Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 760
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
FASEB J ; 36(1): e22098, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34918390

RESUMO

Vascular calcification (VC), in which vascular smooth muscle cells (VSMCs) undergo differentiation and osteogenic transition, is a common complication of chronic kidney disease (CKD). Recent findings show that nuclear factor-erythroid-2-related factor 2 (NRF2) is an evolutionarily conserved antioxidant and beneficial in preventing vascular senescence and calcification. The roles of NRF2 in the initiation and progression of VC in CKD still need further investigation. CKD-associated VC model rats exhibited significant upregulation of NRF2, NAD(P)H: quinone oxidoreductase-1 (NQO1), osteogenic markers such as alkaline phosphatase (ALP), runt-related transcription factor-2 (RUNX2) and osteopontin (OPN), and ß-catenin compared to CKD rats. Immunohistochemistry further verified these results. In addition, rat aortic VSMCs were isolated and subjected to four treatments: normal control, phosphorus-induced (Pi), Pi + NRF2 activator DMF, and Pi + NRF2 inhibitor ML385. The reactive oxygen species (ROS) generation, malondialdehyde (MDA) content, and calcium deposition of the four treatments were determined. The mRNA and protein expression levels of NRF2, NQO1, and haem oxygenase 1 (HO1) and the osteogenic markers ALP, Runx1, OPN, bone morphogenetic protein 2 (BMP2), and ß-catenin were quantified by RT-PCR and western blotting. VSMC apoptosis was calculated by flow cytometry. The in vitro results suggested that intracellular oxidative stress and calcification were closely associated with NRF2 activity and that the activation of NRF2 could significantly suppress osteogenic transition and apoptosis in VSMCs. Thus, this study indicated that the NRF2-related antioxidant pathway can positively respond to and protect against the initiation and progression of VC in CKD by reducing oxidative stress. This study may contribute insights facilitating the application of the NRF2 antioxidative system as a therapeutic treatment for vascular diseases such as CKD.


Assuntos
Antioxidantes/metabolismo , Aorta/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais , Calcificação Vascular/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Modelos Animais de Doenças , Heme Oxigenase (Desciclizante)/biossíntese , Masculino , NAD(P)H Desidrogenase (Quinona)/biossíntese , Ratos , Ratos Wistar
2.
Biotechnol Appl Biochem ; 67(3): 317-322, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31746064

RESUMO

Epigallocatechin 3-gallate (EGCG) is a polyphenol that has been shown to have antioxidant and anti-inflammatory effects. In this study, collagen-induced arthritis (CIA) model, in Wistar albino rats, was used to elucidate the effect of EGCG on pathogenetic pathways in inflammatory arthritis. The levels of serum TNF-α, IL-17, malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx); the expression levels of tissue heme oxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2); histopathologically, perisynovial inflammation and cartilage-bone destruction were examined. In the sham group, serum TNF-α, IL-17, and MDA levels increased, while SOD, CAT, GPx levels, and the expressions of Nrf2 and HO-1 decreased. On the other hand, in the EGCG administered groups, serum TNF-α, IL-17, and MDA levels improved, while SOD, CAT, GPx levels and the expressions of Nrf2 and HO-1 increased. Moreover, histopathological analysis has shown that perisynovial inflammation and cartilage-bone destruction decreased in the EGCG administered groups. These results suggest that EGCG has an antiarthritic effect by regulating the oxidative-antioxidant balance and cytokine levels in the CIA model, which is a surrogate experimental model of rheumatoid arthritis.


Assuntos
Artrite Experimental/tratamento farmacológico , Catequina/análogos & derivados , Citocinas/antagonistas & inibidores , Modelos Animais de Doenças , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/metabolismo , Catequina/farmacologia , Colágeno Tipo II , Citocinas/biossíntese , Feminino , Heme Oxigenase (Desciclizante)/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Ratos , Ratos Wistar
3.
Proc Natl Acad Sci U S A ; 114(45): 11962-11967, 2017 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-29078307

RESUMO

Optogenetics is a powerful tool to precisely manipulate cell signaling in space and time. For example, protein activity can be regulated by several light-induced dimerization (LID) systems. Among them, the phytochrome B (PhyB)-phytochrome-interacting factor (PIF) system is the only available LID system controlled by red and far-red lights. However, the PhyB-PIF system requires phycocyanobilin (PCB) or phytochromobilin as a chromophore, which must be artificially added to mammalian cells. Here, we report an expression vector that coexpresses HO1 and PcyA with Ferredoxin and Ferredoxin-NADP+ reductase for the efficient synthesis of PCB in the mitochondria of mammalian cells. An even higher intracellular PCB concentration was achieved by the depletion of biliverdin reductase A, which degrades PCB. The PCB synthesis and PhyB-PIF systems allowed us to optogenetically regulate intracellular signaling without any external supply of chromophores. Thus, we have provided a practical method for developing a fully genetically encoded PhyB-PIF system, which paves the way for its application to a living animal.


Assuntos
Ferredoxina-NADP Redutase/biossíntese , Ferredoxinas/biossíntese , Heme Oxigenase (Desciclizante)/biossíntese , Optogenética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases/biossíntese , Ficobilinas/biossíntese , Ficocianina/biossíntese , Linhagem Celular Tumoral , Vetores Genéticos/genética , Células HeLa , Humanos , Luz , Ficobilinas/genética , Ficocianina/genética , Transdução de Sinais/genética
4.
J Cell Physiol ; 234(5): 7301-7319, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30362554

RESUMO

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common and potentially lethal clinical syndromes characterized by acute respiratory failure resulting from excessive pulmonary inflammation, noncardiogenic pulmonary edema, and alveolar-capillary barrier disruption. At present, there is no effective and specific therapy for ALI/ARDS. Mesenchymal stem cells (MSCs) have well-known therapeutic potential in patients with ALI/ARDS. Heme oxygenase-1 (HO-1), a cytoprotective enzyme, possesses antioxidative, anti-inflammatory, and antiapoptotic effects. Thus, a combination of MSC transplantation with HO-1 delivery may have an additional protective effect against ALI/ARDS. This study investigated the effect of HO-1-modified bone-marrow-derived MSCs (MSCs-HO-1) on lipopolysaccharide (LPS)-induced ALI and its underlying mechanisms. We established MSCs-HO-1 through lentiviral transduction. The ALI rat model was established by successive LPS inhalations following injection with MSCs-HO-1. The survival rate, histological changes in the lungs, total protein concentration and neutrophil counts in bronchoalveolar lavage fluid, lung wet/dry weight ratio, cytokine levels in serum and lungs, nuclear transcription factor-κB activity, and protein expression of Toll-like receptor 4 signaling adaptors were examined. Furthermore, the cell viability, apoptosis, and paracrine activity of MSCs-HO-1 were examined under inflammatory stimuli in vitro. MSCs-HO-1 injection improved these parameters compared with primary unmodified MSCs. Moreover, MSCs-HO-1 had superior prosurvival and antiapoptotic properties and enhanced paracrine functions in vitro. Therefore, MSCs-HO-1 exert an enhanced protective effect to alleviate LPS-induced ALI in rats, and the mechanisms may be partially associated with superior prosurvival, antiapoptosis, and enhanced paracrine functions of MSCs-HO-1. These findings provide a novel insight into MSC-based therapeutic strategies for treating ALI/ARDS.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Terapia Genética , Heme Oxigenase (Desciclizante)/biossíntese , Pulmão/enzimologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/enzimologia , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/genética , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Fator 7 de Crescimento de Fibroblastos/metabolismo , Heme Oxigenase (Desciclizante)/genética , Fator de Crescimento de Hepatócito/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-10/metabolismo , Lipopolissacarídeos , Pulmão/patologia , Comunicação Parácrina , Ratos Wistar , Transdução de Sinais
5.
Clin Sci (Lond) ; 133(1): 117-134, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30538149

RESUMO

We previously demonstrated in in vitro and ex vivo models that physiological concentrations of unconjugated bilirubin (BR) prevent oxidative stress (OS)-induced hepatocanalicular dysfunction and cholestasis. Here, we aimed to ascertain, in the whole rat, whether a similar cholestatic OS injury can be counteracted by heme oxygenase-1 (HO-1) induction that consequently elevates endogenous BR levels. This was achieved through the administration of hemin, an inducer of HO-1, the rate-limiting step in BR generation. We found that BR peaked between 6 and 8 h after hemin administration. During this time period, HO-1 induction fully prevented the pro-oxidant tert-butylhydroperoxide (tBuOOH)-induced drop in bile flow, and in the biliary excretion of bile salts and glutathione, the two main driving forces of bile flow; this was associated with preservation of the membrane localization of their respective canalicular transporters, bile salt export pump (Bsep) and multidrug resistance-associated protein 2 (Mrp2), which are otherwise endocytosed by OS. HO-1 induction counteracted the oxidation of intracellular proteins and membrane lipids induced by tBuOOH, and fully prevented the increase in the oxidized-to-total glutathione (GSHt) ratio, a sensitive parameter of hepatocellular OS. Compensatory elevations of the activity of the antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) were also prevented. We conclude that in vivo HO-1 induction protects the liver from acute oxidative injury, thus preventing consequent cholestasis. This reveals an important role for the induction of HO-1 and the consequently elevated levels of BR in preserving biliary secretory function under OS conditions, thus representing a novel therapeutic tool to limit the cholestatic injury that bears an oxidative background.


Assuntos
Antioxidantes/farmacologia , Colestase/prevenção & controle , Heme Oxigenase (Desciclizante)/biossíntese , Hemina/farmacologia , Fígado/efeitos dos fármacos , Estresse Oxidativo , Animais , Bile/metabolismo , Bilirrubina/metabolismo , Catalase/metabolismo , Colestase/induzido quimicamente , Colestase/enzimologia , Colestase/patologia , Modelos Animais de Doenças , Indução Enzimática , Glutationa/metabolismo , Fígado/enzimologia , Fígado/patologia , Masculino , Ratos Wistar , Superóxido Dismutase/metabolismo , terc-Butil Hidroperóxido
6.
Alcohol Clin Exp Res ; 43(8): 1651-1661, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31141180

RESUMO

BACKGROUND: This study aimed to investigate the protective effect of 5-aminolevulinic acid (5-ALA) and sodium ferrous citrate (SFC) against binge alcohol-induced gut leakiness and inflammatory liver disease in HIV transgenic (TG) rats. METHODS: TG rats were treated with 3 consecutive doses of binge ethanol (EtOH) with or without 5-ALA/SFC. Blood and liver tissue samples were collected at 6 hours following the last dose of EtOH. RESULTS: Compared with the wild-type (WT) rats, the TG rats showed increased sensitivity to alcohol-mediated inflammation, as evidenced by the significantly elevated levels of serum endotoxin, AST, ALT, ED1, and ED2 staining in liver. In contrast, 5-ALA/SFC improved the above biochemical and histochemical profiles. 5-ALA/SFC also attenuated the up-regulated mRNA expression of leptin and CCL2. Furthermore, down-regulated intestinal ZO-1 protein expression was also inhibited by 5-ALA/SFC. Moreover, the expressions of HO-1, HO-2, Sirt1, and related signal transduction molecules in liver were increased by 5-ALA/SFC. These results demonstrated that 5-ALA/SFC treatment ameliorated binge alcohol exposure liver injury in a rat model of HIV-infected patients by reducing macrophage activation and expression of inflammatory cytokines/chemokines, and by inducing HO-1, HO-2, and Sirt1 expression. CONCLUSIONS: Taken together, these findings suggested that treatment with 5-ALA/SFC has a potential therapeutic effect for binge alcohol exposure liver injury in HIV-infected patients.


Assuntos
Ácido Aminolevulínico/farmacologia , Consumo Excessivo de Bebidas Alcoólicas/fisiopatologia , Etanol/efeitos adversos , Hepatite/prevenção & controle , Intestinos/fisiopatologia , Permeabilidade/efeitos dos fármacos , Alanina Transaminase/sangue , Animais , Antígenos CD/biossíntese , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/biossíntese , Antígenos de Diferenciação Mielomonocítica/imunologia , Aspartato Aminotransferases/sangue , Moléculas de Adesão Celular Neuronais/genética , Ácido Cítrico , Endotoxinas/sangue , Infecções por Enterobacteriaceae/microbiologia , Compostos Ferrosos , Infecções por HIV/complicações , HIV-1/genética , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase-1/biossíntese , Hepatite/sangue , Hepatite/complicações , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/sangue , Fígado/metabolismo , Ratos , Ratos Transgênicos , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/imunologia , Sirtuína 1/biossíntese , Células-Tronco , Triglicerídeos/metabolismo , Proteína da Zônula de Oclusão-1/biossíntese , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
7.
BMC Nephrol ; 18(1): 232, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-28697727

RESUMO

BACKGROUND: Intermedin [IMD, adrenomedullin-2 (ADM-2)] attenuates renal fibrosis by inhibition of oxidative stress. However, the precise mechanisms remain unknown. Heme oxygenase-1 (HO-1), an antioxidant agent, is associated with antifibrogenic effects. ADM is known to induce HO-1. Whether IMD has any effect on HO-1 is unclear. Herein, we determined whether the antifibrotic properties of IMD are mediated by induction of HO-1. METHODS: Renal fibrosis was induced by unilateral ureteral obstruction (UUO) performed on male Wistar rats. Rat proximal tubular epithelial cell line (NRK-52E) was exposed to rhTGF-ß1 (10 ng/ml) to establish an in vitro model of epithelial-mesenchymal transition (EMT). IMD was over-expressed in vivo and in vitro using the vector pcDNA3.1-IMD. Zinc protoporphyrin (ZnPP) was used to block HO-1 enzymatic activity. IMD effects on HO-1 expression in the obstructed kidney of UUO rat and in TGF-ß1-stimulated NRK-52E were analyzed by real-time RT-PCR, Western blotting or immunohistochemistry. HO activity in the obstructed kidney, contralateral kidney of UUO rat and NRK-52E was examined by measuring bilirubin production. Renal fibrosis was determined by Masson trichrome staining and collagen I expression. Macrophage infiltration and IL-6 expression were evaluated using immunohistochemical analysis. In vivo and in vitro EMT was assessed by measuring α-smooth muscle actin (α-SMA) and E-cadherin expression using Western blotting or immunofluorescence, respectively. RESULTS: HO-1 expression and HO activity were increased in IMD-treated UUO kidneys or NRK-52E. The obstructed kidneys of UUO rats demonstrated significant interstitial fibrosis on day 7 after operation. In contrast, kidneys that were treated with IMD gene transfer exhibited minimal interstitial fibrosis. The obstructed kidneys of UUO rats also had greater macrophage infiltration and IL-6 expression. IMD restrained infiltration of macrophages and expression of IL-6 in UUO kidneys. The degree of EMT was extensive in obstructed kidneys of UUO rats as indicated by decreased expression of E-cadherin and increased expression of α-SMA. In vitro studies using NRK-52E confirmed these observations. EMT was suppressed by IMD gene delivery. However, all of the above beneficial effects of IMD were eliminated by ZnPP, an inhibitor of HO enzyme activity. CONCLUSION: This study demonstrates that IMD attenuates renal fibrosis by induction of HO-1.


Assuntos
Adrenomedulina/administração & dosagem , Heme Oxigenase (Desciclizante)/biossíntese , Nefropatias/enzimologia , Nefropatias/prevenção & controle , Neuropeptídeos/administração & dosagem , Obstrução Ureteral/enzimologia , Obstrução Ureteral/terapia , Adrenomedulina/genética , Animais , Células Cultivadas , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/fisiologia , Fibrose/enzimologia , Fibrose/genética , Fibrose/terapia , Técnicas de Transferência de Genes , Heme Oxigenase (Desciclizante)/genética , Nefropatias/genética , Masculino , Neuropeptídeos/genética , Distribuição Aleatória , Ratos , Ratos Wistar , Obstrução Ureteral/genética
8.
Neurochem Res ; 41(6): 1291-304, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26801169

RESUMO

This study aimed to identify the effect of ß-caryophyllene (BCP) pretreatment and elucidate the Nrf2/HO-1 signaling mechanism after focal cerebral ischemia-reperfusion (I-R) injury in rats. Adult male Sprague-Dawley rats were randomly assigned to the sham-operated group, I-R group and BCP pretreated I-R group. At 24 h after reperfusion, neurological deficits and infarct volume were evaluated. Pathological changes of neuron in hippocampuses were observed by Nissil staining and transmission electron microscopy (TEM). Oxidative stress was assessed by malondialdehyde (MDA) level, lipid peroxidation (LPO), nitric oxide (NO), superoxide dismutase (SOD) and Catalase (CAT) activity. The expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) were analysed by Western blotting and real-time quantitative polymerase chain reaction (Q-PCR). The protein expression of Bcl-2 and Bax was determined by immunohistochemistry. Apoptotic cells were detected using TUNEL staining. In I-R group, neurological deficit scores, cerebral infarct volume, MDA levels, LPO content, NO level, expression of Bax and TUNEL-positive cells were found to be increased at 24 h after I-R injury, while SOD activity, CAT activity and expression of Bcl-2 were decreased. However, results in the BCP pretreatment groups were reversed. And the protein and mRNA expressions of Nrf2 and HO-1 were significantly up-regulated in the BCP pretreated I-R group. Results of Nissil staining and TEM scan manifested that BCP remarkablely improved neuronal injury after I-R in rats. All the above suggested that BCP pretreatment played a neuroprotective role in cerebral I-R injury, which might be exerted by upregulating the expression of Nrf2 and HO-1 to ameliorate oxidative damage and neuronal apoptosis.


Assuntos
Isquemia Encefálica/prevenção & controle , Heme Oxigenase (Desciclizante)/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Traumatismo por Reperfusão/prevenção & controle , Sesquiterpenos/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Isquemia Encefálica/metabolismo , Relação Dose-Resposta a Droga , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Sesquiterpenos Policíclicos , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Sesquiterpenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
9.
Pediatr Res ; 79(2): 243-50, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26492285

RESUMO

BACKGROUND: Statins elicit antioxidant effects independently of their lipid-lowering properties. Heme oxygenase-1 (HO-1) induction may be a part of these pleiotropic effects, which are insufficiently described in the kidney. We hypothesize that simvastatin (SIM) transcriptionally activates HO-1 that protects renal proximal tubule cells from lipotoxic injury. METHODS: Impact of SIM on 100 µmol/l oleic acid (OA)-mediated reactive oxygen species (ROS) production and consequent oxidative stress (4-hydroxynonenal (HNE) content) as well as cell injury/apoptosis (lactate dehydrogenase (LDH) release, caspase-3 activation) were determined in cultured renal proximal tubule (NRK52E) cells. Effect of SIM on the HO-1 promoter and its enhancer elements (antioxidant response element (ARE), CCAAT, AP1, and cAMP response element (CRE)) was also determined in reporter luciferase assays. Dominant-negative (dnMEK, M1CREB) and pharmacologic (H89) approaches were used to inhibit activation of extracellular signal regulated kinase (ERK), CREB, and protein kinase A (PKA), respectively. RESULTS: SIM dose-dependently activated the HO-1 promoter that was essential for protection against OA-dependent ROS production/oxidative stress and LDH release/caspase-3 activation. We found that the HO-1 promoter was induced through ERK and PKA-dependent activation of the CRE by SIM. CONCLUSION: SIM may protect the kidney from adverse effects of circulating fatty acids by upregulating the antioxidant HO-1, aside from its well-described lipid-lowering effects.


Assuntos
Antioxidantes/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Heme Oxigenase (Desciclizante)/biossíntese , Túbulos Renais Proximais/efeitos dos fármacos , Ácido Oleico/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Sinvastatina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoproteção , Relação Dose-Resposta a Droga , Indução Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Heme Oxigenase (Desciclizante)/genética , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/patologia , Regiões Promotoras Genéticas , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transfecção
10.
Ren Fail ; 38(9): 1516-1527, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27499091

RESUMO

OBJECTIVES: 18ß-glycyrrhetinic acid (18ß-GA) has multiple beneficial and therapeutic effects. However, its protective roles on methotrexate (MTX)-induced renal injury are not well defined. In the present study, we investigated the possible protective effects of 18ß-GA against MTX-induced nephrotoxicity in rats. MATERIALS: 18ß-GA (50 and 100 mg/kg) was administered for 7 days either before or after MTX. The rats were decapitated and kidney and serum samples were collected. RESULTS: MTX-induced renal injury in rats was evidenced by the significant (p < 0.001) increase in circulating kidney function markers and tumor necrosis factor alpha (TNF-α), as well as the histopathological alterations. MTX-induced rats exhibited significantly increased lipid peroxidation (p < 0.05) and nitric oxide (p < 0.001) levels, with concomitant marked (p < 0.001) decline in the antioxidant defenses. 18ß-GA, administered either before or after MTX, produced a significant amelioration of circulating kidney function markers, TNF-α, kidney lipid peroxidation, nitric oxide, and antioxidant defenses. In addition, 18ß-GA supplementation significantly up-regulated the mRNA abundance of both nuclear factor-erythroid 2-related factor 2 (Nrf2) and hemoxygenase 1 (HO-1) in the kidney of MTX-induced rats. CONCLUSIONS: These results indicate that 18ß-GA has a protective effect on MTX-induced nephrotoxicity with possible mechanisms of attenuating oxidative stress and inflammation through up-regulating the Nrf2/ARE signaling. These findings make 18ß-GA candidate as a potent agent in preventing MTX-induced kidney injury.


Assuntos
Injúria Renal Aguda/prevenção & controle , Hidrolases de Éster Carboxílico/genética , Ácido Glicirretínico/análogos & derivados , Heme Oxigenase (Desciclizante)/genética , Fator 2 Relacionado a NF-E2/genética , RNA/genética , Regulação para Cima , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Animais , Biomarcadores/metabolismo , Hidrolases de Éster Carboxílico/biossíntese , Modelos Animais de Doenças , Junções Comunicantes , Ácido Glicirretínico/farmacologia , Heme Oxigenase (Desciclizante)/biossíntese , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Peroxidação de Lipídeos , Masculino , Metotrexato/toxicidade , Fator 2 Relacionado a NF-E2/biossíntese , Estresse Oxidativo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Pharm Biol ; 54(3): 555-60, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26270131

RESUMO

CONTEXT: Ischemia/hypoxia and reperfusion impair mitochondria and produce a large amount of reactive oxygen species (ROS), which lead to mitochondrial and brain damage. Furthermore, heme oxygenase-1 (HO-1) as a cytoprotective gene protects cells against ROS-induced cell death in ischemia-reperfusion injury. Induction of HO-1 is involved in cytoprotective effects of taxol. OBJECTIVE: We hypothesize that taxol protects cardiac myocytes possibly by preserving myocardial mitochondrial function and inducing HO-1 expression through the JNK pathway. MATERIALS AND METHODS: In this project, the perfused Langendorff hearts isolated from rats were randomly divided into five groups: control, ischemic, ischemic + taxol (0.1 µM), ischemic + taxol (0.3 µM), and ischemic + taxol (1 µM). Briefly, following a 15 min equilibration period, the control group was subject to normoxic perfusion for 120 min; the ischemia group, normoxic reperfusion for 120 min after 30 min ischemia; the taxol groups, normoxic reperfusion for 120 min after 30-min ischemia with taxol (0.1, 0.3, or 1 µM). The microtubule disruption score, ROS levels, and the activity of mitochondrial electron transport chain complexes I and III were examined by using immunohistochemical methods and free radical detection kits. Western blot assay was employed to study the underlying mechanisms. RESULTS: After Taxol treatment (0.1 µM), the ischemic microtubule disruption score was reduced to 9.8 ± 1.9%. The study revealed that 0.1, 0.3, and 1 µM taxol reduced the level of ROS by 33, 46 and 51%, respectively (p < 0.05). In additional, 0.3 and 1 µM taxol dramatically increased the activity of mitochondrial electron transport chain complex I (99.11 ± 2.59, 103.49 ± 3.89) and mitochondrial electron transport chain complex III (877.82 ± 12.08; 907.42 ± 16.21; 914.73 ± 19.39, *p < 0.05). Additionally, phosphorylation levels of JNK1 were significantly increased in the taxol group. Furthermore, the expression level of HO-1 increased with taxol treatments, which could be inhibited by the specific inhibitor of JNK, SP600125. DISCUSSION AND CONCLUSION: Taxol stabilized microtubules and effectively reduced ROS levels during ischemia. It also preserved the activity of mitochondrial complexes I and III. Interestingly, taxol induced the expression of HO-1 via the JNK pathway in cardiac myocytes.


Assuntos
Heme Oxigenase (Desciclizante)/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/enzimologia , Paclitaxel/uso terapêutico , Animais , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/fisiologia , Regulação Enzimológica da Expressão Gênica , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Paclitaxel/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
12.
J Cell Mol Med ; 19(5): 924-33, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25683492

RESUMO

Estrogen-induced cholestasis is characterized by impaired hepatic uptake and biliary bile acids secretion because of changes in hepatocyte transporter expression. The induction of heme oxygenase-1 (HMOX1), the inducible isozyme in heme catabolism, is mediated via the Bach1/Nrf2 pathway, and protects livers from toxic, oxidative and inflammatory insults. However, its role in cholestasis remains unknown. Here, we investigated the effects of HMOX1 induction by heme on ethinylestradiol-induced cholestasis and possible underlying mechanisms. Wistar rats were given ethinylestradiol (5 mg/kg s.c.) for 5 days. HMOX1 was induced by heme (15 µmol/kg i.p.) 24 hrs prior to ethinylestradiol. Serum cholestatic markers, hepatocyte and renal membrane transporter expression, and biliary and urinary bile acids excretion were quantified. Ethinylestradiol significantly increased cholestatic markers (P ≤ 0.01), decreased biliary bile acid excretion (39%, P = 0.01), down-regulated hepatocyte transporters (Ntcp/Oatp1b2/Oatp1a4/Mrp2, P ≤ 0.05), and up-regulated Mrp3 (348%, P ≤ 0.05). Heme pre-treatment normalized cholestatic markers, increased biliary bile acid excretion (167%, P ≤ 0.05) and up-regulated hepatocyte transporter expression. Moreover, heme induced Mrp3 expression in control (319%, P ≤ 0.05) and ethinylestradiol-treated rats (512%, P ≤ 0.05). In primary rat hepatocytes, Nrf2 silencing completely abolished heme-induced Mrp3 expression. Additionally, heme significantly increased urinary bile acid clearance via up-regulation (Mrp2/Mrp4) or down-regulation (Mrp3) of renal transporters (P ≤ 0.05). We conclude that HMOX1 induction by heme increases hepatocyte transporter expression, subsequently stimulating bile flow in cholestasis. Also, heme stimulates hepatic Mrp3 expression via a Nrf2-dependent mechanism. Bile acids transported by Mrp3 to the plasma are highly cleared into the urine, resulting in normal plasma bile acid levels. Thus, HMOX1 induction may be a potential therapeutic strategy for the treatment of ethinylestradiol-induced cholestasis.


Assuntos
Colestase/enzimologia , Heme Oxigenase (Desciclizante)/biossíntese , Heme/farmacologia , Substâncias Protetoras/farmacologia , Transportadores de Cassetes de Ligação de ATP/genética , Fosfatase Alcalina/sangue , Animais , Ácidos e Sais Biliares/sangue , Bilirrubina/sangue , Bilirrubina/farmacologia , Células Cultivadas , Colestase/sangue , Colestase/induzido quimicamente , Indução Enzimática/efeitos dos fármacos , Etinilestradiol , Feminino , Expressão Gênica/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/genética , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Cultura Primária de Células , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácido Taurocólico/farmacologia
13.
Biochem Biophys Res Commun ; 456(2): 549-54, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25264201

RESUMO

Hyperoxic acute lung injury (HALI) is a clinical syndrome as a result of prolonged supplement of high concentrations of oxygen. Previous studies have shown hyperbaric oxygen preconditioning (HBO-PC) had a protective effect on oxidative injury. In the present study, we investigated the effect of HBO-PC on HALI in rats. The results demonstrated that HBO-PC ameliorated the lung biochemical and histological alterations induced by hyperoxia, decreased oxidative products but increased antioxidant enzymes. Furthermore, HBO-PC up-regulated heme oxygenase-1 (HO-1) mRNA and activity in lung tissues. The administration of HO-1 inhibitor, Zinc protoporphyrin IX, abolished its protective effects. The data showed that HBO-PC could protect rats against HALI and the anti-oxidative effect may be related to the up-regulation of HO-1.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Heme Oxigenase (Desciclizante)/biossíntese , Oxigenoterapia Hiperbárica/métodos , Hiperóxia/prevenção & controle , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Lesão Pulmonar Aguda/patologia , Animais , Inibidores Enzimáticos/administração & dosagem , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Hiperóxia/patologia , L-Lactato Desidrogenase/metabolismo , Masculino , Protoporfirinas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
14.
J Recept Signal Transduct Res ; 35(5): 479-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26096165

RESUMO

Sulfur mustard (SM) is a potent alkylating agent which reacts with nucleophilic groups on DNA, RNA and proteins. It is capable of inducing cellular toxicity and oxidative stress via production of reactive oxygen species (ROS) and reactive nitrogen species (RNS). The accumulation of high amounts of the reactive species causes harmful effects such as DNA damage, lipid peroxidation, protein oxidation, inflammation and apoptosis. Although SM (also known as mustard gas) and its derivatives are rapidly removed from the body, long-term damages are much more serious than the short-term effects and may be correlated with the subsequent changes occurred on the genome. In order to defend against oxidative properties of this toxic molecule, cells trigger several anti-oxidant pathways through up-regulating the corresponding genes. Enzymes like heme oxygenase-1, superoxide dismutase and glutathione-S-transferase are the examples of such genes. These enzymes produce anti-oxidant substances that are able to scavenge the reactive species, alleviate their noxious effects and protect the cells. Following SM gas exposure, gene transcription (mRNA levels) of these enzymes are ramped up to help detoxify the cells. Yet, some studies have reported that the up-regulated transcription does not necessarily translate into higher protein expression levels. The exact reason why this phenomenon happens is not clear. Creation of mutations in the genome sequence may lead to protein structure changes. Phosphorylation or other post-translational alterations of proteins upon SM exposure are also considered as possible causes. In addition, alterations in some microRNAs responsible for regulating post-translation events may inhibit the expression of the anti-oxidant proteins in the poisoned cells at translational level.


Assuntos
Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase (Desciclizante)/genética , Gás de Mostarda/intoxicação , Biossíntese de Proteínas/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Humanos , Mutação/efeitos dos fármacos , Mutação/genética
15.
Toxicol Appl Pharmacol ; 289(2): 142-54, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26440581

RESUMO

The natural product totarol, a phenolic diterpenoid and a major constituent isolated from the sap of Podocarpus totara, has been reported to have a potent antimicrobial activity. In this study, we determined whether totarol possessed an additional neuroprotective activity in vitro and in vivo. We found that totarol prevented glutamate- and oxygen and glucose deprivation-induced neuronal death in primary rat cerebellar granule neuronal cells and cerebral cortical neurons. Totarol increased Akt and GSK-3ß phosphorylation, Nrf2 and heme oxygenase-1 (HO-1) protein expressions and suppressed oxidative stress by increasing GSH and SOD activities. The PI3K/Akt inhibitor LY294002 prevented totarol neuroprotective effect by suppressing the totarol-induced changes in HO-1 expression and the activities of GSH and SOD. The HO-1 inhibitor ZnPPIX also prevented totarol-increased GSH and SOD activities. In a model of acute cerebral ischemic injury in Sprague-Dawley rats, produced by occlusion of the middle cerebral artery for 2h followed by 22 h or 46 h of reperfusion, totarol significantly reduced infarct volume and improved the neurological deficit. In this model, totarol increased HO-1 expression and the activities of GSH and SOD. These observations suggest that totarol may be a novel activator of the Akt/HO-1 pathway protecting against ischemic stroke through reduction of oxidative stress.


Assuntos
Antioxidantes/farmacologia , Encéfalo/efeitos dos fármacos , Diterpenos/farmacologia , Heme Oxigenase (Desciclizante)/biossíntese , Infarto da Artéria Cerebral Média/prevenção & controle , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Abietanos , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Morte Celular/efeitos dos fármacos , Hipóxia Celular , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática , Indução Enzimática , Agonistas de Aminoácidos Excitatórios/toxicidade , Glucose/deficiência , Ácido Glutâmico/toxicidade , Glutationa/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/enzimologia , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Fatores de Tempo
16.
J Cardiovasc Pharmacol ; 65(6): 601-6, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25714596

RESUMO

Although mesenchymal stem cells (MSCs) have a therapeutic potential for the repair of tissue injuries, their poor viability in damaged tissue limits their effectiveness. Statins can induce an increased production of heme oxygenase-1 (HO-1), which may prevent this detrimental effect in MSCs. We investigated the protective effect of statin-induced overexpression of HO-1 by examining changes in gene expression and function in MSCs after pitavastatin treatment. The relative expression of the HO-1 and endothelial nitric oxide synthase genes in MSCs was significantly increased after treatment with pitavastatin (MSCs). Immunocytological analysis showed that MSCs also stained with phospho-Akt. After exposure to oxidative stress, MSCs showed increased resistance to induced cell death compared with control MSCs. Under serum starvation conditions, MSCs treated with 1 µM pitavastatin showed enhanced cell proliferation and a marked increase in vascular endothelial growth factor production compared with control MSCs. Interestingly, MSCs showed enhanced tube formation under both normoxia and hypoxia. These results demonstrate that pitavastatin can enhance endogenous HO-1 expression in MSCs, which may protect the cells into the environment of oxidative stress with partial activation of endothelial nitric oxide synthase and Akt phosphorylation.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/biossíntese , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Quinolinas/farmacologia , Indutores da Angiogênese/farmacologia , Animais , Células da Medula Óssea/enzimologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Indução Enzimática , Heme Oxigenase (Desciclizante)/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Células-Tronco Mesenquimais/enzimologia , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/biossíntese , Ratos Endogâmicos Lew , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Nanomedicine ; 11(2): 467-77, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25168934

RESUMO

This study investigated toxicity of nanocarriers comprised of cationic polymer and lipid components often used in gene and drug delivery, formulated as cationic micelles and liposomes. Rats were injected intravenously with 10, 25 or 100 mg/kg and sacrificed after 24 or 48 h, or 24 h after the last of three intravenous injections of 100 mg/kg every other day. Histological evaluation of liver, lung and spleen, clinical chemistry parameters, and hematology indicated little effect of treatment. DNA strand breaks were increased in the lung and spleen. Further, in the dose response study we found unaltered expression levels of genes in the antioxidant response (HMOX1) and repair of oxidized nucleobases (OGG1), whereas expression levels of cytokines (IL6, CXCL2 and CCL2) were elevated in lung, spleen or liver. The results indicate that assessment of genotoxicity and gene expression add information on toxicity of nanocarriers, which is not obtained by histology and hematology. FROM THE CLINICAL EDITOR: This study investigates the toxicity of cationic micelles and liposomes utilized as nanocarriers in gene and drug delivery, demonstrating its effects on the lungs, spleen and liver.


Assuntos
Cátions/efeitos adversos , Dano ao DNA/efeitos dos fármacos , Lipossomos/efeitos adversos , Micelas , Animais , Cátions/uso terapêutico , Quimiocina CCL2/biossíntese , Quimiocina CXCL2/biossíntese , DNA Glicosilases/biossíntese , Sistemas de Liberação de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Heme Oxigenase (Desciclizante)/biossíntese , Lipossomos/uso terapêutico , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Masculino , Ratos , Baço/efeitos dos fármacos
18.
Am J Physiol Regul Integr Comp Physiol ; 306(9): R641-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24553299

RESUMO

Preeclampsia is thought to arise from inadequate cytotrophoblast migration and invasion of the maternal spiral arteries, resulting in placental ischemia and hypertension. Evidence suggests that altered expression of epithelial Na(+) channel (ENaC) proteins may be a contributing mechanism for impaired cytotrophoblast migration. ENaC activity is required for normal cytotrophoblast migration. Moreover, ß-ENaC, the most robustly expressed placental ENaC message, is reduced in placentas from preeclamptic women. We recently demonstrated that heme oxygenase-1 (HO-1) protects against hypertension in a rat model of placental ischemia; however, whether HO-1 regulation of ß-ENaC contributes to the beneficial effects of HO-1 is unknown. The purpose of this study was to determine whether ß-ENaC mediates cytotrophoblast migration and whether HO-1 enhances ENaC-mediated migration. We showed that placental ischemia, induced by reducing uterine perfusion suppressed, and HO-1 induction restored, ß-ENaC expression in ischemic placentas. Using an in vitro model, we found that HO-1 induction, using cobalt protoporphyrin, stimulates cytotrophoblast ß-ENaC expression by 1.5- and 1.8-fold (10 and 50 µM). We then showed that silencing of ß-ENaC in cultured cytotrophoblasts (BeWo cells), by expression of dominant-negative constructs, reduced migration to 56 ± 13% (P < 0.05) of control. Importantly, HO-1 induction enhanced migration (43 ± 5% of control, P < 0.05), but the enhanced migratory response was entirely blocked by ENaC inhibition with amiloride (10 µM). Taken together, our results suggest that ß-ENaC mediates cytotrophoblast migration and increasing ß-ENaC expression by HO-1 induction enhances migration. HO-1 regulation of cytotrophoblast ß-ENaC expression and migration may be a potential therapeutic target in preeclamptic patients.


Assuntos
Movimento Celular , Canais Epiteliais de Sódio/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/metabolismo , Isquemia/enzimologia , Placenta/irrigação sanguínea , Placenta/enzimologia , Trofoblastos/enzimologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Indução Enzimática , Canais Epiteliais de Sódio/genética , Feminino , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase-1/biossíntese , Humanos , Isquemia/fisiopatologia , Circulação Placentária , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/fisiopatologia , Gravidez , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transfecção
19.
J Pharmacol Exp Ther ; 347(2): 346-56, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23995597

RESUMO

An exaggerated release of proinflammatory cytokines and accompanying inflammation contributes to the development of multiple organ failure after hemorrhagic shock. Here, we tested the nuclear factor (NF) κ-light-chain-enhancer of activated B cell (NF-κB)-mediated transcriptional control of inflammatory pathways as a target in the management of hemorrhage-induced inflammation. We performed a study in a rat model of fixed-volume hemorrhage to investigate the anti-inflammatory effects of the diphenyldifluoroketone EF24 [3,5-bis(2-fluorobenzylidene)piperidin-4-one], an NF-κB inhibitor, in lung tissue. EF24 treatment (0.4 mg/kg) significantly prevented the upregulation of inflammatory biomarkers in rats subjected to 50% hemorrhage and preserved the pulmonary histology in hemorrhaged rats. The lung tissue from treated rats showed marked suppression of the hemorrhage-mediated induction of Toll-like receptor 4, phospho-p65 NF-κB, inducible nitric-oxide synthase, heme oxygenase-1, and cyclooxygenase-2 (COX-2). The hemorrhage-induced COX-2 activity was also significantly inhibited by the EF24 treatment. At the same time, EF24 induced nuclear factor (erythroid-derived 2)-like 2-mediated protective mechanisms against oxidative stress. EF24 also reduced hemorrhage-induced lung myeloperoxidase activity. The plasma levels of proinflammatory tumor necrosis factor-α, interleukin (IL)-6, IL-1α, and IL-1ß were lower in EF24-treated rats than in untreated rats. Moreover, there was a significant reduction in the pulmonary expression of high-mobility group B1 protein. These biochemical effects were accompanied by a significant improvement in the survival of rats administered with EF24 as compared with the rats receiving vehicle control (P < 0.05). Overall, the results suggest that EF24 attenuates hemorrhage-induced inflammation and could serve as a salutary anti-inflammatory agent in resuscitation strategies.


Assuntos
Anti-Inflamatórios/uso terapêutico , Compostos de Benzilideno/uso terapêutico , Hipovolemia/prevenção & controle , Piperidonas/uso terapêutico , Choque Hemorrágico/tratamento farmacológico , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Compostos de Benzilideno/administração & dosagem , Compostos de Benzilideno/farmacologia , Ciclo-Oxigenase 2/biossíntese , Citocinas/sangue , Citocinas/imunologia , Modelos Animais de Doenças , Heme Oxigenase (Desciclizante)/biossíntese , Hipovolemia/etiologia , Hipovolemia/imunologia , Hipovolemia/patologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Masculino , Óxido Nítrico Sintase Tipo II/biossíntese , Piperidonas/administração & dosagem , Piperidonas/farmacologia , Ratos , Ratos Sprague-Dawley , Choque Hemorrágico/complicações , Choque Hemorrágico/imunologia , Choque Hemorrágico/patologia , Análise de Sobrevida , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/biossíntese
20.
J Vasc Surg ; 58(3): 756-65.e2, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23562340

RESUMO

OBJECTIVE: To explore the therapeutic potential of endothelial progenitor cells (EPCs) transfected with vascular endothelial growth factor A (VEGFA) and heme oxygenase-1 (HO-1) on rat hindlimb ischemia model. METHODS: Eukaryotic expression vectors encoding VEGFA or HO-1 were constructed and introduced into EPCs isolated from rat bone marrow. In total, 150 Sprague Dawley rat hindlimb ischemia models were established and randomized into five groups which were injected via tail vein with phosphate-buffered saline (PBS), nontransfected EPCs, VEGFA-modified EPCs, HO-1-modified EPCs, and both VEGFA- and HO-1-modified EPCs, respectively. The microvessel density, the expressions of VEGFA and HO-1 in the ischemic limbs, the recovery of blood flow as evaluated by laser-Doppler perfusion imaging, and the rate of limb salvage were compared among different groups. RESULTS: Transplantation of both VEGFA- and HO-1-modified EPCs in recipient rats significantly increased the microvessel density (expressed as capillaries/m(2) at day 21 after operation, group vascular endothelial growth factor (VEGF)+HO-1, 357 ± 14.1; group VEGF, 253.7 ± 9.9; group HO-1, 255.5 ± 12.5; group EPC, 210.7 ± 10.3; group PBS, 144.3 ± 9.3; P < .001), the expressions of VEGFA and HO-1 in ischemic tissue, the recovery of blood flow (at day 21, VEGF+HO-1 group, 85.4 ± 17.8%; VEGF group, 51.2 ± 13.2%; HO-1 group, 50.4 ± 12.9%; EPC group, 39.9 ± 8.5%; PBS group, 28.3 ± 7.8%; P < .001), and the rate of limb salvage (VEGF+HO-1 group, 94.4%; VEGF group or HO-1 group, 63.6%; EPC group, 50.0%; PBS group, 11.1%), compared with transplantation of either VEGFA- or HO-1-modified EPCs alone, or of nontransfected EPCs, or PBS injection. The order of therapeutic effectiveness on ischemic limbs was VEGFA- + HO-1-modifed EPC > either VEGFA- or HO-1-modified EPC alone > nontransfected EPC > PBS. CONCLUSIONS: VEGFA-modified EPC and HO-1-modified EPC synergized with each other in promoting angiogenesis in ischemic limbs of rat hindlimb ischemia model. In addition to VEGF, the introduction of HO-1 in EPC-based transplantation may serve as a novel and useful therapeutic strategy for ischemic disease of lower extremity.


Assuntos
Células Endoteliais/transplante , Terapia Genética/métodos , Heme Oxigenase (Desciclizante)/biossíntese , Isquemia/terapia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Transplante de Células-Tronco , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Biomarcadores/metabolismo , Velocidade do Fluxo Sanguíneo , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/enzimologia , Regulação da Expressão Gênica , Heme Oxigenase (Desciclizante)/genética , Membro Posterior , Isquemia/enzimologia , Isquemia/genética , Isquemia/patologia , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Fatores de Tempo , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA