosothiol levels that may be attributed to upregulation of GSNO reductase activity. The present study demonstrates that 1) GSNO reductase activity is increased in the cystic fibrosis bronchial epithelial (CFBE41o(-)) cells expressing mutant F508del-cystic fibrosis transmembrane regulator (CFTR) compared with the wild-type CFBE41o(-) cells, 2) GSNO reductase expression level is increased in the primary human bronchial epithelial cells expressing mutant F508del-CFTR compared with the wild-type cells, 3) GSNO reductase colocalizes with cochaperone Hsp70/Hsp90 organizing protein (Hop; Stip1) in human airway epithelial cells, 4) GSNO reductase knockdown with siRNA increases the expression and maturation of CFTR and decreases Stip1 expression in human airway epithelial cells, 5) increased levels of GSNO reductase cause a decrease in maturation of CFTR, and 6) a GSNO reductase inhibitor effectively reverses the effects of GSNO reductase on CFTR maturation. These studies provide a novel approach to define the subcellular location of the interactions between Stip1 and GSNO reductase and the role of S-nitrosothiols in these interactions.


Assuntos
Aldeído Oxirredutases/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Aldeído Oxirredutases/farmacologia , Linhagem Celular , Membrana Celular/metabolismo , Humanos , Transdução de Sinais/fisiologia
10.
Curr Drug Targets ; 16(9): 923-36, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25557257

RESUMO

Cystic fibrosis (CF) is a lethal autosomal recessive disease that causes severe damage to the respiratory and digestive systems. It results from a dysfunctional CF Transmembrane Conductance Regulator (CFTR) protein, which is a cAMP- regulated epithelial chloride channel. CFTR is also a subtype of the ABC-transporter superfamily, and is expressed primarily in the apical membrane of epithelial cells in the airways, pancreas, and intestines. A single amino acid deletion of phenylalanine (Phe) is the most common mutation in CF patients known as F508del-CFTR. Normally, wild-type CFTR is largely degraded before reaching the cell membrane and F508del-CFTR virtually never reaches the cell surface. Ultimately, our goal is to correct dysfunctional CFTR proteins in CF patients. Via high-throughput screening techniques, several novel compounds for potential drugs effective in reversing the molecular CF defect and prohibiting further progression of CF have recently been discovered. S-nitrosothiols (SNOs) are small, naturally occurring endogenous cell signaling compounds, which have potential relevance to human lung diseases, including CF. Remarkably, researchers have found that the level of SNOs are reduced in the CF airway. It was previously reported that different types of SNOs, such as GSNO and S-nitrosoglutathione diethyl ester will increase CFTR maturation and function at the plasma membrane in human airway epithelial cells. The mechanisms by which SNOs improve CFTR maturation remain elusive. Currently, clinical trials are still investigating the effectiveness and safety of novel corrector and potentiator drugs for F508del- CFTR. This review article offers a summary of our knowledge on the most up-to-date CF therapies.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/tratamento farmacológico , Terapia de Alvo Molecular/métodos , S-Nitrosotióis/uso terapêutico , Ensaios Clínicos como Assunto , Fibrose Cística/genética , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/química , Regulador de Condutância Transmembrana em Fibrose Cística/efeitos dos fármacos , Humanos , Dobramento de Proteína/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , S-Nitrosotióis/farmacologia , Deleção de Sequência
11.
Biochem Biophys Res Commun ; 443(4): 1257-62, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24393850

RESUMO

S-nitrosothiols (SNOs) are endogenous signaling molecules with a broad spectrum of beneficial airway effects. SNOs are normally present in the airway, but levels tend to be low in cystic fibrosis (CF) patients. We and others have demonstrated that S-nitrosoglutathione (GSNO) increases the expression, maturation, and function of wild-type and mutant F508del cystic fibrosis transmembrane conductance regulator (CFTR) in human bronchial airway epithelial (HBAE) cells. We hypothesized that membrane permeable SNOs, such as S-nitrosoglutathione diethyl ester (GNODE) and S-nitroso-N-acetyl cysteine (SNOAC) may be more efficient in increasing the maturation of CFTR. HBAE cells expressing F508del CFTR were exposed to GNODE and SNOAC. The effects of these SNOs on the expression and maturation of F508del CFTR were determined by cell surface biotinylation and Western blot analysis. We also found for the first time that GNODE and SNOAC were effective at increasing CFTR maturation at the cell surface. Furthermore, we found that cells maintained at low temperature increased cell surface stability of F508del CFTR whereas the combination of low temperature and SNO treatment significantly extended the half-life of CFTR. Finally, we showed that SNO decreased the internalization rate of F508del CFTR in HBAE cells. We anticipate identifying the novel mechanisms, optimal SNOs, and lowest effective doses which could benefit cystic fibrosis patients.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , S-Nitrosotióis/farmacologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Fibrose Cística/genética , Fibrose Cística/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Meia-Vida , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Estabilidade Proteica/efeitos dos fármacos , S-Nitrosotióis/metabolismo , Deleção de Sequência , Transdução de Sinais
12.
Curr Pharm Des ; 19(19): 3509-20, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23331028

RESUMO

Cystic fibrosis (CF) is a multisystem disease associated with mutations in the gene that encodes the CF transmembrane conductance regulatory (CFTR) protein. The majority of wild-type CFTR and virtually all mutant ΔF508 CFTR are degraded before reaching the cell surface. Certain agents and conditions that increase expression and maturation of CFTR enable the protein to function at the cell surface. We and several research groups have reported that S-nitrosoglutathione (GSNO), a class of endogenous S-nitrosothiols, increases the maturation and function of CFTR in human airway epithelial cells. S-nitrosothiols (SNOs) are endogenous molecules with several cell signaling effects and potential relevance to human lung disease. SNOs are normally present in the human airway and have beneficial effects on lung function. Biochemical evidence suggests that SNOs act on post-translational protein modifications through mechanisms involving S-nitrosylation reactions. S-nitrosylation reactions are increasingly recognized to represent metabolically regulated cell signaling processes. Airway epithelial S-nitrosylation signaling disorders have been observed in a range of diseases, including CF. SNO levels are low in CF patients and normal physiological concentrations are effective in increasing CFTR maturation. The mechanisms by which SNOs improve CFTR expression appear to be novel. However, the precise mechanisms by which SNOs exert their beneficial effects are poorly understood. In the near future, we expect to identify the novel mechanisms by which SNO augments CFTR maturation. This information will be critical for optimizing the design and dosing of SNOs that might be used as CFTR corrector therapies in clinical trials.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/tratamento farmacológico , S-Nitrosotióis/metabolismo , S-Nitrosotióis/uso terapêutico , Animais , Membrana Celular/metabolismo , Fibrose Cística/genética , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Retículo Endoplasmático/metabolismo , Degradação Associada com o Retículo Endoplasmático/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Transporte Proteico/efeitos dos fármacos
13.
Proc Natl Acad Sci U S A ; 107(25): 11393-8, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20534503

RESUMO

The endogenous signaling molecule S-nitrosoglutathione (GSNO) and other S-nitrosylating agents can cause full maturation of the abnormal gene product DeltaF508 cystic fibrosis (CF) transmembrane conductance regulator (CFTR). However, the molecular mechanism of action is not known. Here we show that Hsp70/Hsp90 organizing protein (Hop) is a critical target of GSNO, and its S-nitrosylation results in DeltaF508 CFTR maturation and cell surface expression. S-nitrosylation by GSNO inhibited the association of Hop with CFTR in the endoplasmic reticulum. This effect was necessary and sufficient to mediate GSNO-induced cell-surface expression of DeltaF508 CFTR. Hop knockdown using siRNA recapitulated the effect of GSNO on DeltaF508 CFTR maturation and expression. Moreover, GSNO acted additively with decreased temperature, which promoted mutant CFTR maturation through a Hop-independent mechanism. We conclude that GSNO corrects DeltaF508 CFTR trafficking by inhibiting Hop expression, and that combination therapies--using differing mechanisms of action--may have additive benefits in treating CF.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Fibrose Cística/terapia , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Mutação , Nitrogênio/química , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Linhagem Celular , Membrana Celular/metabolismo , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Retículo Endoplasmático/metabolismo , Terapia Genética/métodos , Humanos , Modelos Biológicos , S-Nitrosoglutationa/química , Transdução de Sinais
14.
Respir Res ; 7: 124, 2006 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-17022806

RESUMO

BACKGROUND: It has been suggested that low microM concentrations of S-nitrosoglutathione (GSNO), an endogenous bronchodilator, may promote maturation of the defective cystic fibrosis (CF) transmembrane conductance regulator (CFTR). Because nitric oxide (NO) and GSNO levels appear to be low in the CF airway, there is an interest in the possibility that GSNO replacement could be of therapeutic benefit in CF. METHODS: The effect of GSNO on chloride (Cl-) transport was investigated in primary nasal epithelial cells obtained from CF patients homozygous for the delF508 mutation, as well as in two CF cell lines (CFBE and CFSME), using both a fluorescent Cl- indicator and X-ray microanalysis. Maturation of delF508 CFTR was determined by immunoblotting. RESULTS: Treatment with 60 microM GSNO for 4 hours increased cAMP-induced chloride efflux in nasal epithelial cells from 18 out of 21 CF patients, but did not significantly affect Cl- efflux in cells from healthy controls. This Cl- efflux was confirmed by measurements with a fluorescent Cl- indicator in the CFBE and CFSME cell lines. The effect of GSNO on Cl- efflux in CFBE cells could be inhibited both by a specific thiazolidinone CFTR inhibitor (CFTRinh-172) and by 4,4'-diisothiocyanatodihydrostilbene-2,2'-disulfonic acid (H2DIDS). X-ray microanalysis showed that, following 4 hours incubation with 60 microM GSNO, cAMP agonists caused a decrease in the cellular Cl- concentration in CFBE cells, corresponding to Cl- efflux. GSNO exposure resulted in an increase in the protein expression and maturation, as shown by immunoblot analysis. GSNO did not increase the cytosolic Ca2+ concentration in cultured airway epithelial cells. CONCLUSION: Previous studies have suggested that treatment with GSNO promotes maturation of delF508-CFTR, consistent with our results in this study. Here we show that GSNO increases chloride efflux, both in the two CF cell lines and in primary nasal epithelial cells from delF508-CF patients. This effect is at least in part mediated by CFTR. GSNO may be a candidate for pharmacological treatment of the defective chloride transport in CF epithelial cells.


Assuntos
Canais de Cloreto/metabolismo , Fibrose Cística/metabolismo , Glutationa/análogos & derivados , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/metabolismo , Nitrocompostos/farmacologia , Linhagem Celular , Células Cultivadas , Canais de Cloreto/genética , Fibrose Cística/genética , Glutationa/farmacologia , Humanos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo
15.
Mol Pharmacol ; 70(4): 1435-42, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16857740

RESUMO

The endogenous bronchodilator, S-nitrosoglutathione (GSNO), increases expression, maturation, and function of both the wild-type and the DeltaF508 mutant of the cystic fibrosis transmembrane conductance regulatory protein (CFTR). Though transcriptional mechanisms of action have been identified, GSNO seems also to have post-transcriptional effects on CFTR maturation. Here, we report that 1) GSNO is only one of a class of S-nitrosylating agents that, at low micromolar concentrations, increase DeltaF508 and wild-type CFTR expression and maturation; 2) NO itself (at these concentrations) and 8-bromocyclic GMP are minimally active on CFTR; 3) a novel agent, S-nitrosoglutathione diethyl ester, bypasses the need for GSNO bioactivation by gamma-glutamyl transpeptidase to increase CFTR maturation; 4) surprisingly, expression-but not S-nitrosylation-of cysteine string proteins (Csp) 1 and 2 is increased by GSNO; 5) the effect of GSNO to increase full maturation of wild-type CFTR is inhibited by Csp silencing (si)RNA; 6) proteins relevant to CFTR trafficking are SNO-modified, and SNO proteins traffic through the endoplasmic reticulum (ER) and Golgi after GSNO exposure; and 7) GSNO alters the interactions of DeltaF508 CFTR with Csp and Hsc70 in the ER and Golgi. These data suggest that GSNO is one of a class of S-nitrosylating agents that act independently of the classic NO radical/cyclic GMP pathway to increase CFTR expression and maturation. They also suggest that the effect of GSNO is dependent on Csp and on intracellular SNO trafficking. We speculate that these data will be of relevance to the development of NO donor-based therapies for CF.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/fisiologia , Regulação da Expressão Gênica , Processamento de Proteína Pós-Traducional , S-Nitrosoglutationa/farmacologia , Diferenciação Celular , Linhagem Celular , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico HSP40/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Proteínas de Membrana/fisiologia , Estrutura Molecular , S-Nitrosotióis/farmacologia
16.
Am J Physiol Lung Cell Mol Physiol ; 290(5): L827-32, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16603595

RESUMO

Human rhinovirus infection is a common trigger for asthma exacerbations. Asthma exacerbations and rhinovirus infections are both associated with markedly decreased pH and ammonium levels in exhaled breath condensates. This observation is thought to be related, in part, to decreased activity of airway epithelial glutaminase. We studied whether direct rhinovirus infection and/or the host immune response to the infection decreased airway epithelial cell surface pH in vitro. Interferon-gamma and tumor necrosis factor-alpha, but not direct rhinovirus infection, decreased pH, an effect partly associated with decreased ammonium concentrations. This effect was 1) prevented by nitric oxide synthase inhibition; 2) independent of cyclic GMP; 3) associated with an increase in endogenous airway epithelial cell S-nitrosothiol concentration; 4) mimicked by the exogenous S-nitrosothiol, S-nitroso-N-acetyl cysteine; and 5) independent of glutaminase expression and activity. We then confirmed that decreased epithelial pH inhibits human rhinovirus replication in airway epithelial cells. These data suggest that a nitric oxide synthase-dependent host response to viral infection mediated by S-nitrosothiols, rather than direct infection itself, plays a role in decreased airway surface pH during human rhinovirus infection. This host immune response may serve to protect the lower airways from direct infection in the normal host. In patients with asthma, however, this fall in pH could be associated with the increased mucus production, augmented inflammatory cell degranulation, bronchoconstriction, and cough characteristic of an asthma exacerbation.


Assuntos
Membrana Celular/fisiologia , Infecções por Picornaviridae/imunologia , Mucosa Respiratória/fisiopatologia , S-Nitrosotióis/farmacologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Citocinas/farmacologia , Humanos , Concentração de Íons de Hidrogênio , NG-Nitroarginina Metil Éster/farmacologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/virologia , Rhinovirus , Células Th1/imunologia
17.
Am J Respir Cell Mol Biol ; 34(4): 387-93, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16415251

RESUMO

S-Nitrosoglutathione (GSNO) is an endogenous bronchodilator with several beneficial pulmonary effects. Levels are decreased in the asthmatic airway, and GSNO inhalation has been proposed as an asthma therapy. 5-lipoxygenase (5-LO) is the rate-limiting enzyme in the synthetic pathway for cysteinyl leukotrienes (CysLTs), bronchoconstricting agents that are overproduced in asthma. Here, we have studied the effect of GSNO on the expression of 5-LO in human airway A549 cell lines and in primary normal human tracheobronchial epithelial (NHBE) cells in vitro. GSNO at concentrations of 0.5-1 microM caused a 3- to 6-fold increase in 5-LO expression. However, GSNO at>5 microM significantly inhibited both 5-LO expression and LT production. We also found that airway epithelial cells had gamma-glutamyl transpeptidase (gamma-GT) activity. The effect of 1 microM GSNO on 5-LO expression was prevented by the gamma-GT inhibitor, acivicin, suggesting a convergence of GSNO and CysLT metabolic pathway that may be relevant to asthma. Our data demonstrate that GSNO levels5microM suppresses 5-LO expression. These data suggest that GSNO might inhibit 5-LO expression in the clinical setting.


Assuntos
Araquidonato 5-Lipoxigenase/biossíntese , Mucosa Respiratória/enzimologia , S-Nitrosoglutationa/metabolismo , Broncodilatadores/farmacologia , Células Cultivadas , Ativação Enzimática , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica , Humanos , Doadores de Óxido Nítrico/farmacologia , Plicamicina/análogos & derivados , Plicamicina/farmacologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Mucosa Respiratória/efeitos dos fármacos , S-Nitrosoglutationa/farmacologia , Fator de Transcrição Sp1/fisiologia , Fator de Transcrição Sp3/fisiologia , gama-Glutamiltransferase/antagonistas & inibidores , gama-Glutamiltransferase/metabolismo
18.
Methods Enzymol ; 396: 95-105, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16291225

RESUMO

Protein and peptide S-nitrosothiols (SNOs) are involved in guanylate cyclase-independent signaling associated with nitric oxide synthase (NOS) activation. As a general rule, SNO formation requires the presence of an electron acceptor such as Cu2+. Various proteins have been identified that catalyze SNO formation, including NOS itself, ceruloplasmin, and hemoglobin. Biochemical evidence suggests the existence of other SNO synthases and NOS-associated proteins involved in SNO formation following NOS activation. Indeed, both hydrophilic and hydrophobic consensus motifs have been identified that favor protein S-nitrosylation. Inorganic SNO formation appears also to occur in biological systems at low pH levels and/or in membranes. Once formed, SNOs localized to specific cellular compartments signal specific effects, ranging from gene regulation to ion channel gating. Indeed, the number of cellular and physiological functions appreciated to be regulated through SNO synthesis, localization, and catabolism is increasing. Although research into SNO biosynthesis is in its infancy, the importance of this field of biochemistry has been confirmed repeatedly by investigators from a broad spectrum of disciplines.


Assuntos
S-Nitrosotióis/metabolismo , Peso Molecular , Peptídeos/química , Peptídeos/metabolismo , Proteínas/química , Proteínas/metabolismo , S-Nitrosotióis/química
19.
Biochem J ; 380(Pt 1): 67-74, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-14766015

RESUMO

The activities of certain nuclear regulatory proteins are modified by high concentrations of S-nitrosothiols associated with nitrosative stress. In the present study, we have studied the effect of physiological (low microM) concentrations of the endogenous S-nitrosothiol, GSNO (S-nitrosoglutathione), on the activities of nuclear regulatory proteins Sp3 and Sp1 (specificity proteins 3 and 1). Low concentrations of GSNO increased Sp3 binding, as well as Sp3-dependent transcription of the cystic fibrosis transmembrane conductance regulatory gene, cftr. However, higher GSNO levels prevented Sp3 binding, augmented Sp1 binding and prevented both cftr transcription and CFTR (cystic fibrosis transmembrane conductance regulator) expression. We conclude that low concentrations of GSNO favour Sp3 binding to 'housekeeping' genes such as cftr, whereas nitrosative stress-associated GSNO concentrations shut off Sp3-dependent transcription, possibly to redirect cellular resources. Since low micromolar concentrations of GSNO also increase the maturation and activity of a clinically common CFTR mutant, whereas higher concentrations have the opposite effect, these observations may have implications for dosing of S-nitrosylating agents used in cystic fibrosis clinical trials.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/biossíntese , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , S-Nitrosoglutationa/farmacologia , Fator de Transcrição Sp1/fisiologia , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral/efeitos dos fármacos , Meios de Cultura Livres de Soro , Cicloeximida/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Drosophila melanogaster/citologia , Humanos , Isoxazóis/farmacologia , Neoplasias Pulmonares/patologia , Plicamicina/farmacologia , Ligação Proteica/efeitos dos fármacos , Alvéolos Pulmonares/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/fisiologia , S-Nitrosoglutationa/administração & dosagem , Transdução de Sinais/fisiologia , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp3 , Transcrição Gênica , Transfecção
20.
J Neurosci ; 23(9): 3597-606, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12736330

RESUMO

Neuronal cell death in response to oxidative stress may reflect the failure of endogenous adaptive mechanisms. However, the transcriptional activators induced by oxidative stress in neurons that trigger adaptive genetic responses have yet to be fully elucidated. We report that basal DNA binding of the zinc finger transcription factors Sp1 and Sp3 is unexpectedly low in cortical neurons in vitro and is significantly induced by glutathione depletion-induced or hydrogen peroxide-induced oxidative stress in these cells. The increases in Sp1/Sp3 DNA binding reflect, in part, increased levels of Sp1 and Sp3 protein in the nuclei of cortical neurons. Similar induction of Sp1 and Sp3 protein is also observed in neurons in vivo in a chemical or a genetic model of Huntington's disease, two rodent models in which neuronal loss has been attributed to oxidative stress. Sustained high-level expression of full-length Sp1 or full-length Sp3, but not the Sp1 zinc finger DNA-binding domain alone, prevents death in response to oxidative stress, DNA damage, or both. Taken together, these results establish Sp1 and Sp3 as oxidative stress-induced transcription factors in cortical neurons that positively regulate neuronal survival.


Assuntos
Córtex Cerebral/metabolismo , Proteínas de Ligação a DNA/metabolismo , Homocisteína/análogos & derivados , Doença de Huntington/metabolismo , Neurônios/metabolismo , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , DNA/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/farmacologia , Modelos Animais de Doenças , Glutationa/metabolismo , Homocisteína/farmacologia , Doença de Huntington/induzido quimicamente , Doença de Huntington/genética , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Nitrocompostos , Oxidantes/farmacologia , Estresse Oxidativo/fisiologia , Propionatos , Ratos , Ratos Sprague-Dawley , Elementos de Resposta/fisiologia , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/farmacologia , Fator de Transcrição Sp3 , Fatores de Transcrição/genética , Fatores de Transcrição/farmacologia , Transfecção , Dedos de Zinco/genética , Dedos de Zinco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA