Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Glia ; 72(5): 885-898, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38311982

RESUMO

It is well established that axonal Neuregulin 1 type 3 (NRG1t3) regulates developmental myelin formation as well as EGR2-dependent gene activation and lipid synthesis. However, in peripheral neuropathy disease context, elevated axonal NRG1t3 improves remyelination and myelin sheath thickness without increasing Egr2 expression or activity, and without affecting the transcriptional activity of canonical myelination genes. Surprisingly, Pmp2, encoding for a myelin fatty acid binding protein, is the only gene whose expression increases in Schwann cells following overexpression of axonal NRG1t3. Here, we demonstrate PMP2 expression is directly regulated by NRG1t3 active form, following proteolytic cleavage. Then, using a transgenic mouse model overexpressing axonal NRG1t3 (NRG1t3OE) and knocked out for PMP2, we demonstrate that PMP2 is required for NRG1t3-mediated remyelination. We demonstrate that the sustained expression of Pmp2 in NRG1t3OE mice enhances the fatty acid uptake in sciatic nerve fibers and the mitochondrial ATP production in Schwann cells. In sum, our findings demonstrate that PMP2 is a direct downstream mediator of NRG1t3 and that the modulation of PMP2 downstream NRG1t3 activation has distinct effects on Schwann cell function during developmental myelination and remyelination.


Assuntos
Bainha de Mielina , Remielinização , Camundongos , Animais , Bainha de Mielina/metabolismo , Células de Schwann/metabolismo , Axônios/metabolismo , Nervo Isquiático/metabolismo , Camundongos Transgênicos , Trifosfato de Adenosina/metabolismo
2.
J Immunol ; 207(7): 1812-1823, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34462312

RESUMO

Radiation is associated with tissue damage and increased risk of atherosclerosis, but there are currently no treatments and a very limited mechanistic understanding of how radiation impacts tissue repair mechanisms. We uncovered that radiation significantly delayed temporal resolution programs that were associated with decreased efferocytosis in vivo. Resolvin D1 (RvD1), a known proresolving ligand, promoted swift resolution and restored efferocytosis in sublethally irradiated mice. Irradiated macrophages exhibited several features of senescence, including increased expression of p16INK4A and p21, heightened levels of SA-ß-gal, COX-2, several proinflammatory cytokines/chemokines, and oxidative stress (OS) in vitro, and when transferred to mice, they exacerbated inflammation in vivo. Mechanistically, heightened OS in senescent macrophages led to impairment in their ability to carry out efficient efferocytosis, and treatment with RvD1 reduced OS and improved efferocytosis. Sublethally irradiated Ldlr -/- mice exhibited increased plaque necrosis, p16INK4A cells, and decreased lesional collagen compared with nonirradiated controls, and treatment with RvD1 significantly reduced necrosis and increased lesional collagen. Removal of p16INK4A hematopoietic cells during advanced atherosclerosis with p16-3MR mice reduced plaque necrosis and increased production of key intraplaque-resolving mediators. Our results demonstrate that sublethal radiation drives macrophage senescence and efferocytosis defects and suggest that RvD1 may be a new therapeutic strategy to limit radiation-induced tissue damage.


Assuntos
Aterosclerose/imunologia , Doenças Cardiovasculares/imunologia , Ácidos Docosa-Hexaenoicos/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Macrófagos/imunologia , Lesões por Radiação/imunologia , Cicatrização/efeitos da radiação , Animais , Aterosclerose/genética , Células Cultivadas , Senescência Celular , Ciclo-Oxigenase 2/metabolismo , Genes p16 , Humanos , Inflamação , Camundongos , Camundongos Knockout , Radiação
3.
Am J Respir Cell Mol Biol ; 65(3): 259-271, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33909984

RESUMO

Patients with pulmonary emphysema often develop locomotor muscle dysfunction, which is independently associated with disability and higher mortality in that population. Muscle dysfunction entails reduced force generation capacity, which partially depends on fibers' oxidative potential, yet very little mechanistic research has focused on muscle respiration in pulmonary emphysema. Using a recently established animal model of pulmonary emphysema-driven skeletal muscle dysfunction, we found downregulation of SDHC (succinate dehydrogenase subunit C) in association with lower oxygen consumption and fatigue tolerance in locomotor muscles. Reduced SDH activity has been previously observed in muscles from patients with pulmonary emphysema, and we found that SDHC is required to support respiration in cultured muscle cells. Moreover, in vivo gain of SDH function in emphysema animals' muscles resulted in better oxygen consumption rate and fatigue tolerance. These changes correlated with a larger number of relatively more oxidative type 2-A and 2X fibers and a reduced amount of 2B fibers. Our data suggest that SDHC is a key regulator of respiration and fatigability in pulmonary emphysema-driven skeletal muscles, which could be impactful in developing strategies aimed at attenuating this comorbidity.


Assuntos
Fadiga/enzimologia , Proteínas de Membrana/metabolismo , Músculo Esquelético/enzimologia , Consumo de Oxigênio , Enfisema Pulmonar/enzimologia , Animais , Modelos Animais de Doenças , Fadiga/genética , Fadiga/patologia , Fadiga/fisiopatologia , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Enfisema Pulmonar/genética , Enfisema Pulmonar/patologia , Enfisema Pulmonar/fisiopatologia
4.
Am J Physiol Heart Circ Physiol ; 317(5): H969-H980, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31518169

RESUMO

Multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a multigene family with isoform-specific regulation of vascular smooth muscle (VSM) functions. In previous studies, we found that vascular injury resulted in VSM dedifferentiation and reduced expression of the CaMKIIγ isoform in medial wall VSM. Smooth muscle knockout of CaMKIIγ enhanced injury-induced VSM neointimal hyperplasia, whereas CaMKIIγ overexpression inhibited VSM proliferation and neointimal formation. In this study, we evaluated DNA cytosine methylation/demethylation as a mechanism for regulating CaMKII isoform expression in VSM. Inhibition of cytosine methylation with 5-Aza-2'-deoxycytidine significantly upregulated CaMKIIγ expression in cultured VSM cells and inhibited CaMKIIγ downregulation in organ-cultured aorta ex vivo. With the use of methylated cytosine immunoprecipitation, the rat Camk2g promoter was found hypomethylated in differentiated VSM, whereas injury- or cell culture-induced VSM dedifferentiation coincided with Camk2g promoter methylation and decreased expression. We report for the first time that VSM cell phenotype switching is accompanied by marked induction of thymine DNA glycosylase (TDG) protein and mRNA expression in injured arteries in vivo and in cultured VSM synthetic phenotype cells. Silencing Tdg in VSM promoted expression of CaMKIIγ and differentiation markers, including myocardin, and inhibited VSM cell proliferation and injury-induced neointima formation. This study indicates that CaMKIIγ expression in VSM is regulated by cytosine methylation/demethylation and that TDG is an important determinant of this process and, more broadly, VSM phenotype switching and function.NEW & NOTEWORTHY Expression of the calcium calmodulin-dependent protein kinase II-γ isoform (CaMKIIγ) is associated with differentiated vascular smooth muscle (VSM) and negatively regulates proliferation in VSM synthetic phenotype (VSMSyn) cells. This study demonstrates that thymine DNA glycosylase (TDG) plays a key role in regulating CaMKIIγ expression in VSM through promoter cytosine methylation/demethylation. TDG expression is strongly induced in VSMSyn cells and plays key roles in negatively regulating CaMKIIγ expression and more broadly VSM phenotype switching.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Lesões das Artérias Carótidas/enzimologia , Plasticidade Celular , Metilação de DNA , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Timina DNA Glicosilase/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/enzimologia , Artéria Carótida Primitiva/patologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Masculino , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Neointima , Fenótipo , Regiões Promotoras Genéticas , Ratos Sprague-Dawley , Transdução de Sinais , Timina DNA Glicosilase/genética
5.
Am J Physiol Renal Physiol ; 315(4): F769-F780, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29631355

RESUMO

Most renal transplants ultimately fail secondary to chronic allograft nephropathy (CAN). Vimentin (vim) is a member of the intermediate filament family of proteins and has been shown to be important in the development of CAN. One of the pathways leading to chronic renal fibrosis after transplant is thought to be epithelial to mesenchymal transition (EMT). Even though vim expression is one of the main steps of EMT, it is unknown whether vim expression is required for EMT leading to renal fibrosis and allograft loss. To this end, the role of vim in renal fibrosis was determined via unilateral ureteral obstruction (UUO) in vim knockout mice (129 svs6 vim -/-). Following UUO, kidneys were recovered and analyzed via Western blotting, immunofluorescence, and transcriptomics. Cultured human proximal renal tubular (HK-2) cells were subjected to lentiviral-driven inhibition of vim expression and then treated with transforming growth factor (TGF)-ß to undergo EMT. Immunoblotting as well as wound healing assays were used to determine development of EMT. Western blotting analyses of mice undergoing UUO reveal increased levels of vim soon after UUO. As expected, interstitial collagen deposition increased in control mice following UUO but decreased in vim -/- kidneys. Immunofluorescence analyses also revealed altered localization of ß-catenin in vim -/- mice undergoing UUO without significant changes in mRNA levels. However, RNA sequencing revealed a decrease in ß-catenin-dependent genes in vim -/- kidneys. Finally, vim-silenced HK-2 cell lines undergoing EMT were shown to have decreased cellular migration during wound healing. We conclude that vim inhibition decreases fibrosis following UUO by possibly altering ß-catenin localization and downstream signaling.


Assuntos
Fibrose/patologia , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Vimentina/metabolismo , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/fisiologia , Fibrose/metabolismo , Filamentos Intermediários/metabolismo , Filamentos Intermediários/patologia , Camundongos Knockout , Transdução de Sinais/fisiologia , Sistema Urinário/metabolismo , Sistema Urinário/patologia
6.
FASEB J ; 31(6): 2576-2591, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28258189

RESUMO

Tetraspanins (TSPANs) comprise a large family of 4-transmembrane domain proteins. The importance of TSPANs in vascular smooth muscle cells (VSMCs) is unexplored. Given that TGF-ß1 and myocardin (MYOCD) are potent activators for VSMC differentiation, we screened for TGF-ß1 and MYOCD/serum response factor (SRF)-regulated TSPANs in VSMC by using RNA-seq analyses and RNA-arrays. TSPAN2 was found to be the only TSPAN family gene induced by TGF-ß1 and MYOCD, and reduced by SRF deficiency in VSMCs. We also found that TSPAN2 is highly expressed in smooth muscle-enriched tissues and down-regulated in in vitro models of VSMC phenotypic modulation. TSPAN2 expression is attenuated in mouse carotid arteries after ligation injury and in failed human arteriovenous fistula samples after occlusion by dedifferentiated neointimal VSMC. In vitro functional studies showed that TSPAN2 suppresses VSMC proliferation and migration. Luciferase reporter and chromatin immunoprecipitation assays demonstrated that TSPAN2 is regulated by 2 parallel pathways, MYOCD/SRF and TGF-ß1/SMAD, via distinct binding elements within the proximal promoter. Thus, we identified the first VSMC-enriched and MYOCD/SRF and TGF-ß1/SMAD-dependent TSPAN family member, whose expression is intimately associated with VSMC differentiation and negatively correlated with vascular disease. Our results suggest that TSPAN2 may play important roles in vascular disease.-Zhao, J., Wu, W., Zhang, W., Lu, Y. W., Tou, E., Ye, J., Gao, P., Jourd'heuil, D., Singer, H. A., Wu, M., Long, X. Selective expression of TSPAN2 in vascular smooth muscle is independently regulated by TGF-ß1/SMAD and myocardin/serum response factor.


Assuntos
Músculo Liso Vascular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fator de Resposta Sérica/metabolismo , Proteínas Smad/metabolismo , Tetraspaninas/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Miócitos de Músculo Liso/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Fator de Resposta Sérica/genética , Proteínas Smad/genética , Tetraspaninas/genética , Transativadores/genética , Transcriptoma , Fator de Crescimento Transformador beta1/genética
7.
Arterioscler Thromb Vasc Biol ; 37(10): 1944-1955, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28798140

RESUMO

OBJECTIVE: The role of hemoglobin and myoglobin in the cardiovascular system is well established, yet other globins in this context are poorly characterized. Here, we examined the expression and function of cytoglobin (CYGB) during vascular injury. APPROACH AND RESULTS: We characterized CYGB content in intact vessels and primary vascular smooth muscle (VSM) cells and used 2 different vascular injury models to examine the functional significance of CYGB in vivo. We found that CYGB was strongly expressed in medial arterial VSM and human veins. In vitro and in vivo studies indicated that CYGB was lost after VSM cell dedifferentiation. In the rat balloon angioplasty model, site-targeted delivery of adenovirus encoding shRNA specific for CYGB prevented its reexpression and decreased neointima formation. Similarly, 4 weeks after complete ligation of the left common carotid, Cygb knockout mice displayed little to no evidence of neointimal hyperplasia in contrast to their wild-type littermates. Mechanistic studies in the rat indicated that this was primarily associated with increased medial cell loss, terminal uridine nick-end labeling staining, and caspase-3 activation, all indicative of prolonged apoptosis. In vitro, CYGB could be reexpressed after VSM stimulation with cytokines and hypoxia and loss of CYGB sensitized human and rat aortic VSM cells to apoptosis. This was reversed after antioxidant treatment or NOS2 (nitric oxide synthase 2) inhibition. CONCLUSIONS: These results indicate that CYGB is expressed in vessels primarily in differentiated medial VSM cells where it regulates neointima formation and inhibits apoptosis after injury.


Assuntos
Apoptose , Globinas/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiopatologia , Remodelação Vascular/fisiologia , Animais , Caspase 3/metabolismo , Diferenciação Celular , Citoglobina , Regulação para Baixo , Ativação Enzimática , Camundongos , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Neointima/fisiopatologia , Óxido Nítrico Sintase Tipo II/toxicidade , Oxirredução , Ratos
8.
Arterioscler Thromb Vasc Biol ; 36(10): 2088-99, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27444199

RESUMO

OBJECTIVE: Long noncoding RNAs (lncRNA) represent a growing class of noncoding genes with diverse cellular functions. We previously reported on SENCR, an lncRNA that seems to support the vascular smooth muscle cell (VSMC) contractile phenotype. However, information about the VSMC-specific lncRNAs regulated by myocardin (MYOCD)/serum response factor, the master switch for VSMC differentiation, is unknown. APPROACH AND RESULTS: To define novel lncRNAs with functions related to VSMC differentiation, we performed RNA sequencing in human coronary artery SMCs that overexpress MYOCD. Several novel lncRNAs showed altered expression with MYOCD overexpression and one, named MYOcardin-induced Smooth muscle LncRNA, Inducer of Differentiation (MYOSLID), was activated by MYOCD and selectively expressed in VSMCs. MYOSLID was a direct transcriptional target of both MYOCD/serum response factor and transforming growth factor-ß/SMAD pathways. Functional studies revealed that MYOSLID promotes VSMC differentiation and inhibits VSMC proliferation. MYOSLID showed reduced expression in failed human arteriovenous fistula samples compared with healthy veins. Although MYOSLID did not affect gene expression of transcription factors, such as serum response factor and MYOCD, its depletion in VSMCs disrupted actin stress fiber formation and blocked nuclear translocation of MYOCD-related transcription factor A (MKL1). Finally, loss of MYOSLID abrogated transforming growth factor-ß1-induced SMAD2 phosphorylation. CONCLUSIONS: We have demonstrated that MYOSLID, the first human VSMC-selective and serum response factor/CArG-dependent lncRNA, is a novel modulator in amplifying the VSMC differentiation program, likely through feed-forward actions of both MKL1 and transforming growth factor-ß/SMAD pathways.


Assuntos
Diferenciação Celular , Desenvolvimento Muscular , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , RNA Longo não Codificante/metabolismo , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo , Transporte Ativo do Núcleo Celular , Derivação Arteriovenosa Cirúrgica , Proliferação de Células , Células Cultivadas , Vasos Coronários/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Proteínas Nucleares/genética , Fenótipo , Fosforilação , RNA Longo não Codificante/genética , Fator de Resposta Sérica/genética , Transdução de Sinais , Proteína Smad2/metabolismo , Fibras de Estresse/metabolismo , Fatores de Tempo , Transativadores/genética , Transcrição Gênica , Transfecção , Fator de Crescimento Transformador beta1/metabolismo , Vasoconstrição
9.
Adv Exp Med Biol ; 967: 71-81, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29047082

RESUMO

Pulmonary hypertension (PH) is characterized by increased vasoconstriction and smooth muscle cell hyperplasia driving pathological vascular remodeling of arterial vessels. In this short review, we discuss the primary source of reactive oxygen species (ROS) and nitric oxide (NO) relevant to PH and the mechanism by which dysregulation of their production contributes to PH. Specifically, hypoxia-induced PH is associated with diminished endothelial nitric oxide synthase (eNOS)-derived NO production and increased production of superoxide (O2•-) through eNOS uncoupling and defective mitochondrial respiration. This drives the inhibition of the NO/soluble guanylate cyclase (sGC) pathway and activation of the transcription factor hypoxia-inducible factor-1α (HIF-1α) with consequential dysregulation of the pulmonary vasculature. Therapeutics aimed at increasing NO or cGMP bioavailabilities are amenable to hypoxia disease-induced PH. Similarly, strategies targeting HIF-1α are now considered. Overall, pulmonary hypertension including hypoxia-induced PH offers unique opportunities for the rational development of therapeutics centered on modulating redox signaling.


Assuntos
Hipertensão Pulmonar/metabolismo , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Animais , Humanos , Hipertensão Pulmonar/fisiopatologia , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Superóxidos/metabolismo
10.
Allergy Asthma Proc ; 35(3): 241-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24801467

RESUMO

Measurement of fractional nitric oxide concentration in exhaled breath (FENO) is a simple, noninvasive method to evaluate eosinophilic airway inflammation. Nitric oxide (NO) arising from peripheral small airways/alveoli (alveolar NO concentration [CalvNO]) can be estimated using multiple flow rates and a two-compartment model of the airways and alveoli. Omalizumab, a monoclonal anti-IgE antibody, is approved for the treatment of allergic asthma and also has been shown to decrease FENO levels. This study investigates the effects of omalizumab, when added to an inhaled corticosteroid (ICS) ± long-acting beta-adrenergic agonist (LABA) treatment, on peripheral small airway/alveolar inflammation reflected by FENO measurements at higher flow rates. We hypothesized that compared with placebo, omalizumab would decrease CalvNO levels in asthmatic patients on ICS ± LABA. Forty-two patients with moderate-to-severe asthma were randomly assigned 2:1 to either omalizumab (n = 29) or placebo treatment (n = 13) for 16 weeks. Selection criteria included moderate-to-severe asthmatic patients on an ICS ± LABA, positive skin test to one or more perennial allergen, screening FENO of >13 ppb, and a baseline IgE of 30-700 IU/mL. FENO measured at multiple flow rates was used to calculate CalvNO over the course of 16 weeks. FENO levels decrease with increasing flow rates (p < 0.05 repeated measures ANOVA) but no differences between the placebo and treatment groups in overall CalvNO levels or in the changes of CalvNO with time were found. Omalizumab did not lower the CalvNO, which could have been caused by the initial low CalvNO in this asthmatic population. The model used may not be completely sufficient and/or sensitive enough to detect small changes in CalvNO.


Assuntos
Antiasmáticos/uso terapêutico , Anticorpos Anti-Idiotípicos/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Asma/diagnóstico , Asma/tratamento farmacológico , Expiração , Óxido Nítrico/análise , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Omalizumab , Testes de Função Respiratória , Índice de Gravidade de Doença , Testes Cutâneos , Resultado do Tratamento , Adulto Jovem
11.
Oncogene ; 43(9): 650-667, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38184712

RESUMO

Transient early endosome (EE)-mitochondria interactions can mediate mitochondrial iron translocation, but the associated mechanisms are still elusive. We showed that Divalent Metal Transporter 1 (DMT1) sustains mitochondrial iron translocation via EE-mitochondria interactions in triple-negative MDA-MB-231, but not in luminal A T47D breast cancer cells. DMT1 silencing increases labile iron pool (LIP) levels and activates PINK1/Parkin-dependent mitophagy in MDA-MB-231 cells. Mitochondrial bioenergetics and the iron-associated protein profile were altered by DMT1 silencing and rescued by DMT1 re-expression. Transcriptomic profiles upon DMT1 silencing are strikingly different between 2D and 3D culture conditions, suggesting that the environment context is crucial for the DMT1 knockout phenotype observed in MDA-MB-231 cells. Lastly, in vivo lung metastasis assay revealed that DMT1 silencing promoted the outgrowth of lung metastatic nodules in both human and murine models of triple-negative breast cancer cells. These findings reveal a DMT1-dependent pathway connecting EE-mitochondria interactions to mitochondrial iron translocation and metastatic fitness of breast cancer cells.


Assuntos
Neoplasias da Mama , Ferro , Animais , Feminino , Humanos , Camundongos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Endossomos/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Mitofagia
12.
Nitric Oxide ; 28: 57-64, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23099296

RESUMO

Recent studies indicate the formation of protein nitrosamines in vivo and tryptophan residues in proteins might represent important targets of nitrosative and oxidative stress. In the present work, we examined the mechanism by which xanthine oxidase (XO) denitrosates N-nitroso Trp residues and determined the applicability of the reactions involved to the detection of nitrosated Trp residues by tri-iodide-based chemiluminescence. We found that - in addition to superoxide - denitrosation of N-acetyl-nitroso Trp (NANT) by hypoxanthine and XO occurred via the intermediacy of uric acid. Zero-order dependence of NANT decay rate with uric acid was achieved with increasing concentrations of uric acid (k(0)∼6.0×10(-4)s(-1)) and generated nitric oxide. In contrast, S-nitrosoglutathione and nitrosyl-myoglobin were stable in the presence of uric acid. NANT decomposition by uric acid could be reproducibly measured using the tri-iodide-based chemiluminescence assay in the presence of excess nitrite upon pre-treatment with acidified sulfanilamide. N-nitrosated albumin was sensitive to uric acid-induced decomposition only after proteolytic degradation. In conclusion, XO decomposes nitrosated Trp through superoxide and uric acid pathways and in the case of uric acid generates free nitric oxide. Site-specificity of this reaction may possibly be used in combination with the tri-iodide-based chemiluminescence assay to discern between nitrosated Trp, S-nitrosothiols, and nitrosylated heme proteins.


Assuntos
Nitrosaminas/química , Nitrosaminas/metabolismo , Superóxidos/metabolismo , Triptofano/análogos & derivados , Ácido Úrico/metabolismo , Xantina Oxidase/metabolismo , Óxido Nítrico/análise , Óxido Nítrico/biossíntese , Triptofano/química , Triptofano/metabolismo
13.
J Asthma ; 50(1): 64-70, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23098359

RESUMO

OBJECTIVE: The long-acting ß2-agonist salmeterol in combination with the corticosteroid fluticasone propionate is used in clinical practice for the treatment of mild persistent asthma. Although the effect of fluticasone propionate alone in asthmatic patients is well documented, the effect of fluticasone propionate/salmeterol (FSC) combination therapy on airway inflammation and airway hyperresponsiveness (AHR) is not well characterized. Thus, we evaluated AHR, exhaled nitric oxide (FE(NO)), and nitrite and nitrate in exhaled breath condensates (EBCs) from mild persistent asthmatic patients treated with a low-dose FSC (100/50). METHODS: In this open label study, 18 mild persistent, steroid-naïve asthmatics (age, 22-62 years, forced expiratory volume in 1 s (FEV(1)) > 70% predicted, provocative dose resulting in 20% reduction (PD(20)) < 10 mg/mL) were treated with FSC 100/50 for 4 weeks. PD(20) to methacholine, FEV(1), FE(NO), and EBC nitrite and nitrate was measured before and after treatment. RESULTS: After 4 weeks of therapy with FSC 100/50, FE(NO) decreased from 74 ppb (SD = 37) to 34 ppb (SD = 15) (p < .001). FEV(1) (% predicted) increased from 89.4 (SD = 10.7) to 93.3 (SD = 9.5) (p < .01). The PD(20) for methacholine increased from 3.0 (±3.2) to 10.3 (±8.4) mg/mL (p < .01) in 3 of 18 patients reaching the maximum allowable dose (25 mg/mL). FE(NO) correlated with the log of the methacholine dose. There was no statistically significant change in EBC nitrite and nitrate before and after treatment. CONCLUSIONS: Treatment of mild persistent, steroid-naïve asthmatics with low-dose combination therapy is effective in rapidly reducing airway inflammation and AHR. Our results suggest different metabolic origins for nitrite, nitrate, and FE(NO) in this group of patients.


Assuntos
Albuterol/análogos & derivados , Androstadienos/administração & dosagem , Asma/tratamento farmacológico , Asma/metabolismo , Broncodilatadores/administração & dosagem , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Adulto , Albuterol/administração & dosagem , Testes Respiratórios , Testes de Provocação Brônquica , Estudos Transversais , Combinação de Medicamentos , Combinação Fluticasona-Salmeterol , Humanos , Modelos Lineares , Pessoa de Meia-Idade , Adulto Jovem
14.
bioRxiv ; 2023 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-37034694

RESUMO

The oxidant hydrogen peroxide serves as a signaling molecule that alters many aspects of cardiovascular functions. Recent studies suggest that cytoglobin - a hemoglobin expressed in the vasculature - may promote electron transfer reactions with proposed functions in hydrogen peroxide decomposition. Here, we determined the extent to which cytoglobin regulates intracellular hydrogen peroxide and established mechanisms. We found that cytoglobin decreased the hyperoxidation of peroxiredoxins and maintained the activity of peroxiredoxin 2 following challenge with exogenous hydrogen peroxide. Cytoglobin promoted a reduced intracellular environment and facilitated the reduction of the thiol-based hydrogen peroxide sensor Hyper7 after bolus addition of hydrogen peroxide. Cytoglobin also limited the inhibitory effect of hydrogen peroxide on glycolysis and reversed the oxidative inactivation of the glycolytic enzyme GAPDH. Our results indicate that cytoglobin in cells exists primarily as oxyferrous cytoglobin (CygbFe 2+ -O 2 ) with its cysteine residues in the reduced form. We found that the specific substitution of one of two cysteine residues on cytoglobin (C83A) inhibited the reductive activity of cytoglobin on Hyper7 and GAPDH. Carotid arteries from cytoglobin knockout mice were more sensitive to glycolytic inhibition by hydrogen peroxide than arteries from wildtype mice. Together, these results support a role for cytoglobin in regulating intracellular redox signals associated with hydrogen peroxide through oxidation of its cysteine residues, independent of hydrogen peroxide reaction at its heme center.

15.
Redox Biol ; 65: 102838, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37573836

RESUMO

Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2.


Assuntos
Globinas , Proteína HMGB2 , Animais , Camundongos , Citoglobina/genética , Dano ao DNA , Globinas/genética , Globinas/metabolismo , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Fatores de Transcrição/genética
16.
bioRxiv ; 2023 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-37214992

RESUMO

Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2.

17.
Circ Res ; 107(8): 975-83, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20724704

RESUMO

RATIONALE: Vascular smooth muscle cell (SMC) migration is an important pathological process in several vascular occlusive diseases, including atherosclerosis and restenosis, both of which are accelerated by diabetes mellitus. OBJECTIVE: To determine the mechanisms of abnormal vascular SMC migration in type 2 diabetes, the obese Zucker rat (ZO), a model of obesity and insulin resistance, was studied. METHODS AND RESULTS: In culture, ZO aortic SMCs showed a significant increase in Nox4 mRNA and protein levels compared with the control lean Zucker rat (ZL). The sarco-/endoplasmic reticulum Ca(2+) ATPase (SERCA) nitrotyrosine-294,295 and cysteine-674 (C674)-SO(3)H were increased in ZO SMCs, indicating oxidant stress. Unlike ZL SMC, nitric oxide (NO) failed to inhibit serum-induced SMC migration in ZO. Transfection of Nox4 small interference RNA or overexpression of SERCA2b wild type, but not C674S mutant SERCA, restored the response to NO. Knockdown of Nox4 also decreased SERCA oxidation in ZO SMCs. In addition, transforming growth factor-ß1 via Smad2 was necessary and sufficient to upregulate Nox4, oxidize SERCA, and block the antimigratory action of NO in ZO SMCs. Corresponding to the results in cultured SMCs, immunohistochemistry confirmed that Nox4 and SERCA C674-SO(3)H were significantly increased in ZO aorta. After common carotid artery injury, knockdown of Nox4 by adenoviral Nox4 short hairpin RNA decreased Nox4 and SERCA C674-SO(3)H staining and significantly decreased injury-induced neointima. CONCLUSION: These studies indicate that the upregulation of Nox4 by transforming growth factor-ß1 in ZO SMCs is responsible for the impaired response to NO by a mechanism involving the oxidation of SERCA C674. Knockdown of Nox4 inhibits oxidation of SERCA, as well as neointima formation, after ZO common carotid artery injury.


Assuntos
Lesões das Artérias Carótidas/fisiopatologia , NADPH Oxidases/genética , Óxido Nítrico/metabolismo , Estado Pré-Diabético/fisiopatologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Fator de Crescimento Transformador beta1/farmacologia , Animais , Aorta/citologia , Lesões das Artérias Carótidas/metabolismo , Artéria Carótida Primitiva/fisiopatologia , Movimento Celular/fisiologia , Células Cultivadas , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , Obesidade/metabolismo , Obesidade/fisiopatologia , Estresse Oxidativo/fisiologia , Estado Pré-Diabético/metabolismo , Ratos , Ratos Zucker , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Túnica Íntima/citologia , Túnica Íntima/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
18.
Lung ; 189(4): 295-303, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21667259

RESUMO

The aim of this study was to evaluate whether underlying respiratory disease may be revealed by offline fractional exhaled nitric oxide (FE(NO)) testing among a cohort of New York State (NYS) World Trade Center (WTC) responders in comparison with a control group of similar but unexposed NYS employees, 6 years post-9/11. Participants (92 exposed, 141 unexposed) provided two breath samples that were collected in Mylar bags and sent to a central laboratory for FE(NO) testing. Participants also completed a brief questionnaire. Ambient air pollution was characterized using particulate matter (PM(2.5)) and ozone concentration data from the NYS Department of Environmental Conservation air-monitoring sites closest to each testing site for each day of sample collection. WTC exposure did not appear to be associated with elevated FE(NO) concentrations. FE(NO) concentrations were higher on days with elevated levels of PM(2.5) (≥ 35 µg/m³) and ozone (≥ 0.08 ppm). FE(NO) concentrations were higher in men and lower in smokers. Our results do not suggest an association between WTC exposure and elevated FE(NO) concentrations, 6 years post-9/11, in this moderately exposed cohort of responders. Results do suggest that FE(NO) concentrations were elevated in relation to higher levels of ambient air pollutants. Our results also offer useful reference values for future research involving FE(NO) testing. This study demonstrates that offline FE(NO) testing is a useful method for epidemiological studies requiring collection of samples in the field, potentially over a broad geographic area.


Assuntos
Poluentes Atmosféricos/toxicidade , Expiração , Pneumopatias/induzido quimicamente , Pneumopatias/diagnóstico , Óxido Nítrico/análise , Exposição Ocupacional , Ataques Terroristas de 11 de Setembro , Adulto , Estudos de Coortes , Socorristas , Feminino , Humanos , Pneumopatias/epidemiologia , Masculino , Cidade de Nova Iorque/epidemiologia , Ozônio/toxicidade , Material Particulado/toxicidade , Fumar/epidemiologia
19.
Am J Physiol Cell Physiol ; 298(5): C993-1005, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20107038

RESUMO

We recently demonstrated that thapsigargin-induced passive store depletion activates Ca(2+) entry in vascular smooth muscle cells (VSMC) through stromal interaction molecule 1 (STIM1)/Orai1, independently of transient receptor potential canonical (TRPC) channels. However, under physiological stimulations, despite the ubiquitous depletion of inositol 1,4,5-trisphosphate-sensitive stores, many VSMC PLC-coupled agonists (e.g., vasopressin and endothelin) activate various store-independent Ca(2+) entry channels. Platelet-derived growth factor (PDGF) is an important VSMC promigratory agonist with an established role in vascular disease. Nevertheless, the molecular identity of the Ca(2+) channels activated by PDGF in VSMC remains unknown. Here we show that inhibitors of store-operated Ca(2+) entry (Gd(3+) and 2-aminoethoxydiphenyl borate at concentrations as low as 5 microM) prevent PDGF-mediated Ca(2+) entry in cultured rat aortic VSMC. Protein knockdown of STIM1, Orai1, and PDGF receptor-beta (PDGFRbeta) impaired PDGF-mediated Ca(2+) influx, whereas Orai2, Orai3, TRPC1, TRPC4, and TRPC6 knockdown had no effect. Scratch wound assay showed that knockdown of STIM1, Orai1, or PDGFRbeta inhibited PDGF-mediated VSMC migration, but knockdown of STIM2, Orai2, and Orai3 was without effect. STIM1, Orai1, and PDGFRbeta mRNA levels were upregulated in vivo in VSMC from balloon-injured rat carotid arteries compared with noninjured control vessels. Protein levels of STIM1 and Orai1 were also upregulated in medial and neointimal VSMC from injured carotid arteries compared with noninjured vessels, as assessed by immunofluorescence microscopy. These results establish that STIM1 and Orai1 are important components for PDGF-mediated Ca(2+) entry and migration in VSMC and are upregulated in vivo during vascular injury and provide insights linking PDGF to STIM1/Orai1 during neointima formation.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Actinas , Animais , Canais de Cálcio/genética , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Masculino , Glicoproteínas de Membrana/genética , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Proteína ORAI1 , Ratos , Transdução de Sinais , Molécula 1 de Interação Estromal
20.
Redox Biol ; 32: 101468, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32087552

RESUMO

Cytoglobin is an evolutionary ancient hemoglobin with poor functional annotation. Rather than constrained to penta coordination, cytoglobin's heme iron may exist either as a penta or hexacoordinated arrangement when exposed to different intracellular environments. Two cysteine residues at the surface of the protein form an intramolecular disulfide bond that regulates iron coordination, ligand binding, and peroxidase activity. Overall, biochemical results do not support a role for cytoglobin as a direct antioxidant enzyme that scavenges hydrogen peroxide because the rate of the reaction of cytoglobin with hydrogen peroxide is several orders of magnitude slower than metal and thiol-based peroxidases. Thus, alternative substrates such as fatty acids have been suggested and regulation of nitric oxide bioavailability through nitric oxide dioxygenase and nitrite reductase activities has received experimental support. Cytoglobin is broadly expressed in connective, muscle, and nervous tissues. Rational for differential cellular distribution is poorly understood but inducibility in response to hypoxia is one of the most established features of cytoglobin expression with regulation through the transcription factor hypoxia-inducible factor (HIF). Phenotypic characterization of cytoglobin deletion in the mouse have indicated broad changes that include a heightened inflammatory response and fibrosis, increase tumor burden, cardiovascular dysfunction, and hallmarks of senescence. Some of these changes might be reversed upon inhibition of nitric oxide synthase. However, subcellular and molecular interactions have been seldom characterized. In addition, specific molecular mechanisms of action are still lacking. We speculate that cytoglobin functionality will extend beyond nitric oxide handling and will have to encompass indirect regulatory antioxidant and redox sensing functions.


Assuntos
Globinas , Peroxidase , Animais , Citoglobina , Globinas/genética , Camundongos , Oxigenases , Peroxidases
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa