Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gen Physiol Biophys ; 39(6): 545-555, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33226363

RESUMO

Cardiovascular disease (CVD) states are associated with endothelial dysfunction (ED) and increased production of ROS in endothelial cells. The present study aimed to explore the protective effects of antioxidant protein peroxiredoxin 6 (PRDX6) on angiotensin II (AngII)­induced human umbilical vein endothelial cell (HUVEC) dysfunction. To investigate cell viability, levels of inflammatory molecules and proteins were assayed using the CCK-8 assay and evaluated by ELISA and Western blot. NO and ROS levels were determined by Griess assay and the fluorescent probe DCFH-DA. Cell migration capacity was assessed by Transwell assay. AngII decreased cell viability and PRDX6, upregulated the expression levels of TNF-α, IL-6, IL-1ß, LDH and MDA, stimulated ROS production, and reduced NO synthase, the expressions of eNOS, MnSOD, ICAM-1, VCAM-1, and activated the MAPK family of signaling proteins. However, the stimulatory effects of AngII on HUVECs were remarkably suppressed by PRDX6. Furthermore, mercaptosuccinate (MS; PRDX6 inhibitor) had similar effects as AngII in aggravating HUVECs damage. Conversely, these adverse events caused by AngII and MS were obviously reversed by ML3404 and SP600125. The present study indicated that PRDX6 overexpression inactivated p38 MAPK and JNK pathway through decrease AngII-induced inflammation, oxidative stress and endothelial dysfunction leading to attenuation of endothelial cell damage.


Assuntos
Angiotensina II , Células Endoteliais da Veia Umbilical Humana/fisiologia , Estresse Oxidativo , Peroxirredoxina VI/fisiologia , Angiotensina II/farmacologia , Antioxidantes/fisiologia , Citocinas/fisiologia , Humanos , Inflamação/induzido quimicamente , Espécies Reativas de Oxigênio/metabolismo
2.
Biochemistry (Mosc) ; 84(6): 637-643, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31238863

RESUMO

Taking into account a special role of pancreatic ß-cells in the development of diabetes mellitus, the effects of peroxiredoxin 6 (Prx6) on the viability and functional activity of rat insulinoma RIN-m5F ß-cells were studied under diabetes-simulating conditions. For this purpose, the cells were cultured at elevated glucose concentrations or in the presence of pro-inflammatory cytokines (TNF-α and IL-1) known for their special role in the cytotoxic autoimmune response in diabetes. It was found that the increased glucose concentration of 23-43 mM caused death of 20-60% ß-cells. Prx6 added to cells significantly reduced the level of reactive oxygen species and protected the RIN-m5F ß-cells from hyperglycemia, reducing the death of these cells by several fold. A measurement of insulin secretion by the RIN-m5F ß-cells showed a significant stimulatory effect of Prx6 on the insulin-producing activity of pancreatic ß-cells. It should be noted that the stimulatory activity of Prx6 was detected during culturing the cells under both normal and unfavorable conditions. The regulation of the NF-κB signaling cascade could be one of the mechanisms of Prx6 action on ß-cells, in particular, through activation of RelA/p65 phosphorylation at Ser536.


Assuntos
Citocinas/toxicidade , Glucose/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Peroxirredoxina VI/fisiologia , Animais , Morte Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Citocinas/metabolismo , Glucose/metabolismo , Mediadores da Inflamação/metabolismo , Insulina/biossíntese , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1/metabolismo , NF-kappa B/metabolismo , Fosforilação , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
3.
Oxid Med Cell Longev ; 2019: 9685607, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31949886

RESUMO

Insulin action and often glucose-stimulated insulin secretion are reduced in obesity. In addition, the excessive intake of lipids increases oxidative stress leading to overt type 2 diabetes mellitus (T2DM). Among the antioxidative defense systems, peroxiredoxin 6 (PRDX6) is able to reduce H2O2 and short chain and phospholipid hydroperoxides. Increasing evidences suggest that PRDX6 is involved in the pathogenesis of atherosclerosis and T2DM, but its role in the etiopathology of obesity and its complications is still not known. Therefore, in the present study, we sought to investigate this association by using PRDX6 knockout mice (PRDX6-/-). Metabolic parameters, like carbon dioxide (VCO2) production, oxygen consumption (VO2), and the respiratory exchange ratio (RER), were determined using metabolic cages. Intraperitoneal insulin and glucose tolerance tests were performed to evaluate insulin sensitivity and glucose tolerance, respectively. Liver and pancreas histochemical analyses were also evaluated. The expression of enzymes involved in lipid and glucose metabolism was analyzed by real-time PCR. Following 24 weeks of high-fat-diet (HFD), PRDX6-/- mice showed weight gain and higher food and drink intake compared to controls. VO2 consumption and VCO2 production decreased in PRDX6-/- mice, while the RER was lower than 0.7 indicating a prevalent lipid metabolism. PRDX6-/- mice fed with HFD showed a further deterioration on insulin sensitivity and glucose-stimulated insulin secretion. Furthermore, in PRDX6-/- mice, insulin did not suppress adipose tissue lipolysis with consequent hepatic lipid overload and higher serum levels of ALT, cholesterol, and triglycerides. Interestingly, in PRDX6-/- mice, liver and adipose tissue were associated with proinflammatory gene upregulation. Finally, PRDX6-/- mice showed a higher rate of nonalcoholic steatohepatitis (NASH) compared to control. Our results suggest that PRDX6 may have a functional and protective role in the development of obesity-related metabolic disorders such as liver diseases and T2DM and may be considered a potential therapeutic target against these illnesses.


Assuntos
Adipogenia , Glicemia/metabolismo , Células Secretoras de Insulina/patologia , Insulina/metabolismo , Metabolismo dos Lipídeos , Hepatopatia Gordurosa não Alcoólica/patologia , Peroxirredoxina VI/fisiologia , Animais , Antioxidantes , Dieta Hiperlipídica/efeitos adversos , Feminino , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/complicações
4.
Aging (Albany NY) ; 10(9): 2284-2315, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30215601

RESUMO

Progressive deterioration of antioxidant response in aging is a major culprit in the initiation of age-related pathobiology induced by oxidative stress. We previously reported that oxidative stress leads to a marked reduction in transcription factor Sp1 and its mediated Prdx6 expression in lens epithelial cells (LECs) leading to cell death. Herein, we examined how Sp1 activity goes awry during oxidative stress/aging, and whether it is remediable. We found that Sp1 is hyper-Sumoylated at lysine (K) 16 residue in aging LECs. DNA binding and promoter assays revealed, in aging and oxidative stress, a significant reduction in Sp1 overall binding, and specifically to Prdx6 promoter. Expression/overexpression assay revealed that the observed reduction in Sp1-DNA binding activity was connected to its hyper-Sumoylation due to increased reactive oxygen species (ROS) and Sumo1 levels, and reduced levels of Senp1, Prdx6 and Sp1. Mutagenesis of Sp1 at K16R (arginine) residue restored steady-state, and improved Sp1-DNA binding activity and transactivation potential. Extrinsic expression of Sp1K16R increased cell survival and reduced ROS levels by upregulating Prdx6 expression in LECs under aging/oxidative stress, demonstrating that Sp1K16R escapes the aberrant Sumoylation processes. Intriguingly, the deleterious processes are reversible by the delivery of Sumoylation-deficient Prdx6, an antioxidant, which would be a candidate molecule to restrict aging pathobiology.


Assuntos
Estresse Oxidativo , Peroxirredoxina VI/fisiologia , Fator de Transcrição Sp1/fisiologia , Sumoilação , Envelhecimento , Animais , Morte Celular , Células Cultivadas , DNA/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Peroxirredoxina VI/genética , Espécies Reativas de Oxigênio/metabolismo , Ativação Transcricional
5.
Oncotarget ; 6(25): 20875-84, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26327204

RESUMO

Multiple sclerosis (MS) is a complex disease with an unknown etiology and has no effective medications despite extensive research. Antioxidants suppress oxidative damages which are implicated in the pathogenesis of MS. In this study, we showed that the expression of an antioxidant protein peroxiredoxin 6 (PRDX6) is markedly increased in spinal cord of mice with experimental autoimmune encephalomyelitis (EAE) compared to other PRDXs. PRDX6 transgenic (Tg) mice displayed a significant decrease in clinical severity and attenuated demyelination in EAE compared to wide type mice. The increased PRDX6 expression in astrocytes of EAE mice and MS patients reduced MMP9 expression, fibrinogen leakage, chemokines, and free radical stress, leading to reduction in blood-brain-barrier (BBB) disruption, peripheral immune cell infiltration, and neuroinflammation. Together, these findings suggest that PRDX6 expression may represent a therapeutic way to restrict inflammation in the central nervous system and potentiate oligodendrocyte survival, and suggest a new molecule for neuroprotective therapies in MS.


Assuntos
Barreira Hematoencefálica/fisiologia , Esclerose Múltipla/fisiopatologia , Peroxirredoxina VI/fisiologia , Animais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Perfilação da Expressão Gênica , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla/imunologia , Óxido Nítrico/metabolismo , Oligodendroglia/citologia , Peroxirredoxina VI/imunologia , Proteoma , Espécies Reativas de Oxigênio/metabolismo
6.
FEBS J ; 281(15): 3357-81, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24910119

RESUMO

Loss of the cytoprotective protein peroxiredoxin 6 (Prdx6) in cells that are aging or under oxidative stress is known to be linked to the pathobiology of many age-related diseases. However, the mechanism by which Prdx6 activity goes awry is largely unknown. Using Prdx6-deficient (Prdx6(-/-) ) cells as a model for aging or redox active cells, human/mouse lens epithelial cells (LECs) facing oxidative stress and aging lenses, we found a significant increase in the levels of small ubiquitin-like modifier (Sumo)1 conjugates. These cells displayed increased levels of Sumo1 and reduced the expression of Prdx6. Specifically, we observed that Prdx6 is a target for aberrant sumoylation signaling, and that Sumo1 modification reduces its cellular abundance. LECs overexpressing Sumo1 showed reduced expression and activity of Prdx6 and its transactivator specificity protein 1 (Sp1), mRNA and protein with increased levels of reactive oxygen species; those cells were vulnerable to oxidative stress-induced cell death. A significant reduction in Prdx6, Sp1 protein and mRNA expression was observed in redox active Prdx6(-/-) cells and in aging lenses/LECs. The reduction was correlated with increased expression of Sumo1 and enrichment of the inactive form (dimeric) of Sumo-specific protease (Senp)1. Experiments with Sumo1-fused Prdx6 and Prdx6 promoter-linked to chloramphenicol acetyltransferase reporter gene constructs indicated that Sumo1 dysregulated Prdx6 activity by reducing its abundance and attenuating its transcription; in contrast, the delivery of Senp1 or Prdx6 reversed the process. The data show that reactive oxygen species-evoked aberrant sumoylation signaling affects Prdx6 activity by reducing Prdx6 abundance, as well as transcription. The findings of the present study may provide a foundation for a strategy to repair deleterious oxidative signaling generated by a reduced activity of Prdx6.


Assuntos
Peroxirredoxina VI/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Proteína SUMO-1/metabolismo , Sumoilação , Transcrição Gênica , Envelhecimento , Animais , Sobrevivência Celular , Células Cultivadas , Cisteína Endopeptidases , Endopeptidases/genética , Endopeptidases/metabolismo , Repressão Enzimática , Estabilidade Enzimática , Humanos , Camundongos , Estresse Oxidativo , Transdução de Sinais , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo
7.
Free Radic Biol Med ; 61: 453-63, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23643677

RESUMO

This study compared lung tumor growth in PRDX6-overexpressing transgenic (Tg) mice and normal mice. These mice expressed elevated levels of PRDX6 mRNA and protein in multiple tissues. In vivo, Tg mice displayed a greater increase in the growth of lung tumor compared with normal mice. Glutathione peroxidase and calcium-independent phospholipase 2 (iPLA2) activities in tumor tissues of Tg mice were much higher than in tumor tissues of normal mice. Higher tumor growth in PRDX6-overexpressing Tg mice was associated with an increase in activating protein-1 (AP-1) DNA-binding activity. Moreover, expression of proliferating cell nuclear antigen, Ki67, vascular endothelial growth factor, c-Jun, c-Fos, metalloproteinase-9, cyclin-dependent kinases, and cyclins was much higher in the tumor tissues of PRDX6-overexpressing Tg mice than in tumor tissues of normal mice. However, the expression of apoptotic regulatory proteins including caspase-3 and Bax was slightly less in the tumor tissues of normal mice. In tumor tissues of PRDX6-overexpressing Tg mice, activation of mitogen-activated protein kinases (MAPKs) was much higher than in normal mice. In cultured lung cancer cells, PRDX6 siRNA suppressed glutathione peroxidase and iPLA2 activities and cancer cell growth, but the enforced overexpression of PRDX6 increased cancer cell growth associated with their increased activities. In vitro, among the tested MAPK inhibitors, c-Jun NH2-terminal kinase (JNK) inhibitor clearly suppressed the growth of lung cancer cells and AP-1 DNA binding, glutathione peroxidase activity, and iPLA2 activity in normal and PRDX6-overexpressing lung cancer cells. These data indicate that overexpression of PRDX6 promotes lung tumor growth via increased glutathione peroxidase and iPLA2 activities through the upregulation of the AP-1 and JNK pathways.


Assuntos
Neoplasias Pulmonares/patologia , Peroxirredoxina VI/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , DNA/metabolismo , Glutationa Peroxidase/análise , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Peroxirredoxina VI/análise , Fosfolipases A2/metabolismo , Fator de Transcrição AP-1/metabolismo
9.
Free Radic Biol Med ; 51(2): 552-61, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21586322

RESUMO

The disruption of redox control, i.e., oxidative stress, is one of the most destructive causes of ischemia-reperfusion (IR) injury. Thioredoxin (Trx) family proteins play a major role in the cellular response to oxidative stress. Here, we systematically investigated the levels and tissue distribution of 15 members of this family (Trx and TrxR 1 and 2, Nrx, Prx 1-6, and Grx 1-3 and 5) in mouse kidneys after induction of IR by comparing control, clamped, and contralateral organs. After IR, levels of various redoxins were quantified. Immunohistochemical analysis revealed segment-specific alterations induced by the ischemic insult. Grx2, Prx3, and Prx6 were highly expressed in proximal tubule cells. Overexpression of these proteins in HEK293 and HeLa cells subjected to hypoxia and reoxygenation revealed higher survival and proliferation rates and lower oxidative damage compared to controls. Furthermore, we report for the first time the accumulation of Grx1 at the apical side of distal convoluted cells and the specific secretion of Grx1 into the urine after IR. The differences in both the basal equipment and the segment-specific responses of the antioxidant proteins may contribute to the distinct susceptibilities and regeneration processes of the various segments of the nephron to the IR insult.


Assuntos
Glutarredoxinas/fisiologia , Rim/irrigação sanguínea , Peroxirredoxina VI/fisiologia , Peroxirredoxinas/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/enzimologia
10.
Crit Care Med ; 39(4): 756-64, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21200322

RESUMO

OBJECTIVE: To investigate the role and signaling pathway of peroxiredoxin 6, a newly identified peroxidase, in lipopolysaccharide-induced acute lung injury. DESIGN: Prospective, randomized, controlled study. SETTING: Research laboratory. SUBJECTS: Peroxiredoxin 6 (-/-) and wild-type C57BL/6 mice. INTERVENTIONS: Wild-type or peroxiredoxin 6 (-/-) mice were challenged by intratracheal instillation of lipopolysaccharide (5 mg/kg) for 4 hrs or 24 hrs for lung injury measurement. In other studies, peritoneal macrophages, isolated from wild-type and peroxiredoxin 6 (-/-) mice, were preincubated in presence or absence of mitogen-activated protein kinases inhibitors for 30 mins before being stimulated with lipopolysaccharide (1 µg/mL) for 4 hrs. MEASUREMENTS AND MAIN RESULTS: Bronchoalveolar lavage myeloperoxidase activity and the lung injury score were significantly increased in peroxiredoxin 6 (-/-) mice compared with wild-type mice after lipopolysaccharide instillation at both 4 hrs and 24 hrs. Hydrogen peroxide and malondialdehyde levels, as well as nuclear factor-κB activities, tumor necrosis factor-α, interleukin-1ß, and matrix metalloproteinase-9 messenger RNA, protein concentration, and activities were significantly increased whereas total antioxidative capability was markedly decreased in lungs of peroxiredoxin 6 (-/-) mice compared with wild-type mice. In vitro studies showed intracellular reactive oxygen species levels and release of tumor necrosis factor-α, interleukin-1, and matrix metalloproteinase-9 were significantly increased in macrophages from peroxiredoxin 6 (-/-) mice compared with that from wild-type mice after lipopolysaccharide stimulation. Cytokines release was partially suppressed by extracellular signal-regulated kinase and c-Jun N-terminal kinase inhibitors, but not by the p38 mitogen-activated protein kinase inhibitor. CONCLUSIONS: Deletion of peroxiredoxin 6 exaggerates lipopolysaccharide-induced acute lung injury and inflammation with increased oxidative stress, inflammatory responses, and matrix degradation, all of which were partially dependent on nuclear factor-κB, extracellular signal-regulated kinase, and c-Jun N-terminal kinase pathways.


Assuntos
Lesão Pulmonar Aguda/etiologia , Peroxirredoxina VI/fisiologia , Lesão Pulmonar Aguda/fisiopatologia , Animais , Peróxido de Hidrogênio/análise , Interleucina-1beta/análise , Lipopolissacarídeos/farmacologia , Pulmão/química , Pulmão/efeitos dos fármacos , Macrófagos Peritoneais/química , Masculino , Malondialdeído/análise , Metaloproteinase 9 da Matriz/análise , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/análise , Estresse Oxidativo , Peroxirredoxina VI/deficiência , Espécies Reativas de Oxigênio/análise , Fator de Necrose Tumoral alfa/análise
11.
BMC Neurosci ; 11: 125, 2010 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-20923568

RESUMO

BACKGROUND: The ability to respond to changes in the extra-intracellular environment is prerequisite for cell survival. Cellular responses to the environment include elevating defense systems, such as the antioxidant defense system. Hypoxia-evoked reactive oxygen species (ROS)-driven oxidative stress is an underlying mechanism of retinal ganglion cell (RGC) death that leads to blinding disorders. The protein peroxiredoxin 6 (PRDX6) plays a pleiotropic role in negatively regulating death signaling in response to stressors, and thereby stabilizes cellular homeostasis. RESULTS: We have shown that RGCs exposed to hypoxia (1%) or hypoxia mimetic cobalt chloride display reduced expression of PRDX6 with higher ROS expression and activation of NF-κB. These cells undergo apoptosis, while cells with over-expression of PRDX6 demonstrate resistance against hypoxia-driven RGC death. The RGCs exposed to hypoxia either with 1% oxygen or cobalt chloride (0-400 µM), revealed ~30%-70% apoptotic cell death after 48 and 72 h of exposure. Western analysis and real-time PCR showed elevated expression of PRDX6 during hypoxia at 24 h, while PRDX6 protein and mRNA expression declined from 48 h onwards following hypoxia exposure. Concomitant with this, RGCs showed increased ROS expression and activation of NF-κB with IkB phosphorylation/degradation, as examined with H2DCF-DA and transactivation assays. These hypoxia-induced adverse reactions could be reversed by over-expression of PRDX6. CONCLUSION: Because an abundance of PRDX6 in cells was able to attenuate hypoxia-induced RGC death, the protein could possibly be developed as a novel therapeutic agent acting to postpone RGC injury and delay the progression of glaucoma and other disorders caused by the increased-ROS-generated death signaling related to hypoxia.


Assuntos
Hipóxia Celular/efeitos dos fármacos , Fármacos Neuroprotetores , Peroxirredoxina VI/farmacologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Cobalto/farmacologia , Genes Reporter , Proteínas I-kappa B/metabolismo , Marcação In Situ das Extremidades Cortadas , NF-kappa B/metabolismo , Peroxirredoxina VI/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
PLoS One ; 4(6): e6075, 2009 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-19562038

RESUMO

Oxidative stress plays a prominent role in the pathophysiology of cystic fibrosis (CF). Despite the presence of oxidative stress markers and a decreased antioxidant capacity in CF airway lining fluid, few studies have focused on the oxidant/antioxidant balance in CF cells. The aim of the current study was to investigate the cellular levels of reactive oxygen species (ROS), oxidative damage and enzymatic antioxidant defenses in the lung of Cftr-knockout mice in basal conditions and as a response to oxidative insult.The results show that endogenous ROS and lipid peroxidation levels are higher in Cftr(-/-) lung when compared to wild-type (Cftr(+/+)) in basal conditions, despite a strong enzymatic antioxidant response involving superoxide dismutases, glutathione peroxidases and peroxiredoxin 6 (Prdx6). The latter has the unique capacity to directly reduce membrane phospholipid hydroperoxides (PL-OOH). A dramatic increase in PL-OOH levels in Cftr(-/-) lung consecutive to in vivo oxidative challenge by paraquat (PQ) unmasks a susceptibility to phospholipid peroxidation. PQ strongly decreases Prdx6 expression in Cftr(-/-) mice compared to Cftr(+/+). Similar results were obtained after P. aeruginosa LPS challenge. Two-dimensional gel analysis of Prdx6 revealed one main molecular form in basal conditions and a PQ-induced form only detected in Cftr(+/+) lung. Mass spectrometry experiments suggested that, as opposed to the main basal form, the one induced by PQ is devoid of overoxidized catalytic Cys47 and could correspond to a fully active form that is not induced in Cftr(-/-) lung. These results highlight a constitutive redox imbalance and a vulnerability to oxidative insult in Cftr(-/-) lung and present Prdx6 as a key component in CF antioxidant failure. This impaired PL-OOH detoxification mechanism may enhance oxidative damage and stress-related signaling, contributing to an exaggerated inflammatory response in CF lung.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Pulmão/metabolismo , Peroxirredoxina VI/fisiologia , Fosfolipídeos/metabolismo , Animais , Antioxidantes/metabolismo , Catálise , Inflamação , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxidantes/metabolismo , Estresse Oxidativo , Peroxirredoxina VI/metabolismo , Espécies Reativas de Oxigênio , Transdução de Sinais
13.
J Cell Biochem ; 104(4): 1274-85, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18260127

RESUMO

We evaluated the antioxidant role of peroxiredoxin 6 (Prdx6) in primary lung alveolar epithelial type II cells (AEC II) that were isolated from wild type (WT), Prdx6-/-, or Prdx6 transgenic (Tg) overexpressing mice and exposed to H(2)O(2) at 50-500 microM for 1-24 h. Expression of Prdx6 in Tg AEC II was sevenfold greater than WT. Prdx6 null AEC II exposed to H(2)O(2) showed concentration-dependent cytotoxicity indicated by decreased "live/dead" cell ratio, increased propidium iodide (PI) staining, increased annexin V binding, increased DNA fragmentation by TUNEL assay, and increased lipid peroxidation by diphenylpyrenylphosphine (DPPP) fluorescence. Compared to Prdx6 null cells, oxidant-mediated damage was significantly less in WT AEC II and was least in Prdx6 Tg cells. Thus, Prdx6 functions as an antioxidant enzyme in mouse AEC II. Prdx6 has been shown previously to reduce phospholipid hydroperoxides and we postulate that this activity is a major mechanism for the effectiveness of Prdx6 as an antioxidant enzyme.


Assuntos
Células Epiteliais/metabolismo , Peroxirredoxina VI/fisiologia , Alvéolos Pulmonares/citologia , Animais , Antioxidantes , Apoptose , Sobrevivência Celular , Peróxido de Hidrogênio/farmacologia , Peroxidação de Lipídeos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos
14.
J Cell Biol ; 179(4): 747-60, 2007 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-18025307

RESUMO

Peroxiredoxin 6 (Prdx6) is a cytoprotective enzyme with largely unknown in vivo functions. Here, we use Prdx6 knockout mice to determine its role in UV protection and wound healing. UV-mediated keratinocyte apoptosis is enhanced in Prdx6-deficient mice. Upon skin injury, we observe a severe hemorrhage in the granulation tissue of knockout animals, which correlates with the extent of oxidative stress. At the ultrastructural level endothelial cells appear highly damaged, and their rate of apoptosis is enhanced. Knock-down of Prdx6 in cultured endothelial cells also increases their susceptibility to oxidative stress, thus confirming the sensitivity of this cell type to loss of Prdx6. Wound healing studies in bone marrow chimeric mice demonstrate that Prdx6-deficient inflammatory and endothelial cells contribute to the hemorrhage phenotype. These results provide insight into the cross-talk between hematopoietic and resident cells at the wound site and the role of reactive oxygen species in this interplay.


Assuntos
Peroxirredoxina VI/fisiologia , Pele/irrigação sanguínea , Pele/lesões , Animais , Apoptose/genética , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Vasos Sanguíneos/ultraestrutura , Células Cultivadas , Quimera/genética , Quimera/metabolismo , Células Endoteliais/patologia , Células Endoteliais/ultraestrutura , Endotélio Vascular/ultraestrutura , Feminino , Técnica Direta de Fluorescência para Anticorpo , Hemorragia/etiologia , Hemorragia/patologia , Humanos , Imuno-Histoquímica , Queratinócitos/enzimologia , Queratinócitos/efeitos da radiação , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Estresse Oxidativo , Peroxirredoxina VI/deficiência , Peroxirredoxina VI/genética , Espécies Reativas de Oxigênio/metabolismo , Raios Ultravioleta , Veias Umbilicais/citologia , Irradiação Corporal Total , Cicatrização/genética
15.
J Lipid Res ; 48(10): 2306-18, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652308

RESUMO

Peroxiredoxin 6 (Prdx6) is a bifunctional protein with glutathione peroxidase and phospholipase A(2) (PLA(2)) activities, and it alone among mammalian peroxiredoxins can hydrolyze phospholipids. After identifying a potential catalytic triad (S32, H26, D140) from the crystal structure, site-specific mutations were used to evaluate the role of these residues in protein structure and function. The S32A mutation increased Prdx6 alpha-helical content, whereas secondary structure was unchanged by mutation to H26A and D140A. Lipid binding by wild-type Prdx6 to negatively charged unilamellar liposomes showed an apparent rate constant of 11.2 x 10(6) M(-1) s(-1) and a dissociation constant of 0.36 microM. Both binding and PLA(2) activity were abolished in S32A and H26A; in D140A, activity was abolished but binding was unaffected. Overoxidation of the peroxidatic C47 had no effect on lipid binding or PLA(2) activity. Fluorescence resonance energy transfer from endogenous tryptophanyls to lipid probes showed binding of the phospholipid polar head in close proximity to S32. Thus, H26 is a site for interfacial binding to the liposomal surface, S32 has a key role in maintaining Prdx6 structure and for phospholipid substrate binding, and D140 is involved in catalysis. This putative catalytic triad plays an essential role for interactions of Prdx6 with phospholipid substrate to optimize the protein-substrate complex for hydrolysis.


Assuntos
Peroxirredoxina VI/fisiologia , Fosfolipases/metabolismo , Animais , Catálise , Dicroísmo Circular , Glutationa Peroxidase/metabolismo , Hidrólise , Modelos Biológicos , Modelos Moleculares , Conformação Molecular , Mutagênese Sítio-Dirigida , Mutação , Peroxirredoxina VI/metabolismo , Fosfolipídeos/química , Ligação Proteica , Estrutura Secundária de Proteína , Ratos , Proteínas Recombinantes/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...