Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 370
Filtrar
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.
Oxid Med Cell Longev ; 2021: 5521196, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34194603

RESUMO

Carbon monoxide releasing molecule-3 (CORM-3) has been shown to protect inflammatory diseases via the upregulation of heme oxygenases-1 (HO-1). However, in rat brain astrocytes (RBA-1), the mechanisms underlying CORM-3-induced HO-1 remain poorly defined. This study used western blot, real-time PCR, and promoter activity assays to determine the levels of HO-1 expression and 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) and dihydroethidium (DHE) to measure reactive oxygen species (ROS). We found that CORM-3-induced HO-1 expression was mediated through ROS generation by Nox or mitochondria. The signaling components were differentiated by pharmacological inhibitors and small interfering RNA (siRNA). Subcellular fractions, immunofluorescent staining, and chromatin immunoprecipitation assay were used to evaluate the nuclear translocation and promoter binding activity of Nrf2 induced by CORM-3. The roles of mTOR and FoxO1 in CORM-3-stimulated responses are still unknown in RBA-1 cells. Our results demonstrated that transfection with siRNAs or pretreatment with pharmacological inhibitors attenuated the levels of HO-1 and phosphorylation of signaling components including Akt, mTOR, FoxO1, and Nrf2 stimulated by CORM-3. Moreover, pretreatment with N-acetyl-L-cysteine, diphenyleneiodonium chloride, apocynin, or rotenone blocked nuclear translocation and promoter binding activity of Nrf2 induced by CORM-3. The present study concluded that in RBA-1 cells, CORM-3-induced HO-1 expression is, at least partially, mediated through Nox and mitochondria/ROS-dependent PI3K/Akt/mTOR cascade to activate FoxO1 or ROS leading to activation of Nrf2 activity.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Compostos Organometálicos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Indução Enzimática/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/biossíntese , Humanos , Ratos , Transfecção
3.
Drug Deliv ; 27(1): 1329-1341, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32945205

RESUMO

Rheumatoid arthritis (RA), autoimmune disease that is categorized via chronic inflammation manifestation, obesity, cardiovascular risk and even enhanced the mortality and affect the 0.3 and 1% of population worldwide. The current experimental study was scrutinize the anti-arthritic effect of ß-sitosterol loaded solid lipid nanoparticles (SLN) against complete Fruend adjuvant (CFA)-induced arthritis via dual pathway. Double emulsion solvent displacement method was used for the preparation of ß-sitosterol solid lipid nanoparticles (SLN). CFA was used to induce arthritis and rats were divided into different groups for 28 days. Biochemical, anti-inflammatory, pro-inflammatory cytokines and inflammatory mediator were estimated, respectively. Receptor activator of nuclear factor kappa-B ligand (RANKL), signal transducer and activator of transcription-3 (STAT3) nuclear factor erythroid 2-related factor 2 (Nrf2), Heme Oxygenase-1(HO-1) and Nuclear factor-κB (NF-κB) expression were estimated. ß-sitosterol-SLN significantly (p < .001) reduced the paw edema, arthritic index and increased the body weight. ß-sitosterol-SLN increased the redox status of synovium {reduce the malonaldehyde (MDA) and increase superoxide dismutase (SOD), glutathione (GSH) and catalase (CAT)} level and reduced the cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), interleukin-2, interleukin-6, interleukin-16, interleukin-17 and increased level of interleukin-10, Transforming growth factor beta (TGF-ß). ß-sitosterol-SLN significantly (p < .001) reduced the level of cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), vascular Endothelial Growth Factor (VEGF) and NF-κB. ß-sitosterol-SLN significantly increased the expression of HO-1,Nrf2 and decreased the expression of NF-κB, RANKL, STAT3. In conclusion, ß-sitosterol SLN showed the antiarthritic effect via suppression of NF-kB and activation of HO-1/Nrf-2 pathway.


Assuntos
Artrite Reumatoide/metabolismo , Heme Oxigenase (Desciclizante)/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , NF-kappa B/biossíntese , Nanopartículas/administração & dosagem , Sitosteroides/administração & dosagem , Animais , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/tratamento farmacológico , Relação Dose-Resposta a Droga , Portadores de Fármacos/administração & dosagem , Adjuvante de Freund/toxicidade , Hipolipemiantes/administração & dosagem , Masculino , Fator 2 Relacionado a NF-E2/agonistas , NF-kappa B/antagonistas & inibidores , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
4.
Biomolecules ; 10(3)2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32121307

RESUMO

Selective estrogen receptor modulators (SERMs) were discovered in the mid-1900s in connection with estrogen-related pathological conditions. They were developed to antagonize the adverse effects of estrogen and have been shown to be effective against postmenopausal disorders manifested by estrogen deficiency. Raloxifene (RAL), one of the most widely used SERMs, expresses estrogen-like effects on bones, while it is found to be an antagonist on breast and uterus. RAL has multiple beneficial effects throughout the body, including antioxidant and anti-inflammatory properties, because of which it gains particular attention. Additionally, previous studies have revealed that RAL is an efficient modulator of heme-oxygenase (HO) expression. HO, through its general activity, participates in comprehensive cell defense processes, thus the induction of HO by RAL administration indicates a major role in its therapeutic efficacy. In this review, we compile the current knowledge about the overall metabolic, neurocognitive, and cardiovascular effects of RAL involving the cytoprotective HO-system.


Assuntos
Antioxidantes/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/biossíntese , Cloridrato de Raloxifeno/uso terapêutico , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Feminino , Humanos
5.
Neuromolecular Med ; 22(2): 278-292, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31900786

RESUMO

Spinal cord injury (SCI) is a deliberating disorder with impairments in locomotor deficits and incapacitating sensory abnormalities. Harpagophytum procumbens (Hp) is a botanical widely used for treating inflammation and pain related to various inflammatory and musculoskeletal conditions. Using a modified rodent contusion model of SCI, we explored the effects of this botanical on locomotor function and responses to mechanical stimuli, and examined possible neurochemical changes associated with SCI-induced allodynia. Following spinal cord contusion at T10 level, Hp (300 mg/kg, p.o.) or vehicle (water) was administered daily starting 24 h post-surgery, and behavioral measurements made every-other day until sacrifice (Day 21). Hp treatment markedly ameliorated the contusion-induced decrease in locomotor function and increased sensitivity to mechanical stimuli. Determination of Iba1 expression in spinal cord tissues indicated microglial infiltration starting 3 days post-injury. SCI results in increased levels of 4-hydroxynonenal, an oxidative stress product and proalgesic, which was diminished at 7 days by treatment with Hp. SCI also enhanced antioxidant heme oxygenase-1 (HO-1) expression. Concurrent studies of cultured murine BV-2 microglial cells revealed that Hp suppressed oxidative/nitrosative stress and inflammatory responses, including production of nitric oxide and reactive oxygen species, phosphorylation of cytosolic phospholipases A2, and upregulation of the antioxidative stress pathway involving the nuclear factor erythroid 2-related factor 2 and HO-1. These results support the use of Hp for management of allodynia by providing resilience against the neuroinflammation and pain associated with SCI and other neuropathological conditions.


Assuntos
Harpagophytum/química , Hiperalgesia/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Fitoterapia , Extratos Vegetais/uso terapêutico , Traumatismos da Medula Espinal/complicações , Aldeídos/metabolismo , Animais , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase (Desciclizante)/genética , Hiperalgesia/etiologia , Inflamação , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/biossíntese , Fator 2 Relacionado a NF-E2/genética , Ácido Nítrico/metabolismo , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Método Simples-Cego , Tato
6.
Exp Neurol ; 324: 113135, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31778663

RESUMO

Traumatic brain injury (TBI) is one of the most common causes of death and disability worldwide. We investigated whether inhibition of p53 using pifithrin (PFT)-α or PFT-µ provides neuroprotective effects via p53 transcriptional dependent or -independent mechanisms, respectively. Sprague Dawley rats were subjected to controlled cortical impact TBI followed by the administration of PFTα or PFT-µ (2 mg/kg, i.v.) at 5 h after TBI. Brain contusion volume, as well as sensory and motor functions were evaluated at 24 h after TBI. TBI-induced impairments were mitigated by both PFT-α and PFT-µ. Fluoro-Jade C staining was used to label degenerating neurons within the TBI-induced cortical contusion region that, together with Annexin V positive neurons, were reduced by PFT-µ. Double immunofluorescence staining similarly demonstrated that PFT-µ significantly increased HO-1 positive neurons and mRNA expression in the cortical contusion region as well as decreased numbers of 4-hydroxynonenal (4HNE)-positive cells. Levels of mRNA encoding for p53, autophagy, mitophagy, anti-oxidant, anti-inflammatory related genes and proteins were measured by RT-qPCR and immunohistochemical staining, respectively. PFT-α, but not PFT-µ, significantly lowered p53 mRNA expression. Both PFT-α and PFT-µ lowered TBI-induced pro-inflammatory cytokines (IL-1ß and IL-6) mRNA levels as well as TBI-induced autophagic marker localization (LC3 and p62). Finally, treatment with PFT-µ mitigated TBI-induced declines in mRNA levels of PINK-1 and SOD2. Our data suggest that both PFT-µ and PFT-α provide neuroprotective actions through regulation of oxidative stress, neuroinflammation, autophagy, and mitophagy mechanisms, and that PFT-µ, in particular, holds promise as a TBI treatment strategy.


Assuntos
Autofagia/efeitos dos fármacos , Benzotiazóis/uso terapêutico , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/patologia , Encefalite/tratamento farmacológico , Mitofagia/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Sulfonamidas/uso terapêutico , Tolueno/análogos & derivados , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Antioxidantes/metabolismo , Comportamento Animal , Contusão Encefálica/tratamento farmacológico , Contusão Encefálica/patologia , Contusão Encefálica/psicologia , Lesões Encefálicas Traumáticas/psicologia , Citocinas/metabolismo , Encefalite/patologia , Heme Oxigenase (Desciclizante)/biossíntese , Masculino , Ratos , Ratos Sprague-Dawley , Tolueno/uso terapêutico
7.
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
8.
Neurosci Lett ; 709: 134386, 2019 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-31330225

RESUMO

Neuroinflammation is a predisposing factor for several neurodegenerative diseases. The purpose of this study was to evaluate the protective effect of madecassoside (MA) in lipopolysaccharide (LPS)-induced cognitive impairment and neuroinflammation in rats. MA has many protective effects such as antioxidant and anti-inflammatory properties. We investigated whether MA could improve neurocognitive dysfunction caused by intracerebroventricular injection of LPS. We examined the effects and mechanisms of action of MA on LPS-induced neuroinflammation in the cortex and hippocampus. Our study revealed that MA (120 mg/kg, i.g) treatment for 14 days reduced LPS-induced neurotoxicity by reducing cognitive impairments and suppressing the production of inflammatory cytokines such as interleukin 1 beta (IL-1ß), tumor necrosis factor alpha(TNF-α), and interleukin 6(IL-6) via activation of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Furthermore, MA treatment enhanced protein levels of heme oxygenase (HO)-1 by upregulating Nrf2 in LPS-stimulated neurotoxicity. Collectively, these results suggest that MA is effective in preventing neurodegenerative diseases by improving memory functions due to its anti-inflammatory activities and activation of Keap1-Nrf2/HO-1 signaling. As such, MA may be a potential therapy for addressing memory impairment caused by neuroinflammation.


Assuntos
Heme Oxigenase (Desciclizante)/biossíntese , Lipopolissacarídeos/toxicidade , Fator 2 Relacionado a NF-E2/biossíntese , Transtornos Neurocognitivos/tratamento farmacológico , Transtornos Neurocognitivos/metabolismo , Triterpenos/administração & dosagem , Animais , Feminino , Injeções Intraventriculares , Masculino , Fator 2 Relacionado a NF-E2/agonistas , Transtornos Neurocognitivos/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
9.
Neuroscience ; 410: 140-149, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31085280

RESUMO

Sestrin2 (Sesn2) is a stress response protein which expresses neuroprotective characteristics in some neurodegenerative disorders. However, the impact of Sesn2 on the clinical outcome of stroke is unclear. The nuclear factor-erythroid 2 related factor 2/heme oxygenase-1 (Nrf2/HO-1) pathway is a key factor in angiogenesis, which aids in attenuating cerebral ischemia damage. In this study the investigators examine the effects of Sesn2 on cerebral ischemia damage by increasing angiogenesis through the Nrf2/HO-1 signaling pathway. Healthy adult Sprague-Dawley (SD) rats were exposed to photochemical cerebral ischemia while AAV injection was used to overexpress Sesn2. At 5 days after photochemical embolization, the investigators observed a reduction in neurological problems, decreased infarct volume, and diminished neuronal injury in the Sesn2 overexpression samples compared to the controls. To further explore these defensive mechanisms, the investigators also silenced Nrf2. While Sesn2, Nrf2, HO-1, and VEGF were significantly increased following cerebral ischemia, overexpression of Sesn2 further increased their expression. After silencing Nrf2, the opposite effect was observed. These results imply that Sestrin2 may activate the Nrf2 / HO-1 pathway, leading to enhanced angiogenesis following photothrombotic cerebral ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Heme Oxigenase (Desciclizante)/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Neovascularização Patológica/metabolismo , Proteínas Nucleares/biossíntese , Animais , Isquemia Encefálica/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Expressão Gênica , Heme Oxigenase (Desciclizante)/genética , Masculino , Fator 2 Relacionado a NF-E2/genética , Proteínas Nucleares/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
10.
Stem Cell Res Ther ; 10(1): 85, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30867050

RESUMO

BACKGROUND: Islets of Langerhans transplantation is a promising therapy for type 1 diabetes mellitus, but this technique is compromised by transplantation stresses including inflammation. In other tissues, co-transplantation with mesenchymal stem cells has been shown to reduce damage by improving anti-inflammatory and anti-oxidant defences. Therefore, we probed the protection afforded by bone marrow mesenchymal stem cells to islets under pro-inflammatory cytokine stress. METHODS: In order to evaluate the cytoprotective potential of mesenchymal stem cells on rat islets, co-cultures were exposed to the interleukin-1, tumour necrosis factor α and interferon γ cocktail for 24 h. Islet viability and functionality tests were performed. Reactive oxygen species and malondialdehyde were measured. Expression of stress-inducible genes acting as anti-oxidants and detoxifiers, such as superoxide dismutases 1 and 2, NAD(P)H quinone oxidoreductase 1, heme oxygenase-1 and ferritin H, was compared to non-stressed cells, and the corresponding proteins were measured. Data were analysed by a two-way ANOVA followed by a Holm-Sidak post hoc analysis. RESULTS: Exposure of rat islets to cytokines induces a reduction in islet viability and functionality concomitant with an oxidative status shift with an increase of cytosolic ROS production. Mesenchymal stem cells did not significantly increase rat islet viability under exposure to cytokines but protected islets from the loss of insulin secretion. A drastic reduction of the antioxidant factors heme oxygenase-1 and ferritin H protein levels was observed in islets exposed to the cytokine cocktail with a prevention of this effect by the presence of mesenchymal stem cells. CONCLUSIONS: Our data evidenced that MSCs are able to preserve islet insulin secretion through a modulation of the oxidative imbalance mediated by heme and iron via heme oxygenase-1 and ferritin in a context of cytokine exposure.


Assuntos
Citocinas/farmacologia , Ferritinas/biossíntese , Heme Oxigenase (Desciclizante)/biossíntese , Ilhotas Pancreáticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Animais , Técnicas de Cocultura , Humanos , Ilhotas Pancreáticas/citologia , Células-Tronco Mesenquimais/citologia , Ratos
11.
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
12.
Curr Clin Pharmacol ; 14(1): 61-67, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30179140

RESUMO

BACKGROUND: Cilostazol, a phosphodiesterase 3 inhibitor (PDE3I), is a platelet aggregation inhibitor and vasodilator that is useful for treating intermittent claudication. Experimental studies have shown that cilostazol has potent anti-inflammatory, anti-oxidant effects effects. OBJECTIVES: Although the hepatoprotective effect cilostazol has been studied, the molecular mechanisms of such protection, including: the nuclear factor-erythroid 2-related factor 2 (Nrf2) / hemoxygenase (HO-1) and the phosphoinositide 3-kinase (PI3K) /serine/threonine kinase (Akt) pathways are not fully explored, which is the aim of this study. METHODS: To achieve the aim of this study, 35 rats were grouped into: control groups, liver injury group (model- non treated: injected with thioacetamide (TAA), 150 mg/kg, i.p.), and two cilostazoltreated groups (treated with cilostazol 10 and 50 mg/kg, p.o.). The rats were treated for 8 days and injected with TAA on the 7th day of the experiment and sacrificed 48 hours after TAA injection. RESULTS: The model group showed evidence of liver injury as indicated by the elevation of liver enzymes and confirmed by histopathological findings. TAA-induced liver injury was accompanied by down-regulation of the cytoprotective pathways: PI3K/Akt and Nrf2/HO-1 mRNAs. Cilostazol administration ameliorated TAA-induced liver injury, where it caused a significant improvement in the activity of liver enzymes as well as in the histopathological changes. Such an effect was associated with a significant increase in the expression of PI3K/Akt and Nrf2/HO-1 mRNAs as detected by Real-time reverse transcription polymerase chain reaction (RT-PCR). CONCLUSION: Cilostazol protected rats against TAA hepatotoxicity through up-regulation of PI3K/Akt and Nrf2/HO-1 gene expression.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Cilostazol/uso terapêutico , Heme Oxigenase (Desciclizante)/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Fosfatidilinositol 3-Quinases/biossíntese , Proteínas Proto-Oncogênicas c-akt/biossíntese , Animais , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Masculino , Inibidores da Fosfodiesterase 3/uso terapêutico , Ratos , Tioacetamida/toxicidade , Resultado do Tratamento
13.
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
14.
Toxicol Lett ; 288: 111-118, 2018 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-29447957

RESUMO

Lead (Pb) is an environmental neurotoxic metal. Chronic exposure to Pb causes deficits of learning and memory in children and spatial learning deficits in developing rats. In this study we investigated the effects of Pb exposure on the expression of HMGB1 and HO-1 in rats and PC12 cells. The animals were randomly divided to three groups: control group; low lead exposure group; high lead exposure group; PC12 cells were divided into 3 groups: 0 µM (control group), 1 µM and 100 µM Pb acetate. The results showed that Pb levels in blood and brain of Pb exposed groups were significantly higher than that of the control group (p < 0.05). The expression of HMGB1 and HO-1 were increased in Pb exposed groups than that of the control group (p < 0.05). Moreover, we found that the up-regulation of HO-1 in Pb exposure environment inhibited the expression of HMGB1.


Assuntos
Proteína HMGB1/biossíntese , Heme Oxigenase (Desciclizante)/biossíntese , Intoxicação por Chumbo/genética , Chumbo/toxicidade , Animais , Encéfalo/metabolismo , Feminino , Proteína HMGB1/genética , Heme Oxigenase (Desciclizante)/genética , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Chumbo/farmacocinética , Intoxicação por Chumbo/psicologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Células PC12 , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
15.
J Pharm Pharmacol ; 70(3): 383-392, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29355950

RESUMO

OBJECTIVES: We previously demonstrated that lansoprazole provided hepatoprotection in a drug-induced hepatitis animal model partially through the Nrf2/HO-1 pathway. Here, we examined whether lansoprazole could also provide hepatoprotection in a rat model of non-alcoholic steatohepatitis (NASH). METHODS: Six-week-old rats were fed a normal chow or a choline-deficient amino acid-defined (CDAA) diet to establish a rat model of NASH. The groups fed a CDAA diet for 5 weeks were subcutaneously administered either a vehicle or a lansoprazole suspension for 4 weeks beginning the second week of the experiment. KEY FINDINGS: Bridging fibrosis was observed in the livers of almost all the NASH model rats (six of seven), but it was not always observed in NASH model rats (one of seven) continuously administered lansoprazole. The serum aspartate aminotransferase level elevated by the CDAA diet was significantly decreased following lansoprazole administration. Lansoprazole also increased the expression of Nrf2, but not HO-1, in the liver of NASH model rats. Lansoprazole decreased the level of activated TGF-ß protein. Furthermore, interleukin-6 gene and protein expression were decreased. CONCLUSIONS: Lansoprazole inhibits hepatic fibrogenesis, at least during the early stages, in CDAA diet-induced NASH model rats. The mechanisms might be associated with cytokine suppression but not the inhibition of reactive oxygen species.


Assuntos
Lansoprazol/farmacologia , Cirrose Hepática/prevenção & controle , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Animais , Aspartato Aminotransferases/sangue , Dieta , Modelos Animais de Doenças , Progressão da Doença , Heme Oxigenase (Desciclizante)/biossíntese , Interleucina-6/biossíntese , Cirrose Hepática/complicações , Masculino , Fator 2 Relacionado a NF-E2/biossíntese , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ratos , Fator de Crescimento Transformador beta/metabolismo
16.
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
17.
Oxid Med Cell Longev ; 2017: 2176749, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28770019

RESUMO

Estrogens and raloxifene (RAL) have beneficial effects on certain cardiovascular indices in postmenopausal women characterized by estrogen deficiency. Heme oxygenase (HO) activity is increased by 17ß-estradiol (E2) and RAL in estrogen-deficient rat resulting in vasorelaxation mediated by carbon monoxide. We determined the expressions of HO in cardiac and aortic tissues after ovariectomy (OVX) and subsequent RAL or E2 treatment. We investigated the effects of pharmacological inhibition of HO enzyme on the arginine vasopressin- (AVP-) induced blood pressure in vivo, the epinephrine- and phentolamine-induced electrocardiogram ST segment changes in vivo, and the myeloperoxidase (MPO) enzyme activity. When compared with intact females, OVX decreased the HO-1 and HO-2 expression, aggravated the electrocardiogram signs of heart ischemia and the blood pressure response to AVP, and increased the cardiac MPO. E2 and RAL are largely protected against these negative impacts induced by OVX. The pharmacological inhibition of HO in E2- or RAL-treated OVX animals, however, restored the cardiovascular status close to that observed in nontreated OVX animals. The decreased expression of HO enzymes and the changes in blood pressure ischemia susceptibility and inflammatory state in OVX rat can be reverted by the administration of E2 or RAL partly through its antioxidant and anti-inflammatory roles.


Assuntos
Cardiotônicos/farmacologia , Estrogênios/deficiência , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase (Desciclizante)/biossíntese , Isquemia Miocárdica/prevenção & controle , Cloridrato de Raloxifeno/farmacologia , Receptores de Estrogênio/metabolismo , Animais , Feminino , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Ovariectomia , Ratos , Ratos Wistar
18.
J Neurotrauma ; 34(3): 695-706, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27526795

RESUMO

The mechanism by which spinal cord injury (SCI) induces neuronal death has not been thoroughly understood. Investigation on the molecular signal pathways involved in SCI-mediated neuronal apoptosis is important for development of new therapeutics for SCI. In the current study, we explore the role of heme oxygenase-1 (HO-1) in the modulation of mixed lineage kinase 3/mitogen-activated protein kinase kinase/cJUN N-terminal kinase 3 (MLK3/MKK7/JNK3) signaling, which is a pro-apoptotic pathway, after SCI. We found that MLK3/MKK7/JNK3 signaling was activated by SCI in a time-dependent manner, demonstrated by increase in activating phosphorylation of MLK3, MKK7, and JNK3. SCI also induced HO-1 expression. Administration of HO-1-expressing adeno-associated virus before SCI introduced expression of exogenous HO-1 in injured spinal cords. Exogenous HO-1 reduced phosphorylation of MLK3, MKK7, and JNK3. Consistent with its inhibitory effect on MLK3/MKK7/JNK3 signaling, exogenous HO-1 decreased SCI-induced neuronal apoptosis and improved neurological score. Further, we found that exogenous HO-1 inhibited expression of cell division cycle 42 (Cdc42), which is crucial for MLK3 activation. In vitro experiments indicated that Cdc42 was essential for neuronal apoptosis, while transduction of neurons with HO-1-expressing adeno-associated virus significantly reduced neuronal apoptosis to enhance neuronal survival. Therefore, our study disclosed a novel mechanism by which HO-1 exerted its neuroprotective efficacy. Our discovery might be valuable for developing a new therapeutic approach for SCI.


Assuntos
Heme Oxigenase (Desciclizante)/biossíntese , MAP Quinase Quinase 7/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Traumatismos da Medula Espinal/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Apoptose/fisiologia , Células Cultivadas , Regulação para Baixo/fisiologia , MAP Quinase Quinase 7/antagonistas & inibidores , MAP Quinase Quinase Quinases/antagonistas & inibidores , Masculino , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Neurônios/metabolismo , Neurônios/patologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Traumatismos da Medula Espinal/patologia , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
19.
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
20.
PLoS One ; 11(6): e0157280, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27280718

RESUMO

Aggressive tumors induce tumor-supporting changes in the benign parts of the prostate. One factor that has increased expression outside prostate tumors is hemoxygenase-1 (HO-1). To investigate HO-1 expression in more detail, we analyzed samples of tumor tissue and peritumoral normal prostate tissue from rats carrying cancers with different metastatic capacity, and human prostate cancer tissue samples from primary tumors and bone metastases. In rat prostate tumor samples, immunohistochemistry and quantitative RT-PCR showed that the main site of HO-1 synthesis was HO-1+ macrophages that accumulated in the tumor-bearing organ, and at the tumor-invasive front. Small metastatic tumors were considerably more effective in attracting HO-1+ macrophages than larger non-metastatic ones. In clinical samples, accumulation of HO-1+ macrophages was seen at the tumor invasive front, almost exclusively in high-grade tumors, and it correlated with the presence of bone metastases. HO-1+ macrophages, located at the tumor invasive front, were more abundant in bone metastases than in primary tumors. HO-1 expression in bone metastases was variable, and positively correlated with the expression of macrophage markers but negatively correlated with androgen receptor expression, suggesting that elevated HO-1 could be a marker for a subgroup of bone metastases. Together with another recent observation showing that selective knockout of HO-1 in macrophages reduced prostate tumor growth and metastatic capacity in animals, the results of this study suggest that extratumoral HO-1+ macrophages may have an important role in prostate cancer.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase-1/biossíntese , Macrófagos/enzimologia , Proteínas de Neoplasias/biossíntese , Neoplasias da Próstata/enzimologia , Animais , Humanos , Macrófagos/patologia , Masculino , Metástase Neoplásica , Neoplasias da Próstata/patologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA