Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
2.
Am J Respir Cell Mol Biol ; 66(3): 323-336, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34890296

RESUMO

Administration of high concentrations of oxygen (hyperoxia) is one of few available options to treat acute hypoxemia-related respiratory failure, as seen in the current coronavirus disease (COVID-19) pandemic. Although hyperoxia can cause acute lung injury through increased production of superoxide anion (O2•-), the choice of high-concentration oxygen administration has become a necessity in critical care. The objective of this study was to test the hypothesis that UCP2 (uncoupling protein 2) has a major function of reducing O2•- generation in the lung in ambient air or in hyperoxia. Lung epithelial cells and wild-type; UCP2-/-; or transgenic, hTrx overexpression-bearing mice (Trx-Tg) were exposed to hyperoxia and O2•- generation was measured by using electron paramagnetic resonance, and lung injury was measured by using histopathologic analysis. UCP2 expression was analyzed by using RT-PCR analysis, Western blotting analysis, and RNA interference. The signal transduction pathways leading to loss of UCP2 expression were analyzed by using IP, phosphoprotein analysis, and specific inhibitors. UCP2 mRNA and protein expression were acutely decreased in hyperoxia, and these decreases were associated with a significant increase in O2•- production in the lung. Treatment of cells with rhTrx (recombinant human thioredoxin) or exposure of Trx-Tg mice prevented the loss of UCP2 protein and decreased O2•- generation in the lung. Trx is also required to maintain UCP2 expression in normoxia. Loss of UCP2 in UCP2-/- mice accentuated lung injury in hyperoxia. Trx activates the MKK4-p38MAPK (p38 mitogen-activated protein kinase)-PGC1α (PPARγ [peroxisome proliferator-activated receptor γ] coactivator 1α) pathway, leading to rescue of UCP2 and decreased O2•- generation in hyperoxia. Loss of UCP2 in hyperoxia is a major mechanism of O2•- production in the lung in hyperoxia. rhTrx can protect against lung injury in hyperoxia due to rescue of the loss of UCP2.


Assuntos
Pulmão/metabolismo , Oxigênio/metabolismo , Tiorredoxinas/metabolismo , Proteína Desacopladora 2/metabolismo , Animais , COVID-19/metabolismo , COVID-19/terapia , Linhagem Celular , Humanos , Hiperóxia/metabolismo , Pulmão/citologia , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oxigênio/toxicidade , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fosforilação , Transdução de Sinais , Superóxidos/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/farmacologia , Proteína Desacopladora 2/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 41(8): 2293-2314, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34039018

RESUMO

OBJECTIVE: Remote ischemic preconditioning (RIPC) is an intervention process where the application of multiple cycles of short ischemia/reperfusion (I/R) in a remote vascular bed provides protection against I/R injury. However, the identity of the specific RIPC factor and the mechanism by which RIPC alleviates I/R injury remains unclear. Here, we have investigated the identity and the mechanism by which the RIPC factor provides protection. APPROACH AND RESULTS: Using fluorescent in situ hybridization and immunofluorescence, we found that RIPC induces Nrg1ß expression in the endothelial cells, which is secreted into the serum. Whereas, RIPC protected against myocardial apoptosis and infarction, treatment with neutralizing-Nrg1 antibodies abolished the protective effect of RIPC. Further, increased superoxide anion generated in RIPC is required for Nrg1 expression. Improved myocardial perfusion and nitric oxide production were achieved by RIPC as determined by contrast echocardiography and electron spin resonance. However, treatment with neutralizing-Nrg1ß antibody abrogated these effects, suggesting Nrg1ß is a RIPC factor. ErbB2 (Erb-B2 receptor tyrosine kinase 2) is not expressed in the adult murine cardiomyocytes, but expressed in the endothelial cells of heart which is degraded in I/R. RIPC-induced Nrg1ß interacts with endothelial ErbB2 and thereby prevents its degradation. Mitochondrial Trx2 (thioredoxin) is degraded in I/R, but rescue of ErbB2 by Nrg1ß prevents Trx-2 degradation that decreased myocardial apoptosis in I/R. CONCLUSIONS: Nrg1ß is a RIPC factor that interacts with endothelial ErbB2 and prevents its degradation, which in turn prevents Trx2 degradation due to phosphorylation and inactivation of ATG5 (autophagy-related 5) by ErbB2. Nrg1ß also restored loss of eNOS (endothelial nitric oxide synthase) function in I/R via its interaction with Src.


Assuntos
Autofagia , Células Endoteliais/metabolismo , Membro Posterior/irrigação sanguínea , Precondicionamento Isquêmico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Neuregulina-1/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Receptor ErbB-2/metabolismo , Tiorredoxinas/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Proteína 5 Relacionada à Autofagia/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Neuregulina-1/antagonistas & inibidores , Fosforilação , Estabilidade Proteica , Proteólise , Receptor ErbB-2/genética , Transdução de Sinais , Quinases da Família src/metabolismo
4.
Microcirculation ; 28(4): e12674, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33316843

RESUMO

OBJECTIVE: Thioredoxin (Trx) is a small cellular redox protein with established antioxidant and disulfide reductase properties. We hypothesized that Trx deficiency in mice would cause increased oxidative stress with consequent redox imbalance that would exacerbate obesity-induced vascular dysfunction. METHODS: Non-transgenic (NT, C57BL/6) and dominant-negative Trx (dnTrx-Tg, low levels of redox-active protein) mice were either fed a normal diet (NC) or high fat diet plus sucrose (HFS) diet for 4 months (3-month HFD+ 1-month HFS). Weight gain, glucose tolerance test (GTT), insulin tolerance test (ITT), and other metabolic parameters were performed following NC or HFS diet. Arterial structural remodeling and functional parameters were assessed by myography. RESULTS: Our study found that dnTrx mice with lower levels of active Trx exacerbated myogenic tone, inward arterial remodeling, arterial stiffening, phenylephrine-induced contraction, and endothelial dysfunction of MA. Additionally, FeTMPyP, a peroxynitrite decomposition catalyst, acutely decreased myogenic tone and contraction and normalized endothelial function in MA from dnTrx-Tg mice on HFS via increasing nitric oxide (NO)-mediated relaxation. CONCLUSIONS: Our results indicate that deficiency of active Trx exacerbates MA contractile and relaxing properties during diet-induced obesity demonstrating that loss of redox balance in obesity is a key mechanism of vascular endothelial dysfunction.


Assuntos
Transtornos do Metabolismo de Glucose , Artérias Mesentéricas , Obesidade , Tiorredoxinas/metabolismo , Doenças Vasculares , Animais , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Feminino , Intolerância à Glucose/metabolismo , Transtornos do Metabolismo de Glucose/etiologia , Transtornos do Metabolismo de Glucose/metabolismo , Resistência à Insulina/fisiologia , Masculino , Artérias Mesentéricas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/metabolismo , Oxirredução , Estresse Oxidativo/fisiologia , Fenótipo , Doenças Vasculares/etiologia , Doenças Vasculares/metabolismo , Rigidez Vascular/fisiologia
5.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L903-L917, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30810065

RESUMO

High concentrations of oxygen (hyperoxia) are routinely used during anesthesia, and supplemental oxygen is also administered in connection with several other clinical conditions. Although prolonged hyperoxia is known to cause acute lung injury (ALI), whether short-duration hyperoxia causes lung toxicity remains unknown. We exposed mice to room air (RA or 21% O2) or 60% oxygen alone or in combination with 2% isoflurane for 2 h and determined the expression of oxidative stress marker genes, DNA damage and DNA repair genes, and expression of cell cycle regulatory proteins using quantitative PCR and Western analyses. Furthermore, we determined cellular apoptosis using TUNEL assay and assessed the DNA damage product 8-hydroxy-2'-deoxyguanosine (8-Oxo-dG) in the urine of 60% hyperoxia-exposed mice. Our study demonstrates that short-duration hyperoxia causes mitochondrial and nuclear DNA damage and that isoflurane abrogates this DNA damage and decreases apoptosis when used in conjunction with hyperoxia. In contrast, isoflurane mixed with RA caused significant 8-Oxo-dG accumulations in the mitochondria and nucleus. We further show that whereas NADPH oxidase is a major source of superoxide anion generated by isoflurane in normoxia, isoflurane inhibits superoxide generation in hyperoxia. Additionally, isoflurane also protected the mouse lungs against ALI (95% O2 for 36-h exposure). Our study established that short-duration hyperoxia causes genotoxicity in the lungs, which is abrogated when hyperoxia is used in conjunction with isoflurane, but isoflurane alone causes genotoxicity in the lung when delivered with ambient air.


Assuntos
Lesão Pulmonar Aguda , Dano ao DNA , Hiperóxia , Isoflurano/farmacologia , Pulmão , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/prevenção & controle , Animais , Linhagem Celular , Hiperóxia/metabolismo , Hiperóxia/patologia , Hiperóxia/prevenção & controle , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , NADPH Oxidases/metabolismo , Superóxidos/metabolismo
6.
Curr Hypertens Rep ; 20(1): 6, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29445879

RESUMO

PURPOSE OF REVIEW: Although the roles of oxidant stress and redox perturbations in hypertension have been the subject of several reviews, role of thioredoxin (Trx), a major cellular redox protein in age-related hypertension remains inadequately reviewed. The purpose of this review is to bring readers up-to-date with current understanding of the role of thioredoxin in age-related hypertension. RECENT FINDINGS: Age-related hypertension is a major underlying cause of several cardiovascular disorders, and therefore, intensive management of blood pressure is indicated in most patients with cardiovascular complications. Recent studies have shown that age-related hypertension was reversed and remained lowered for a prolonged period in mice with higher levels of human Trx (Trx-Tg). Additionally, injection of human recombinant Trx (rhTrx) decreased hypertension in aged wild-type mice that lasted for several days. Both Trx-Tg and aged wild-type mice injected with rhTrx were normotensive, showed increased NO production, decreased arterial stiffness, and increased vascular relaxation. These studies suggest that rhTrx could potentially be a therapeutic molecule to reverse age-related hypertension in humans. The reversal of age-related hypertension by restoring proteins that have undergone age-related modification is conceptually novel in the treatment of hypertension. Trx reverses age-related hypertension via maintaining vascular redox homeostasis, regenerating critical vasoregulatory proteins oxidized due to advancing age, and restoring native function of proteins that have undergone age-related modifications with loss-of function. Recent studies demonstrate that Trx is a promising molecule that may ameliorate or reverse age-related hypertension in older adults.


Assuntos
Envelhecimento/fisiologia , Pressão Sanguínea/fisiologia , Hipertensão , Tiorredoxinas/metabolismo , Animais , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Descoberta de Drogas , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Camundongos , Oxirredução
7.
J Biol Chem ; 291(45): 23374-23389, 2016 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-27587398

RESUMO

Reversible glutathionylation plays a critical role in protecting protein function under conditions of oxidative stress generally and for endothelial nitric-oxide synthase (eNOS) specifically. Glutathione-dependent glutaredoxin-mediated deglutathionylation of eNOS has been shown to confer protection in a model of heart damage termed ischemia-reperfusion injury, motivating further study of eNOS deglutathionylation in general. In this report, we present evidence for an alternative mechanism of deglutathionylation. In this pathway thioredoxin (Trx), a small cellular redox protein, is shown to rescue eNOS from glutathionylation during ischemia-reperfusion in a GSH-independent manner. By comparing mice with global overexpression of Trx and mice with cardiomyocyte-specific overexpression of Trx, we demonstrate that vascular Trx-mediated deglutathionylation of eNOS protects against ischemia-reperfusion-mediated myocardial infarction. Trx deficiency in endothelial cells promoted eNOS glutathionylation and reduced its enzymatic activity, whereas increased levels of Trx led to deglutathionylated eNOS. Thioredoxin-mediated deglutathionylation of eNOS in the coronary artery in vivo protected against reperfusion injury, even in the presence of normal levels of GSH. We further show that Trx directly interacts with eNOS, and we confirmed that Cys-691 and Cys-910 are the glutathionylated sites, as mutation of these cysteines partially rescued the decrease in eNOS activity, whereas mutation of a distal site, Cys-384, did not. Collectively, this study shows for the first time that Trx is a potent deglutathionylating protein in vivo and in vitro that can deglutathionylate proteins in the presence of high levels of GSSG in conditions of oxidative stress.


Assuntos
Glutationa/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Tiorredoxinas/metabolismo , Animais , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Interferência de RNA , RNA Interferente Pequeno/genética , Tiorredoxinas/genética , Regulação para Cima
8.
J Biol Chem ; 290(28): 17505-19, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26028649

RESUMO

The mitogen-activated protein kinase kinase 4 (MKK4) is activated via phosphorylation of Ser-257 and Thr-261 by upstream MAP3Ks and activates JNK and p38 MAPKs in response to cellular stress. We show that thioredoxin (Trx), a cellular redox protein, activates MKK4 via Cys-246 and Cys-266 residues as mutation of these residues renders MKK4 insensitive to phosphorylation by MAP3Ks, TNFα, or Trx. MKK4 is activated in vitro by reduced Trx but not oxidized Trx in the absence of an upstream kinase, suggesting that autophosphorylation of this protein occurs due to reduction of Cys-246 and Cys-266 by Trx. Additionally, mutation of Cys-246 and Cys-266 resulted in loss of kinase activity suggesting that the redox state of Cys-246 and Cys-266 is a critical determinant of MKK4 activation. Trx induces manganese superoxide dismutase (MnSOD) gene transcription by activating MKK4 via redox control of Cys-246 and Cys-266, as mutation of these residues abrogates MKK4 activation and MnSOD expression. We further show that MKK4 activates NFκB for its binding to the MnSOD promoter, which leads to AP-1 dissociation followed by MnSOD transcription. Taken together, our studies show that the redox status of Cys-246 and Cys-266 in MKK4 controls its activities independent of MAP3K, demonstrating integration of the endothelial redox environment to MAPK signaling.


Assuntos
Células Endoteliais/metabolismo , MAP Quinase Quinase 4/metabolismo , NF-kappa B/metabolismo , Superóxido Dismutase/genética , Tiorredoxinas/metabolismo , Substituição de Aminoácidos , Células Cultivadas , Cisteína/química , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Células HEK293 , Humanos , MAP Quinase Quinase 4/química , MAP Quinase Quinase 4/genética , MAP Quinase Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases , Mutagênese Sítio-Dirigida , Oxirredução , Fosforilação , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Fator de Transcrição AP-1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 308(5): L429-42, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25539854

RESUMO

Pulmonary oxygen toxicity is a major clinical problem for patients undergoing supplemental oxygen therapy. Thioredoxin (Trx) is an endogenous antioxidant protein that regenerates oxidatively inactivated proteins. We examined how Trx contributes to oxygen tolerance by creating transgenic mice with decreased levels of functional thioredoxin (dnTrx-Tg) using a dominant-negative approach. These mice showed decreased Trx activity in the lung although the expression of mutant protein is three times higher than the wild-type mice. Additionally, we found that these mice showed increased oxidation of endogenous Trx in room air. When exposed to hyperoxia (>90% O2) for 4 days, they failed to recover and showed significant mortality. Even in normal oxygen levels, these mice displayed a significant decrease in aconitase and NADH dehydrogenase activities, decreased mitochondrial energy metabolism, increased p53 and Gadd45α expression, and increased synthesis of proinflammatory cytokines. These effects were further increased by hyperoxia. We also generated mice overexpressing Trx (Trx-Tg) and found they maintained lung redox balance during exposure to high oxygen and thus were resistant to hyperoxia-induced lung injury. These mice had increased levels of reduced Trx in the lung in normoxia as well as hyperoxia. Furthermore, the levels of aconitase and NADH dehydrogenase activities were maintained in these mice concomitant with maintenance of mitochondrial energy metabolism. The genotoxic stress markers such as p53 or Gadd45α remained in significantly lower levels in hyperoxia compared with dnTrx-Tg or wild-type mice. These studies establish that mice deficient in functional Trx exhibit a phenotype of sensitivity to ambient air and hypersensitivity to hyperoxia.


Assuntos
Ar , Hiperóxia/complicações , Lesão Pulmonar/etiologia , Lesão Pulmonar/patologia , Tiorredoxinas/metabolismo , Aconitato Hidratase/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Respiração Celular , Galinhas , Citocinas/metabolismo , Humanos , Hiperóxia/patologia , Mediadores da Inflamação/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Lesão Pulmonar/metabolismo , Camundongos Transgênicos , Mitocôndrias/metabolismo , NADH Desidrogenase/metabolismo , Proteínas Nucleares/metabolismo , Oxirredução , Oxigênio , Consumo de Oxigênio , Fenótipo , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Análise de Sobrevida , Proteína Supressora de Tumor p53/metabolismo
10.
Mol Cell Biochem ; 395(1-2): 187-98, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24939362

RESUMO

Breathing high concentrations of oxygen (hyperoxia) causes lung injury and is associated with lung diseases such as bronchopulmonary dysplasia (BPD), respiratory distress syndrome and persistent pulmonary hypertension of the newborns. Hyperoxia (95-100 %O2) causes DNA damage and growth arrest of lung cells and consequently cells die by apoptosis or necrosis. Although supplemental oxygen therapy is clinically important, the level and duration of hyperoxic exposure that would allow lung cells to reenter the cell cycle remains unclear. We hypothesized that cells exposed to lower concentrations of hyperoxia will retain the capacity to enter cell cycle when recovered in room air. We employed varying concentrations of oxygen (21-95 %) to determine the response of lung cells to hyperoxia. Our results indicate that cells were growth arrested and failed to reenter the cell cycle when exposed to greater than 60 % oxygen. Cell cycle checkpoint proteins were increased in a biphasic manner, increasing until 70 % oxygen, but declined in greater than 90 % oxygen. Microarray analysis shows that there is significant decrease in the abundance of Cdks 6-8 and retinoblastoma protein (Rb), p107 and p130 in exposure to 90 % oxygen for 48 h. We further tested the effect of clinically relevant as needed oxygen [(pro-re-nata (prn)] in premature infant (125-days and 140-days) baboon model of BPD. The microarray results show that 6 or 14d PRN oxygen-exposed animals had induced expression of chromosomal maintenance genes (MCMs), genes related to anti-inflammation, proliferation, and differentiation.


Assuntos
Displasia Broncopulmonar/etiologia , Proteínas de Ciclo Celular/genética , Hiperóxia/genética , Pulmão/patologia , Animais , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patologia , Proteínas de Ciclo Celular/metabolismo , Cromossomos de Mamíferos/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hiperóxia/metabolismo , Hiperóxia/patologia , Pulmão/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Papio
11.
Cardiovasc Diabetol ; 12: 142, 2013 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-24093550

RESUMO

BACKGROUND: Endothelial dysfunction precedes pathogenesis of vascular complications in diabetes. In recent years, the mechanisms of endothelial dysfunction were investigated to outline strategies for its treatment. However, the therapies for dysfunctional endothelium resulted in multiple clinical trial failures and remain elusive. There is a need for defining hyperglycemia-induced endothelial dysfunction with both generic and specific dysfunctional changes in endothelial cells (EC) using a systems approach. In this study, we investigated hyperglycemia-induced endothelial dysfunction in HUVEC and HMVEC. We investigated hyperglycemia-induced functional changes (superoxide (O2⁻), and hydrogen peroxide (H2O2) production and mitochondrial membrane polarization) and gene expression fingerprints of related enzymes (nitric oxide synthase, NAD(P)H oxidase, and reactive oxygen species (ROS) neutralizing enzymes) in both ECs. METHOD: Gene expression of NOS2, NOS3, NOX4, CYBA, UCP1, CAT, TXNRD1, TXNRD2, GPX1, NOX1, SOD1, SOD2, PRDX1, 18s, and RPLP0 were measured using real-time PCR. O2⁻ production was measured with dihydroethidium (DHE) fluorescence measurement. H2O2 production was measured using Amplex Red assay. Mitochondrial membrane polarization was measured using JC-10 based fluorescence measurement. RESULTS: We showed that the O2⁻ levels increased similarly in both ECs with hyperglycemia. However, these endothelial cells showed significantly different underlying gene expression profile, H2O2 production and mitochondrial membrane polarization. In HUVEC, hyperglycemia increased H2O2 production, and hyperpolarized mitochondrial membrane. ROS neutralizing enzymes SOD2 and CAT gene expression were downregulated. In contrast, there was an upregulation of nitric oxide synthase and NAD(P)H oxidase and a depolarization of mitochondrial membrane in HMVEC. In addition, ROS neutralizing enzymes SOD1, GPX1, TXNRD1 and TXNRD2 gene expression were significantly upregulated in high glucose treated HMVEC. CONCLUSION: Our findings highlighted a unique framework for hyperglycemia-induced endothelial dysfunction. We showed that multiple pathways are differentially affected in these endothelial cells in hyperglycemia. High occurrences of gene expression changes in HMVEC in this study supports the hypothesis that microvasculature precedes macrovasculature in epigenetic regulation forming vascular metabolic memory. Identifying genomic phenotype and corresponding functional changes in hyperglycemic endothelial dysfunction will provide a suitable systems biology approach for understanding underlying mechanisms and possible effective therapeutic intervention.


Assuntos
Células Endoteliais/enzimologia , Glucose/metabolismo , Células Endoteliais da Veia Umbilical Humana/enzimologia , Hiperglicemia/enzimologia , Microvasos/enzimologia , Estresse Oxidativo/genética , Células Cultivadas , Regulação Enzimológica da Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Hiperglicemia/genética , Canais Iônicos/genética , Potencial da Membrana Mitocondrial , Proteínas Mitocondriais/genética , NADPH Oxidases/genética , Fator 2 Relacionado a NF-E2/genética , Óxido Nítrico Sintase/genética , RNA Mensageiro/metabolismo , Superóxidos/metabolismo , Biologia de Sistemas , Fatores de Tempo , Proteína Desacopladora 1
12.
Cardiovasc Toxicol ; 21(2): 142-151, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32880787

RESUMO

Cardiotoxicity is a major limitation for anthracycline chemotherapy although anthracyclines are potent antitumor agents. The precise mechanism underlying clinical heart failure due to anthracycline treatment is not fully understood, but is believed to be due, in part, to lipid peroxidation and the generation of free radicals by anthracycline-iron complexes. Thioredoxin (Trx) is a small redox-active antioxidant protein with potent disulfide reductase properties. Here, we present evidence that cancer cells overexpressing Trx undergo enhanced apoptosis in response to daunomycin. In contrast, cells overexpressing redox-inactive mutant Trx were not effectively killed. However, rat embryonic cardiomyocytes (H9c2 cells) overexpressing Trx were protected against daunomycin-mediated apoptosis, but H9c2 cells with decreased levels of active Trx showed enhanced apoptosis in response to daunomycin. We further demonstrate that increased level of Trx is specifically effective in anthracycline toxicity, but not with other topoisomerase II inhibitors such as etoposide. Collectively these data demonstrate that whereas high levels of Trx protect cardiomyocytes against anthracycline toxicity, it potentiates toxicity of anthracyclines in cancer cells.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Daunorrubicina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Tiorredoxinas/metabolismo , Animais , Cardiotoxicidade , Células HCT116 , Humanos , Células MCF-7 , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Tiorredoxinas/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Células U937
13.
J Biol Chem ; 284(25): 17069-17081, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19369702

RESUMO

We have investigated the role of cellular redox state on the regulation of cell cycle in hypoxia and shown that whereas cells expressing mutant thioredoxin (Trx) or a normal level of Trx undergo increased apoptosis, cells overexpressing Trx are protected against apoptosis. We show that hypoxia activates p53 and Chk1/Chk2 proteins in cells expressing normal or mutant Trx but not in cells overexpressing Trx. We also show that the activity of ribonucleotide reductase decreases in hypoxia in cells expressing redox-inactive Trx. Although hypoxia has been shown to induce reactive oxygen species (ROS) generation in the mitochondria resulting in enhanced p53 expression, our data demonstrate that hypoxia-induced p53 expression and phosphorylation are independent of ROS. Furthermore, hypoxia induces oxidation of Trx, and this oxidation is potentiated in the presence of 6-aminonicotinamide, an inhibitor of glucose-6-phosphate dehydrogenase. Taken together our study shows that Trx redox state is modulated in hypoxia independent of ROS and is a critical determinant of cell cycle regulation.


Assuntos
Ciclo Celular/fisiologia , Hipóxia Celular/fisiologia , Ribonucleotídeo Redutases/antagonistas & inibidores , Tiorredoxinas/metabolismo , 6-Aminonicotinamida/farmacologia , Apoptose , Sequência de Bases , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Linhagem Celular , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Expressão Gênica , Glucosefosfato Desidrogenase/antagonistas & inibidores , Humanos , Mutação , Oxirredução , Fosforilação , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribonucleotídeo Redutases/genética , Ribonucleotídeo Redutases/metabolismo , Tiorredoxinas/antagonistas & inibidores , Tiorredoxinas/genética , Proteína Supressora de Tumor p53/metabolismo
14.
Mol Cell Biochem ; 337(1-2): 53-63, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19859790

RESUMO

Thioredoxin (Trx) is a small ubiquitous protein, which has been shown to be involved in redox-dependent cellular functions. In this article, we demonstrate that the increased level of Trx induces AP-1 DNA binding in a redox-dependent manner by activating JNK subgroup of MAPKs. The majority of AP-1 DNA binding complex was found to be composed of cJun, JunB, and Fra-1. Increased expression of Trx resulted in phosphorylation of cJun, Jun B, and Fra-1. Further, increased expression of Trx induced the phosphorylation of MKK4 and MKK7 which are upstream kinases of the JNK signaling cascade. In co-transfection studies, AP-1-dependent luciferase reporter vector and pcDNA3-Trx increased luciferase activity demonstrating that increased expression of Trx increases AP-1 transactivation. In addition, dominant-negative JNK kinase (dnJNK/MKK4) or dominant-negative JNK (dnJNK) inhibited Trx-mediated AP-1 transactivation, as well as AP-1 DNA binding. Furthermore, transfection of kinase-dead MEKK1, an initiating kinase of the JNK pathway inhibited Trx-mediated AP-1 transactivation and DNA binding, suggesting that MEKK1 may mediate Trx-induced AP-1 activation. In contrast, wild-type MEKK1 overexpression did not inhibit Trx-mediated AP-1 activation. Taken together, our data demonstrate that increased expression of Trx induces MKK4/MKK7-dependent JNK activation, resulting in enhanced DNA binding, and transactivation of AP-1 transcription factor.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Tiorredoxinas/farmacologia , Fator de Transcrição AP-1/agonistas , DNA/metabolismo , Genes Reporter/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Luciferases/genética , Luciferases/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/fisiologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Ligação Proteica/efeitos dos fármacos , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Transfecção , Células Tumorais Cultivadas
15.
Aging (Albany NY) ; 12(19): 19809-19827, 2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-33049718

RESUMO

Aging is an independent risk factor for cardiovascular diseases, such as myocardial infarction due to ischemia-reperfusion injury (I/R) of the heart. Cytosolic thioredoxin (Trx) is a multifunctional redox protein which has antioxidant and protein disulfide reducing properties. We hypothesized that high levels of Trx will protect against multifactorial disease such as myocardial infarction due to I/R injury in aged mice. Aged mice overexpressing human Trx (Trx-Tg), mice expressing redox-inactive mutant of human Trx (dnTrx-Tg) and non-transgenic litter-mates (NT) were subjected to I/R (60/30 min), and cardiac function, mitochondrial structure and function, and biogenesis involving PGC1α pathway were evaluated in these mice. While aged Trx-Tg mice were protected from I/R-induced reduction in ejection fraction (EF) and fractional shortening (FS), had smaller infarct with decreased apoptosis and preserved mitochondrial function, aged dnTrx-Tg mice showed enhanced myocardial injury and mitochondrial dysfunction. Further, Trx-Tg mice were protected from I/R induced loss of PGC1α, ACO2, MFN1 and MFN2 in the myocardium. The dnTrx-Tg mice were highly sensitive to I/R induced apoptosis. Overall, our study demonstrated that the loss of Trx redox balance in I/R in aged NT or dnTrx-Tg mice resulted in decreased PGC1α expression that decreased mitochondrial gene expression with increased myocardial apoptosis. High levels of Trx, but not mitochondrial thioredoxin (Trx-2) maintained Trx redox balance in I/R resulting in increased PGC1α expression via AKT/CREB activation upregulating mitochondrial gene expression and protection against I/R injury.

16.
Cell Signal ; 20(4): 675-83, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18222647

RESUMO

Mitogen-activated protein kinases (MAPKs) are key signaling molecules that respond to mitogenic stimulation or environmental stress, resulting in the expression of target proteins. c-Jun N-terminal kinase (JNK) and p38 MAPKs are activated by inflammatory cytokines or environmental stress. Specific p38 MAPK inhibitors, such as SB202190 or SB203580, are widely used to dissect p38 MAPK-related signal transduction mechanisms. While using SB202190 to inhibit p38 MAPK-related signaling, we observed that SB202190 treatment could activate JNK. Further experiments showed that treatment of cells with SB202190 could phosphorylate JNK and activating transcription factor 2 (ATF-2), and increased AP-1 DNA binding. Using multiple cell lines and primary endothelial cells, we demonstrated that specific p38 MAPK inhibitors SB202190 or SB203580 induces the activation of the JNK pathway. Further, using with RNA interference and kinase-inactive expression of intermediates of the JNK pathway, we demonstrated SB202190- or SB203580-induced JNK activation is dependent on the MLK-3-MKK4/MKK7-dependent signal transduction pathway. Finally, we demonstrate that treatment of cells with SB202190 or SB203580 induces the phosphorylation and activation of MLK3.


Assuntos
Ativadores de Enzimas/farmacologia , Imidazóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 7/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Fator 2 Ativador da Transcrição/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ativação Enzimática , Humanos , MAP Quinase Quinase 7/genética , MAP Quinase Quinase Quinases/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação , Proteínas Proto-Oncogênicas c-jun/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Transcrição AP-1/metabolismo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
17.
Physiol Rep ; 5(23)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29212858

RESUMO

High-fat sucrose (HFS) diet in aged individuals causes severe weight gain (obesity) with much higher risk of cardiovascular diseases such as hypertension or atherosclerosis. Endothelial dysfunction is a major contributor for these vascular disorders. We hypothesize that prolonged ingestion of HFS diet by aged mice would accentuate endothelial dysfunction in the small resistance arteries. Male C57BL/6J mice at 12 weeks of age were divided into four groups and fed either normal chow (NC) or high-fat sucrose diet (HFS). Young group received NC for 4 months, and high-fat diet (HFD) for 3 months and 1 month HFS + 10% Sucrose (HFS diet). Aged mice received NC for 12 months. Aged HFS group received HFD for 4 months + 1 month HFD + 10% sucrose + 8 months HFD. Total body weight, plasma blood glucose levels, and glucose tolerance were determined in all groups. Isolated mesenteric arteries were assessed for arterial remodeling, myogenic tone, and vasomotor responses using pressure and wire myography. Both young and aged HFS mice showed impaired glucose tolerance (Y-NC, 137 ± 8.5 vs. Y-NC HFS, 228 ± 11.71; A-NC, 148 ± 6.42 vs. A-HFS, 225 ± 10.99), as well as hypercholesterolemia (Y-NC 99.50 ± 6.35 vs. Y-HFS 220.40 ± 16.34 mg/dL; A-NC 108.6 ± vs. A-HFS 279 ± 21.64) and significant weight gain (Y-NC 32.13 ± 0.8 g vs. Y-HFS 47.87 ± 2.18 g; A-NC 33.72 vs. A-HFS 56.28 ± 3.47 g) compared to both groups of mice on NC. The mesenteric artery from mice with prolonged HFS diet resulted in outward hypertrophic remodeling, increased stiffness, reduced myogenic tone, impaired vasodilation, increased contractility and blunted nitric oxide (NO) and EDH-mediated relaxations. Ebselen, a peroxinitrite scavenger rescued the endothelium derived relaxing factor (EDHF)-mediated relaxations. Our findings suggest that prolonged diet-induced obesity of aged mice can worsen small resistance artery endothelial dysfunction due to decrease in NO and EDHF-mediated relaxation, but, EDHF-mediated relaxation is a major contributor to overall endothelial dysfunction.


Assuntos
Fatores Biológicos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Artérias Mesentéricas/fisiopatologia , Doenças Vasculares/etiologia , Vasodilatação , Animais , Sacarose Alimentar/administração & dosagem , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Masculino , Artérias Mesentéricas/crescimento & desenvolvimento , Artérias Mesentéricas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Tono Muscular , Óxido Nítrico/metabolismo , Doenças Vasculares/metabolismo , Doenças Vasculares/fisiopatologia , Rigidez Vascular , Vasoconstrição
18.
Sci Transl Med ; 9(376)2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28179506

RESUMO

The incidence of high blood pressure with advancing age is notably high, and it is an independent prognostic factor for the onset or progression of a variety of cardiovascular disorders. Although age-related hypertension is an established phenomenon, current treatments are only palliative but not curative. Thus, there is a critical need for a curative therapy against age-related hypertension, which could greatly decrease the incidence of cardiovascular disorders. We show that overexpression of human thioredoxin (TRX), a redox protein, in mice prevents age-related hypertension. Further, injection of recombinant human TRX (rhTRX) for three consecutive days reversed hypertension in aged wild-type mice, and this effect lasted for at least 20 days. Arteries of wild-type mice injected with rhTRX or mice with TRX overexpression exhibited decreased arterial stiffness, greater endothelium-dependent relaxation, increased nitric oxide production, and decreased superoxide anion (O2•-) generation compared to either saline-injected aged wild-type mice or mice with TRX deficiency. Our study demonstrates a potential translational role of rhTRX in reversing age-related hypertension with long-lasting efficacy.


Assuntos
Envelhecimento/patologia , Vasos Sanguíneos/metabolismo , Hipertensão/tratamento farmacológico , Hipertensão/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Tiorredoxinas/uso terapêutico , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Artérias Carótidas/fisiopatologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Glutationa/metabolismo , Humanos , Hipertensão/fisiopatologia , Artéria Mesentérica Superior/efeitos dos fármacos , Artéria Mesentérica Superior/patologia , Artéria Mesentérica Superior/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NADPH Oxidases/metabolismo , Oxirredução , Fosforilação/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Superóxidos/metabolismo , Tiorredoxinas/farmacologia , Resistência Vascular/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
19.
Antioxid Redox Signal ; 7(11-12): 1740-3, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16356134

RESUMO

Thioredoxin (Trx) is a redox-active protein that has been shown to regulate various cellular processes due to its thiol-disulfide exchange reaction. It has antioxidant properties and also induces the expression of critical antioxidant enzymes such as manganese superoxide dismutase. Trx along with thioredoxin reductase and peroxiredoxins forms a complete system similar to the glutathione system, but with different and divergent functions. This review is a mini-update on key advances in the role of Trx in signal transduction and premature newborn biology. In addition, this mini-update also reviews recently reported prooxidant properties of Trx that relate to anthracycline redox cycling.


Assuntos
Recém-Nascido Prematuro/metabolismo , Tiorredoxinas/metabolismo , Animais , Antraciclinas/química , Antraciclinas/metabolismo , Antioxidantes/metabolismo , Humanos , Recém-Nascido , Oxirredução , Transdução de Sinais
20.
Hypertension ; 65(1): 130-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312439

RESUMO

Vascular thiol redox state has been shown to modulate vasodilator functions in large conductance Ca2+ -activated K+ channels and other related channels. However, the role of vascular redox in small resistance arteries is unknown. To determine how in vivo modulation of thiol redox state affects small resistance arteries relaxation, we generated a transgenic mouse strain that overexpresses thioredoxin, a small redox protein (Trx-Tg), and another strain that is thioredoxin-deficient (dnTrx-Tg). The redox state of the mesenteric arteries (MAs) in Trx-Tg mice is found to be predominantly in reduced state; in contrast, MAs from dnTrx-Tg mice remain in oxidized state. Thus, we created an in vivo redox system of mice and isolated the second-order branches of the main superior MAs from wild-type, Trx-Tg, or dnTrx-Tg mice to assess endothelium-dependent relaxing responses in a wire myograph. In MAs isolated from Trx-Tg mice, we observed an enhanced intermediate-conductance Ca2+ -activated potassium channel contribution resulting in a larger endothelium-dependent hyperpolarizing (EDH) relaxation in response to indirect (acetylcholine) and direct (NS309) opening of endothelial calcium-activated potassium channels. MAs derived from dnTrx-Tg mice showed both blunted nitric oxide-mediated and EDH-mediated relaxation compared with Trx-Tg mice. In a control study, diamide decreased EDH relaxations in MAs of wild-type mice, whereas dithiothreitol improved EDH relaxations and was able to restore the diamide-induced impairment in EDH response. Furthermore, the basal or angiotensin II-mediated systolic blood pressure remained significantly lower in Trx-Tg mice compared with wild-type or dnTrx-Tg mice, thus directly establishing redox-mediated EDH in blood pressure control.


Assuntos
Pressão Sanguínea/fisiologia , Hipertensão/metabolismo , Artérias Mesentéricas/metabolismo , Óxido Nítrico/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Resistência Vascular/fisiologia , Vasodilatação/fisiologia , Animais , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Hipertensão/fisiopatologia , Artérias Mesentéricas/fisiopatologia , Camundongos , Camundongos Transgênicos , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA