Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 766
Filtrar
1.
Comput Math Methods Med ; 2022: 8920861, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35047060

RESUMEN

BACKGROUND: Hepatic stellate cells (HSCs) are reported to play significant roles in the development of liver fibrosis. Heme oxygenase-1 (HO-1) is a key rate-limiting enzyme, which could decrease collagen synthesis and liver damage. Nevertheless, it was yet elusive towards the function and mechanism of HO-1. METHODS: An HO-1 inducer Hemin or an HO-1 inhibitor ZnPP-IX was used to treat the activated HSC-T6, respectively. MTT assay was adopted to detect cell proliferation. Immunocytochemical staining was employed to test the levels of alpha-smooth muscle actin (α-SMA), peroxisome proliferator-activated receptor-γ (PPARγ), and nuclear factor-kappa B (NF-kappa B) levels in HSC-T6. HO-1, PPARγ, and NF-κB expression levels were measured by qRT-PCR and Western blotting. ELISA was then used to detect the levels of transforming growth factor- (TGF-) beta 1 (TGF-ß1), interleukin-6 (IL-6), serum hyaluronic acid (HA), and serum type III procollagen aminopeptide (PIIIP). RESULTS: HSC-T6 proliferation was inhibited in Hemin-treated HSCs. The levels of α-SMA, HA, and PIIIP and the production of ECM were lower in Hemin-treated HSCs, whereas those could be rescued by ZnPP-IX. NF-κB activation was decreased, but PPARγ expression was increased after HO-1 upregulation. Furthermore, the levels of TGF-ß1 and IL-6, which were downstream of activated NF-κB in HSC-T6, were reduced. The PPAR-specific inhibitor GW9662 could block those mentioned effects. CONCLUSIONS: Our data demonstrated that HO-1 induction could inhibit HSC proliferation and activation by regulating PPARγ expression and NF-κB activation directly or indirectly, which makes it a promising therapeutic target for liver fibrosis.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/metabolismo , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Biología Computacional , Inducción Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemina/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Modelos Biológicos , FN-kappa B/antagonistas & inhibidores , PPAR gamma/agonistas , PPAR gamma/genética , Protoporfirinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
2.
FASEB J ; 36(1): e22098, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34918390

RESUMEN

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.


Asunto(s)
Antioxidantes/metabolismo , Aorta/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Calcificación Vascular/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Modelos Animales de Enfermedad , Hemo Oxigenasa (Desciclizante)/biosíntesis , Masculino , NAD(P)H Deshidrogenasa (Quinona)/biosíntesis , Ratas , Ratas Wistar
3.
Oxid Med Cell Longev ; 2021: 5521196, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34194603

RESUMEN

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.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Compuestos Organometálicos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/biosíntesis , Humanos , Ratas , Transfección
4.
Oxid Med Cell Longev ; 2020: 4640605, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33354277

RESUMEN

BACKGROUND: Di-N-butylphthalate (DBP) is a kind of unique endocrine toxicity linked to hormonal disruptions that affects the male reproductive system and has given rise to more and more attention. However, the mechanism of DBP-induced testicular injury remains unclear. Here, the objective of this study was to investigate the potential molecular mechanism of miR-506-3p in DBP-induced rat testicular oxidative stress injury via ANXA5 (Annexin A5)/Nrf2/HO-1 signaling pathway. METHODS: In vivo, a total of 40 adolescent male rats were treated from 2 weeks with 800 mg/kg/day of DBP in 1 mL/kg corn oil administered daily by oral gavage. Among them, some rats were also injected subcutaneously with 2 nmol agomir-506-3p and/or 10 nmol recombinant rat ANXA5. The pathomorphological changes of testicular tissue were assessed by histological examination, and the antioxidant factors were evaluated. Subsequently, ANXA5, Nrf2, and its dependent antioxidant enzymes, such as HO-1, NQO1, and GST, were detected by Western blotting or immunohistochemical staining. In vitro, TM3 cells (Leydig cells) were used to detect the cell activity by CCK-8 and the transfection in the DBP-treated group. RESULTS: Differentially expressed miRNAs between the DBP-treated and normal rats were analyzed, and qRT-PCR showed miR-506-3p was highly expressed in testicular tissues of the DBP-treated rats. DBP-treated rats presented severe inflammatory infiltration, increased abnormal germ cells, and missed cell layers frequently existed in seminiferous tubules, resulted in oxidative stress and decreased testicular function. Meanwhile, upregulation of miR-506-3p aggravated the above changes. In addition, miR-506-3p directly bound to ANXA5, and overexpression of miR-506-3p could reduce the ANXA5 expression and also decrease the protein levels of Nrf2/HO-1 signaling pathway. Additionally, we found that recombinant rat ANXA5 reversed the DBP-treated testicular oxidative stress promoting injury of miR-506-3p in rats. In vivo results were reproduced in in vitro experiments. CONCLUSIONS: This study provided evidence that miR-506-3p could aggravate the DBP-treated testicular oxidative stress injury in vivo and in vitro by inhibiting ANXA5 expression and downregulating Nrf2/HO-1 signaling pathway, which might provide novel understanding in DBP-induced testicular injury therapy.


Asunto(s)
Anexina A5/biosíntesis , Dibutil Ftalato/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/biosíntesis , MicroARNs/biosíntesis , Transducción de Señal/efectos de los fármacos , Testículo/metabolismo , Animales , Masculino , Factor 2 Relacionado con NF-E2 , Ratas , Ratas Sprague-Dawley , Testículo/patología
5.
Drug Deliv ; 27(1): 1329-1341, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32945205

RESUMEN

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.


Asunto(s)
Artritis Reumatoide/metabolismo , Hemo Oxigenasa (Desciclizante)/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , FN-kappa B/biosíntesis , Nanopartículas/administración & dosificación , Sitoesteroles/administración & dosificación , Animales , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/administración & dosificación , Adyuvante de Freund/toxicidad , Hipolipemiantes/administración & dosificación , Masculino , Factor 2 Relacionado con NF-E2/agonistas , FN-kappa B/antagonistas & inhibidores , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
6.
Biomolecules ; 10(3)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32121307

RESUMEN

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.


Asunto(s)
Antioxidantes/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/biosíntesis , Clorhidrato de Raloxifeno/uso terapéutico , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Femenino , Humanos
7.
Neuromolecular Med ; 22(2): 278-292, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31900786

RESUMEN

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.


Asunto(s)
Harpagophytum/química , Hiperalgesia/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Fitoterapia , Extractos Vegetales/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Aldehídos/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo Oxigenasa (Desciclizante)/genética , Hiperalgesia/etiología , Inflamación , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Ácido Nítrico/metabolismo , Extractos Vegetales/química , Extractos Vegetales/farmacología , Ratas , Ratas Sprague-Dawley , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Método Simple Ciego , Tacto
8.
Biotechnol Appl Biochem ; 67(3): 317-322, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31746064

RESUMEN

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.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Catequina/análogos & derivados , Citocinas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/metabolismo , Catequina/farmacología , Colágeno Tipo II , Citocinas/biosíntesis , Femenino , Hemo Oxigenasa (Desciclizante)/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , Ratas , Ratas Wistar
9.
Exp Neurol ; 324: 113135, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31778663

RESUMEN

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.


Asunto(s)
Autofagia/efectos de los fármacos , Benzotiazoles/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Encefalitis/tratamiento farmacológico , Mitofagia/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Sulfonamidas/uso terapéutico , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Animales , Antioxidantes/metabolismo , Conducta Animal , Contusión Encefálica/tratamiento farmacológico , Contusión Encefálica/patología , Contusión Encefálica/psicología , Lesiones Traumáticas del Encéfalo/psicología , Citocinas/metabolismo , Encefalitis/patología , Hemo Oxigenasa (Desciclizante)/biosíntesis , Masculino , Ratas , Ratas Sprague-Dawley , Tolueno/uso terapéutico
10.
Naunyn Schmiedebergs Arch Pharmacol ; 392(11): 1435-1445, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31273394

RESUMEN

Oxidative stress and persistent inflammation play crucial role in the progression of diabetic wound complications. Hemeoxgenase-1 (HO-1) by degrading hemin has been shown to display anti-oxidant and anti-inflammatory effects. Further, hemin is a potent HO-1 inducer. Thus, the current study was aimed to evaluate the effect of topical application of hemin on diabetic wound in rats. Four hundred square millimeter open excision wound were created 2 weeks after induction of diabetes with single intraperitoneal injection of streptozotocin (60 mg/kg), and the diabetic rats were divided into three groups namely diabetic control, hemin, and tin protoporphyrin (SnPPIX). Ointment base, hemin (0.5% in ointment base), and SnPPIX (0.5% in ointment base) were applied topically to wounded area in diabetic control, hemin, and SnPPIX group rats, respectively, twice daily for 19 days. Hemin significantly increased the wound contraction in comparison to control and SnPPIX-treated rats. Time-dependent analysis revealed significant increase in anti-oxidants with concomitant decrease in oxidants in hemin-treated rats as compared to diabetic control rats. Further, mRNA expression decreased for inflammatory cytokine and increased for anti-inflammatory cytokine in hemin group as compared to diabetic control rats. Expression of HO-1 also increased in hemin group as compared to diabetic control rats. However, SnPPIX group results were in disagreement with results of hemin which is clearly reflected in histopathology. Results indicate the ability of hemin to accelerate wound healing in diabetic rats by combating inflammation and oxidative stress probably via HO-1.


Asunto(s)
Antioxidantes/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Hemina/farmacología , Mediadores de Inflamación/metabolismo , Estrés Oxidativo/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Administración Cutánea , Animales , Antioxidantes/administración & dosificación , Glucemia/análisis , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/metabolismo , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemina/administración & dosificación , Masculino , Metaloporfirinas/administración & dosificación , Metaloporfirinas/farmacología , Pomadas , Estrés Oxidativo/inmunología , Protoporfirinas/administración & dosificación , Protoporfirinas/farmacología , Ratas , Piel/efectos de los fármacos , Piel/inmunología , Piel/lesiones , Estreptozocina , Cicatrización de Heridas/inmunología
11.
Neurosci Lett ; 709: 134386, 2019 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-31330225

RESUMEN

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.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/biosíntesis , Lipopolisacáridos/toxicidad , Factor 2 Relacionado con NF-E2/biosíntesis , Trastornos Neurocognitivos/tratamiento farmacológico , Trastornos Neurocognitivos/metabolismo , Triterpenos/administración & dosificación , Animales , Femenino , Inyecciones Intraventriculares , Masculino , Factor 2 Relacionado con NF-E2/agonistas , Trastornos Neurocognitivos/inducido químicamente , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
12.
Alcohol Clin Exp Res ; 43(8): 1651-1661, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31141180

RESUMEN

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.


Asunto(s)
Ácido Aminolevulínico/farmacología , Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Etanol/efectos adversos , Hepatitis/prevención & control , Intestinos/fisiopatología , Permeabilidad/efectos de los fármacos , Alanina Transaminasa/sangre , Animales , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Antígenos de Diferenciación Mielomonocítica/inmunología , Aspartato Aminotransferasas/sangre , Moléculas de Adhesión Celular Neuronal/genética , Ácido Cítrico , Endotoxinas/sangre , Infecciones por Enterobacteriaceae/microbiología , Compuestos Ferrosos , Infecciones por VIH/complicaciones , VIH-1/genética , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo-Oxigenasa 1/biosíntesis , Hepatitis/sangre , Hepatitis/complicaciones , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/sangre , Hígado/metabolismo , Ratas , Ratas Transgénicas , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/inmunología , Sirtuina 1/biosíntesis , Células Madre , Triglicéridos/metabolismo , Proteína de la Zonula Occludens-1/biosíntesis , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
13.
Neuroscience ; 410: 140-149, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31085280

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Hemo Oxigenasa (Desciclizante)/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , Neovascularización Patológica/metabolismo , Proteínas Nucleares/biosíntesis , Animales , Isquemia Encefálica/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Expresión Génica , Hemo Oxigenasa (Desciclizante)/genética , Masculino , Factor 2 Relacionado con NF-E2/genética , Proteínas Nucleares/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
14.
Stem Cell Res Ther ; 10(1): 85, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30867050

RESUMEN

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.


Asunto(s)
Citocinas/farmacología , Ferritinas/biosíntesis , Hemo Oxigenasa (Desciclizante)/biosíntesis , Islotes Pancreáticos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Estrés Fisiológico/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Técnicas de Cocultivo , Humanos , Islotes Pancreáticos/citología , Células Madre Mesenquimatosas/citología , Ratas
15.
Neurochem Int ; 126: 96-108, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30880045

RESUMEN

Post stroke recanalization has been associated with increased risk of oxidative stress. Stimulating endogenous antioxidant pathway by activation of nuclear factor erythroid-2-related factor-2 (Nrf2) plays a key role in neuronal defense against inflammation and oxidative stress in penumbra. Here, we explored whether monomethyl fumarate (MMF) could produce neuro-protection after ischemia/reperfusion (I/R) injury via Nrf2/HO1 activation. In male SD rats, middle cerebral artery was occluded for 90 min and confirmed using Laser Doppler flowmeter. MMF (10, 20 and 40 mg/kg) was administered in two divided doses at 30 min post ischemia and 5-10 min after reperfusion. After 24 h, effect on neurobehavioral parameters, infarct damage by TTC staining and MRI, oxidative stress and inflammatory cytokines were assessed. Expression studies of nuclear Nrf2 and cytoplasmic HO1 were performed in peri-infarct cortex and striatum; followed by dual immunofluorescence study to check the specific cell type. I/R induced neurobehavioral deficits and infarct damage were significantly (p < 0.05) attenuated by MMF (20 and 40 mg/kg). MMF, 20 mg/kg, significantly normalized I/R induced altered redox status and increased levels of TNF-α, IL-1ß in the ipsilateral cortex. MRI data showed significantly reduced infarct in cortex but not in striatum after MMF treatment. Expression of nuclear Nrf2 and cytoplasmic HO1 were significantly (p < 0.05) increased in peri-infarct cortex after treatment with MMF. Additionally, dual immunofluorescence showed increased Nrf2 expression in neurons and HO1 expression in neurons as well as astrocytes in peri-infarct cortex after MMF treatment. Our results show the neuro-protective potential of MMF probably by restricting the progression of damage from striatum to cortex through activation of Nrf2/HO1 pathway in peri-infarct cortex.


Asunto(s)
Fumaratos/uso terapéutico , Hemo Oxigenasa (Desciclizante)/biosíntesis , Infarto de la Arteria Cerebral Media/metabolismo , Maleatos/uso terapéutico , Factor 2 Relacionado con NF-E2/biosíntesis , Fármacos Neuroprotectores/uso terapéutico , Daño por Reperfusión/metabolismo , Animales , Fumaratos/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Masculino , Maleatos/farmacología , Factor 2 Relacionado con NF-E2/agonistas , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
Biomed Pharmacother ; 111: 676-685, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30611992

RESUMEN

Olive leaf extract (OLE) has potential health benefits and protects against cytotoxicity in different organs. However, nothing has yet been reported on its potential to prevent cyclophosphamide (CP)-induced nephrotoxicity. This study investigated the possible protective effect of OLE on CP-induced kidney injury in rats, focusing on oxidative stress, inflammation, apoptosis and Nrf2/ARE/HO-1 signaling. Rats received 100 or 200 mg/kg body weight OLE for 15 days and a single injection of 150 mg/kg CP at day 16. CP induced kidney injury evidenced by the significantly increased serum creatinine and urea, and histopathological alterations, including glomerular atrophy, interstitial hemorrhage, dilated urinary space and necrosis. CP-induced rats exhibited increased kidney lipid peroxidation, protein carbonyl, nitric oxide (NO) and pro-inflammatory cytokines, and up-regulated NF-κB, Bax, cytochrome c and caspase-3. OLE ameliorated kidney function markers and prevented CP-induced tissue damage. In addition, OLE significantly prevented oxidative stress, inflammation and apoptosis by enhancing the antioxidant defenses and Bcl-2 expression, and suppressing the pro-inflammatory and pro-apoptotic markers NF-κB, Bax, cytochrome c and caspase-3. OLE up-regulated Nrf2, HO-1 and NQO-1 expression in the kidney of CP-induced rats. In conclusion, OLE has a substantial protective role against CP-induced nephrotoxicity in rats by up-regulating the Nrf2/ARE/HO-1 signaling, enhancing the antioxidant activity and attenuating inflammation and apoptosis.


Asunto(s)
Elementos de Respuesta Antioxidante/efectos de los fármacos , Ciclofosfamida/toxicidad , Hemo Oxigenasa (Desciclizante)/biosíntesis , Riñón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/biosíntesis , Olea , Estrés Oxidativo/efectos de los fármacos , Animales , Elementos de Respuesta Antioxidante/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Relación Dosis-Respuesta a Droga , Depuradores de Radicales Libres/aislamiento & purificación , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Inmunosupresores/toxicidad , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/prevención & control , Riñón/metabolismo , Riñón/patología , Masculino , Estrés Oxidativo/fisiología , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Hojas de la Planta , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
17.
J Cereb Blood Flow Metab ; 39(2): 352-366, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29533123

RESUMEN

The major pathophysiological process of vascular cognitive impairment (VCI) is chronic cerebral ischemia, which causes disintegration of the blood-brain barrier (BBB), neuronal death, and white matter injury. This study aims to test whether sulforaphane (Sfn), a natural activator of nuclear factor erythroid 2-related factor 2 (Nrf2), reduces the chronic ischemic injury and cognitive dysfunction after VCI. Experimental VCI was induced in rats by permanent occlusion of both common carotid arteries for six weeks. This procedure caused notable neuronal death in the cortex and hippocampal CA1, myelin loss in the corpus callosum and hippocampal fimbria, accumulation of myelin debris in the corpus callosum, and remarkable cognitive impairment. Sfn treatment alleviated these ischemic injuries and the cognitive dysfunction. Sfn-mediated neuroprotection was associated with enhanced activation of Nrf2 and upregulation of heme oxygenase 1. Sfn also reduced neuronal and endothelial death and maintained the integrity of BBB after oxygen-glucose deprivation in vitro in an Nrf2 dependent manner. Furthermore, Nrf2 knockdown in endothelial cells decreased claudin-5 protein expression with downregulated claudin-5 promoter activity, suggesting that claudin-5 might be a target gene of Nrf2. Our results demonstrate that Sfn provides robust neuroprotection against chronic brain ischemic injury and may be a promising agent for VCI treatment.


Asunto(s)
Isquemia Encefálica , Encéfalo , Disfunción Cognitiva , Isotiocianatos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/prevención & control , Muerte Celular/efectos de los fármacos , Enfermedad Crónica , Claudina-5/metabolismo , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/prevención & control , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/biosíntesis , Masculino , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Sulfóxidos , Regulación hacia Arriba/efectos de los fármacos
18.
Endocrine ; 63(1): 70-78, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30128961

RESUMEN

PURPOSE: Oxidative stress is an important mechanism for diabetic nephropathy. Studies showed that hemo oxygenase-1 (HO-1) expression in renal tissue of patients with diabetic nephropathy has upregulated, while the HO-1 can protect the body through anti-oxidative stress. The study aimed to preliminarily explore the molecular mechanism by observing the effect of Sitagliptin on HO-1 expression in renal tissue of rats with diabetic nephropathy. METHODS: The diabetic nephropathy rat model was established by STZ injection followed by intraperitoneal injection of sitagliptin with different concentrations. The mRNA expressions of HO-1 were detected by real-time PCR and Western blot and HO-1 enzyme activity change was detected by colorimetry. Human renal mesangial cell (HRMC) were cultured in vitro with high glucose concentration (30 µmol/L), phosphatidylinositol-3-kinase (PI3K) level and nuclear factor erythroid-2-related factor (Nrf2) content in cytoplasm and cell nucleus were observed before and after treatment with sitagliptin, as well as the action of in meditating HO-1 expression. RESULTS: HO-1 mRNA, protein level, and HO-1 enzyme activity in renal tissue of rats with diabetic nephropathy were significantly increased after treatment with sitagliptin (P < 0.05). As comparison, the 24 h urinary microalbumin, creatinine, and boold urea nitrogen were all decreased after treatment of sitagliptin (P < 0.05). Similar results were observed after CoPP (an agonist of HO-1) treatment (P < 0.05). In contrast, ZnPP, an inhibitor of HO-1, significantly abrogated the inhibitory effect of sitagliptin (P < 0.05). Phosphorylation of PI3K and Nrf2 nuclear translocation under high-glucose concentration condition was induced by sitagliptin in HRMC. HO-1 expression was suppressed by pretreating HRMC with PI3K inhibitor or RNA interference. CONCLUSIONS: Sitagliptin may induce HO-1 expression via activation of PI3K and Nrf2 in rats with diabetic nephropathy; HO-1 can improve the oxidative stress of diabetic nephropathy, eventually protect from diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/enzimología , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hipoglucemiantes/uso terapéutico , Fosfato de Sitagliptina/uso terapéutico , Animales , Células Cultivadas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Nefropatías Diabéticas/patología , Mesangio Glomerular/citología , Mesangio Glomerular/efectos de los fármacos , Glucosa/farmacología , Hemo Oxigenasa (Desciclizante)/efectos de los fármacos , Humanos , Riñón/patología , Pruebas de Función Renal , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
19.
Curr Clin Pharmacol ; 14(1): 61-67, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30179140

RESUMEN

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.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Cilostazol/uso terapéutico , Hemo Oxigenasa (Desciclizante)/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , Fosfatidilinositol 3-Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Masculino , Inhibidores de Fosfodiesterasa 3/uso terapéutico , Ratas , Tioacetamida/toxicidad , Resultado del Tratamiento
20.
Clin Sci (Lond) ; 133(1): 117-134, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30538149

RESUMEN

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.


Asunto(s)
Antioxidantes/farmacología , Colestasis/prevención & control , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemina/farmacología , Hígado/efectos de los fármacos , Estrés Oxidativo , Animales , Bilis/metabolismo , Bilirrubina/metabolismo , Catalasa/metabolismo , Colestasis/inducido químicamente , Colestasis/enzimología , Colestasis/patología , Modelos Animales de Enfermedad , Inducción Enzimática , Glutatión/metabolismo , Hígado/enzimología , Hígado/patología , Masculino , Ratas Wistar , Superóxido Dismutasa/metabolismo , terc-Butilhidroperóxido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...