Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nature ; 565(7737): 96-100, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30487609

RESUMEN

Endothelial nitric oxide synthase (eNOS) is protective against kidney injury, but the molecular mechanisms of this protection are poorly understood1,2. Nitric oxide-based cellular signalling is generally mediated by protein S-nitrosylation, the oxidative modification of Cys residues to form S-nitrosothiols (SNOs). S-nitrosylation regulates proteins in all functional classes, and is controlled by enzymatic machinery that includes S-nitrosylases and denitrosylases, which add and remove SNO from proteins, respectively3,4. In Saccharomyces cerevisiae, the classic metabolic intermediate co-enzyme A (CoA) serves as an endogenous source of SNOs through its conjugation with nitric oxide to form S-nitroso-CoA (SNO-CoA), and S-nitrosylation of proteins by SNO-CoA is governed by its cognate denitrosylase, SNO-CoA reductase (SCoR)5. Mammals possess a functional homologue of yeast SCoR, an aldo-keto reductase family member (AKR1A1)5 with an unknown physiological role. Here we report that the SNO-CoA-AKR1A1 system is highly expressed in renal proximal tubules, where it transduces the activity of eNOS in reprogramming intermediary metabolism, thereby protecting kidneys against acute kidney injury. Specifically, deletion of Akr1a1 in mice to reduce SCoR activity increased protein S-nitrosylation, protected against acute kidney injury and improved survival, whereas this protection was lost when Enos (also known as Nos3) was also deleted. Metabolic profiling coupled with unbiased mass spectrometry-based SNO-protein identification revealed that protection by the SNO-CoA-SCoR system is mediated by inhibitory S-nitrosylation of pyruvate kinase M2 (PKM2) through a novel locus of regulation, thereby balancing fuel utilization (through glycolysis) with redox protection (through the pentose phosphate shunt). Targeted deletion of PKM2 from mouse proximal tubules recapitulated precisely the protective and mechanistic effects of S-nitrosylation in Akr1a1-/- mice, whereas Cys-mutant PKM2, which is refractory to S-nitrosylation, negated SNO-CoA bioactivity. Our results identify a physiological function of the SNO-CoA-SCoR system in mammals, describe new regulation of renal metabolism and of PKM2 in differentiated tissues, and offer a novel perspective on kidney injury with therapeutic implications.


Asunto(s)
Lesión Renal Aguda/enzimología , Lesión Renal Aguda/prevención & control , Coenzima A/metabolismo , Ingeniería Metabólica , Oxidorreductasas/metabolismo , Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Animales , Línea Celular , Femenino , Glucólisis , Células HEK293 , Humanos , Túbulos Renales Proximales/enzimología , Masculino , Ratones , Mutación , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxidación-Reducción , Vía de Pentosa Fosfato , Multimerización de Proteína , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/deficiencia , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo
2.
J Mol Cell Cardiol ; 118: 183-192, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29627295

RESUMEN

Pathological cardiac hypertrophy is associated with the accumulation of lipid peroxidation-derived aldehydes such as 4-hydroxy-trans-2-nonenal (HNE) and acrolein in the heart. These aldehydes are metabolized via several pathways, of which aldose reductase (AR) represents a broad-specificity route for their elimination. We tested the hypothesis that by preventing aldehyde removal, AR deficiency accentuates the pathological effects of transverse aortic constriction (TAC). We found that the levels of AR in the heart were increased in mice subjected to TAC for 2 weeks. In comparison with wild-type (WT), AR-null mice showed lower ejection fraction, which was exacerbated 2 weeks after TAC. Levels of atrial natriuretic peptide and myosin heavy chain were higher in AR-null than in WT TAC hearts. Deficiency of AR decreased urinary levels of the acrolein metabolite, 3-hydroxypropylmercapturic acid. Deletion of AR did not affect the levels of the other aldehyde-metabolizing enzyme - aldehyde dehydrogenase 2 in the heart, or its urinary product - (N-Acetyl-S-(2-carboxyethyl)-l-cystiene). AR-null hearts subjected to TAC showed increased accumulation of HNE- and acrolein-modified proteins, as well as increased AMPK phosphorylation and autophagy. Superfusion with HNE led to a greater increase in p62, LC3II formation, and GFP-LC3-II punctae formation in AR-null than WT cardiac myocytes. Pharmacological inactivation of JNK decreased HNE-induced autophagy in AR-null cardiac myocytes. Collectively, these results suggest that during hypertrophy the accumulation of lipid peroxidation derived aldehydes promotes pathological remodeling via excessive autophagy, and that metabolic detoxification of these aldehydes by AR may be essential for maintaining cardiac function during early stages of pressure overload.


Asunto(s)
Aldehído Reductasa/deficiencia , Autofagia , Corazón/fisiopatología , Presión , Aldehído Reductasa/metabolismo , Aldehídos/metabolismo , Animales , Aorta/patología , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/enzimología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Constricción Patológica , Eliminación de Gen , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones Endogámicos C57BL , Contracción Miocárdica , Miocardio/enzimología , Proteína Sequestosoma-1/metabolismo
3.
Biochem Biophys Res Commun ; 478(2): 765-71, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27501753

RESUMEN

Aldehyde reductase (Akr1a) has been reported to be involved in the biosynthesis of ascorbic acid (AsA) in the mouse liver. Because Akr1a is expressed at high levels in the liver, we aimed to investigate the role of Akr1a in liver homeostasis by employing a carbon tetrachloride (CCl4)-induced hepatotoxicity model. Akr1a-deficient (Akr1a(-/-)) and wild-type (WT) mice were injected intraperitoneally with CCl4 and the extent of hepatic injury in the acute phase was assessed. Liver damage was heavier in the Akr1a(-/-) mice than in the WT mice. Furthermore, severe hepatic steatosis was observed in the livers of Akr1a(-/-) mice compared to WT mice and was restored to the levels in WT mice by AsA supplementation. Since the presence or absence of AsA had no effect on the decrease in CYP2E1 activity after the CCl4 treatment, it appears that AsA plays a role in the process after the bioactivation of CCl4. Biomarkers for oxidative stress and ER stress were markedly increased in the livers of Akr1a(-/-) mice and were effectively suppressed by AsA supplementation. Based on these collective results, we conclude that Akr1a exerts a protective effect against CCl4-induced hepatic steatosis by replenishing AsA via its antioxidative properties.


Asunto(s)
Aldehído Reductasa/deficiencia , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Estrés del Retículo Endoplásmico/genética , Hígado Graso/genética , Estrés Oxidativo/genética , Aldehído Reductasa/genética , Animales , Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Biomarcadores/metabolismo , Tetracloruro de Carbono , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Citocromo P-450 CYP2E1/genética , Citocromo P-450 CYP2E1/metabolismo , Chaperón BiP del Retículo Endoplásmico , Hígado Graso/inducido químicamente , Hígado Graso/enzimología , Hígado Graso/prevención & control , Expresión Génica , Glutatión/agonistas , Glutatión/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Ratones , Ratones Noqueados , Perilipina-2/genética , Perilipina-2/metabolismo , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo
4.
Graefes Arch Clin Exp Ophthalmol ; 253(9): 1503-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25921391

RESUMEN

Retinopathy of prematurity (ROP) is a leading cause of childhood blindness where vascular abnormality and retinal dysfunction are reported. We showed earlier that genetic deletion of aldose reductase (AR), the rate-limiting enzyme in the polyol pathway, reduced the neovascularization through attenuating oxidative stress induction in the mouse oxygen-induced retinopathy (OIR) modeling ROP. In this study, we further investigated the effects of AR deficiency on retinal neurons in the mouse OIR. Seven-day-old wild-type and AR-deficient mice were exposed to 75 % oxygen for 5 days and then returned to room air. Electroretinography was used to assess the neuronal function at postnatal day (P) 30. On P17 and P30, retinal cytoarchitecture was examined by morphometric analysis and immunohistochemistry for calbindin, protein kinase C alpha, calretinin, Tuj1, and glial fibrillary acidic protein. In OIR, attenuated amplitudes and delayed implicit time of a-wave, b-wave, and oscillatory potentials were observed in wild-type mice, but they were not significantly changed in AR-deficient mice. The morphological changes of horizontal, rod bipolar, and amacrine cells were shown in wild-type mice and these changes were partly preserved with AR deficiency. AR deficiency attenuated the Müller cell gliosis induced in OIR. Our observations demonstrated AR deficiency preserved retinal functions in OIR and AR deficiency could partly reduce the extent of retinal neuronal histopathology. These findings suggested a therapeutic potential of AR inhibition in ROP treatment with beneficial effects on the retinal neurons.


Asunto(s)
Aldehído Reductasa/deficiencia , Modelos Animales de Enfermedad , Gliosis/prevención & control , Neuronas Retinianas/enzimología , Retinopatía de la Prematuridad/prevención & control , Animales , Animales Recién Nacidos , Calbindina 2/metabolismo , Calbindinas/metabolismo , Electrorretinografía , Proteína Ácida Fibrilar de la Glía , Gliosis/enzimología , Inmunohistoquímica , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C-alfa/metabolismo , Retina/fisiopatología , Retinopatía de la Prematuridad/enzimología , Tubulina (Proteína)/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 303(3): H297-308, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661511

RESUMEN

Earlier studies have demonstrated that aldose reductase (AR) plays a key role in mediating ischemia-reperfusion (I/R) injury. Our objective was to investigate if AR mediates I/R injury by influencing phosphorylation of glycogen synthase kinase-3ß (p-GSK3ß). To investigate this issue, we used three separate models to study the effects of stress injury on the heart. Hearts isolated from wild-type (WT), human expressing AR transgenic (ARTg), and AR knockout (ARKO) mice were perfused with/without GSK3ß inhibitors (SB-216763 and LiCl) and subjected to I/R. Ad-human AR (Ad-hAR)-expressing HL-1 cardiac cells were exposed to hypoxia (0.5% O(2)) and reoxygenation (20.9% O(2)) conditions. I/R in a murine model of transient occlusion and reperfusion of the left anterior descending coronary artery (LAD) was used to study if p-GSK3ß was affected through increased AR flux. Lactate dehydrogenase (LDH) release and left ventricular developed pressure (LVDP) were measured. LVDP was decreased in hearts from ARTg mice compared with WT and ARKO after I/R, whereas LDH release and apoptotic markers were increased (P < 0.05). p-GSK3ß was decreased in ARTg hearts compared with WT and ARKO (P < 0.05). In ARKO, p-GSK3ß and apoptotic markers were decreased compared with WT (P < 0.05). WT and ARTg hearts perfused with GSK3ß inhibitors improved p-GSK3ß expression and LVDP and exhibited decreased LDH release, apoptosis, and mitochondrial pore opening (P < 0.05). Ad-hAR-expressing HL-1 cardiac cells, exposed to hypoxia (0.5% O(2)) and reoxygenation (20.9% O(2)), had greater LDH release compared with control HL-1 cells (P < 0.05). p-GSK3ß was decreased and correlated with increased apoptotic markers in Ad-hAR HL-1 cells (P < 0.05). Treatment with phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) inhibitor increased injury demonstrated by increased LDH release in ARTg, WT, and ARKO hearts and in Ad-hAR-expressing HL-1 cells. Cells treated with protein kinase C (PKC) α/ß inhibitor displayed significant increases in p-Akt and p-GSK3ß expression, and resulted in decreased LDH release. In summary, AR mediates changes in p-GSK3ß, in part, via PKCα/ß and Akt during I/R.


Asunto(s)
Aldehído Reductasa/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Animales , Apoptosis , Línea Celular , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Recuperación de la Función , Transfección , Función Ventricular Izquierda , Presión Ventricular
6.
Vis Neurosci ; 29(6): 267-74, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23101909

RESUMEN

Streptozotocin (STZ)-induced diabetes is associated with reductions in the electrical response of the outer retina and retinal pigment epithelium (RPE) to light. Aldose reductase (AR) is the first enzyme required in the polyol-mediated metabolism of glucose, and AR inhibitors have been shown to improve diabetes-induced electroretinogram (ERG) defects. Here, we used control and AR -/- mice to determine if genetic inactivation of this enzyme likewise inhibits retinal electrophysiological defects observed in a mouse model of type 1 diabetes. STZ was used to induce hyperglycemia and type 1 diabetes. Diabetic and age-matched nondiabetic controls of each genotype were maintained for 22 weeks, after which ERGs were used to measure the light-evoked components of the RPE (dc-ERG) and the neural retina (a-wave, b-wave). In comparison to their nondiabetic controls, wildtype (WT) and AR -/- diabetic mice displayed significant decreases in the c-wave, fast oscillation, and off response components of the dc-ERG but not in the light peak response. Nondiabetic AR -/- mice displayed larger ERG component amplitudes than did nondiabetic WT mice; however, the amplitude of dc-ERG components in diabetic AR -/- animals were similar to WT diabetics. ERG a-wave amplitudes were not reduced in either diabetic group, but b-wave amplitudes were lower in WT and AR -/-diabetic mice. These findings demonstrate that the light-induced responses of the RPE and outer retina are disrupted in diabetic mice, but these defects are not due to photoreceptor dysfunction, nor are they ameliorated by deletion of AR. This latter finding suggests that benefits observed in other studies utilizing pharmacological inhibitors of AR might have been secondary to off-target effects of the drugs.


Asunto(s)
Aldehído Reductasa , Diabetes Mellitus Experimental , Retinopatía Diabética , Epitelio Pigmentado de la Retina , Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/genética , Retinopatía Diabética/enzimología , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Modelos Animales de Enfermedad , Electrorretinografía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Epitelio Pigmentado de la Retina/enzimología , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/fisiopatología , Estreptozocina/efectos adversos
7.
Life Sci ; 304: 120694, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35679914

RESUMEN

AIMS: Acetaminophen (APAP) is a relatively safe analgesic drug, but overdosing can cause acute liver failure. Ingested APAP is detoxified by metabolic conversion through conjugation reactions with glucuronate, sulfate, or glutathione (GSH). The consumption of GSH through conjugation as well as mitochondrial dysfunction is considered to be responsible for the increased susceptibility to APAP-induced hepatotoxicity. Compared to wild-type (WT) mice, Akr1a-knockout (KO) mice are vulnerable to developing hepatotoxicity due to the fact that ascorbate synthesis is attenuated. We used such KO mice to investigate how these conjugation reactions are involved in the hepatotoxicity caused by an overdose of APAP under ascorbate-deficient conditions. MAIN METHODS: APAP (400 mg/kg) was intraperitoneally administered to WT mice and KO mice. In addition to histological and blood biochemical analyses, metabolites in the liver, blood plasma, and urine were measured at several time points by liquid chromatography-mass spectrometry. KEY FINDINGS: Liver damage occurred earlier in the KO mice than in the WT mice. The levels of APAP-Cys, a final metabolite of GSH-conjugated APAP, as well as glucuronidated APAP and sulfated APAP were all higher in the KO mice compared to the WT mice. Treatment of the APAP-administered KO mice with N-acetylcysteine or supplementation of ascorbate suppressed the conjugation reactions at 6 h after APAP had been administrated, which mitigated the degree of liver damage. SIGNIFICANCE: An ascorbate deficiency coordinately stimulates conjugation reactions of APAP, which, combined with the mitochondrial damage caused by APAP metabolites, collectively results in the aggravation of the acute liver failure.


Asunto(s)
Acetaminofén , Aldehído Reductasa , Enfermedad Hepática Inducida por Sustancias y Drogas , Acetaminofén/farmacocinética , Acetaminofén/toxicidad , Aldehído Reductasa/deficiencia , Aldehído Reductasa/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Glutatión/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Diabetologia ; 54(5): 1242-51, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21267539

RESUMEN

AIMS/HYPOTHESIS: The aim of the study was to investigate the effects of genetic deficiency of aldose reductase in mice on the development of key endpoints of diabetic nephropathy. METHODS: A line of Ar (also known as Akr1b3)-knockout (KO) mice, a line of Ar-bitransgenic mice and control C57BL/6 mice were used in the study. The KO and bitransgenic mice were deficient for Ar in the renal glomeruli and all other tissues, with the exception of, in the bitransgenic mice, a human AR cDNA knockin-transgene that directed collecting-tubule epithelial-cell-specific AR expression. Diabetes was induced in 8-week-old male mice with streptozotocin. Mice were further maintained for 17 weeks then killed. A number of serum and urinary variables were determined for these 25-week-old mice. Periodic acid-Schiff staining, western blots, immunohistochemistry and protein kinase C (PKC) activity assays were performed for histological analyses, and to determine the levels of collagen IV and TGF-ß1 and PKC activities in renal cortical tissues. RESULTS: Diabetes-induced extracellular matrix accumulation and collagen IV overproduction were completely prevented in diabetic Ar-KO and bitransgenic mice. Ar deficiency also completely or partially prevented diabetes-induced activation of renal cortical PKC, TGF-ß1 and glomerular hypertrophy. Loss of Ar results in a 43% reduction in urine albumin excretion in the diabetic Ar-KO mice and a 48% reduction in the diabetic bitransgenic mice (p < 0.01). CONCLUSIONS/INTERPRETATION: Genetic deficiency of Ar significantly ameliorated development of key endpoints linked with early diabetic nephropathy in vivo. Robust and specific inhibition of aldose reductase might be an effective strategy for the prevention and treatment of diabetic nephropathy.


Asunto(s)
Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Nefropatías Diabéticas/genética , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
9.
Respir Res ; 12: 145, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-22054012

RESUMEN

BACKGROUND: Childhood hospitalization related to asthma remains at historically high levels, and its incidence is on the rise world-wide. Previously, we have demonstrated that aldose reductase (AR), a regulatory enzyme of polyol pathway, is a major mediator of allergen-induced asthma pathogenesis in mouse models. Here, using AR null (AR-/-) mice we have investigated the effect of AR deficiency on the pathogenesis of ragweed pollen extract (RWE)-induced allergic asthma in mice and also examined the efficacy of enteral administration of highly specific AR inhibitor, fidarestat. METHODS: The wild type (WT) and AR-/- mice were sensitized and challenged with RWE to induce allergic asthma. AR inhibitor, fidarestat was administered orally. Airway hyper-responsiveness was measured in unrestrained animals using whole body plethysmography. Mucin levels and Th2 cytokine in broncho-alveolar lavage (BAL) were determined using mouse anti-Muc5A/C ELISA kit and multiplex cytokine array, respectively. Eosinophils infiltration and goblet cells were assessed by H&E and periodic acid Schiff (PAS)-staining of formalin-fixed, paraffin-embedded lung sections. T regulatory cells were assessed in spleen derived CD4+CD25+ T cells population. RESULTS: Deficiency of AR in mice led to significantly decreased PENH, a marker of airway hyper-responsiveness, metaplasia of airway epithelial cells and mucus hyper-secretion following RWE-challenge. This was accompanied by a dramatic decrease in infiltration of eosinophils into sub-epithelium of lung as well as in BAL and release of Th2 cytokines in response to RWE-challenge of AR-/- mice. Further, enteral administration of fidarestat significantly prevented eosinophils infiltration, airway hyper-responsiveness and also markedly increased population of T regulatory (CD4+CD25+FoxP3+) cells as compared to RWE-sensitized and challenged mice not treated with fidarestat. CONCLUSION: Our results using AR-/- mice strongly suggest the role of AR in allergic asthma pathogenesis and effectiveness of oral administration of AR inhibitor in RWE-induced asthma in mice supports the use of AR inhibitors in the treatment of allergic asthma.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Asma/enzimología , Asma/prevención & control , Imidazolidinas/administración & dosificación , Polen , Rinitis Alérgica Estacional/enzimología , Rinitis Alérgica Estacional/prevención & control , Aldehído Reductasa/deficiencia , Aldehído Reductasa/metabolismo , Ambrosia/química , Animales , Ratones , Ratones Noqueados , Extractos Vegetales , Resultado del Tratamiento
10.
J Neurosci Res ; 88(9): 2026-34, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20143423

RESUMEN

Aldose reductase (AR), the first enzyme in the polyol pathway, has been implicated in a wide variety of physiological and pathological functions, such as diabetic vascular and neural complications. It is known that diabetes mellitus can exacerbate brain and retina damage after ischemic injuries. However, the underlying mechanisms are not clear. In the present study, we made use of db/db mice with an AR null mutation (AR(-/-)db/db) to understand better the role of AR in the pathogenesis of brain and retina ischemic injuries under diabetic conditions. Cerebral and retinal ischemia was induced by transient middle cerebral artery occlusion in control and diabetic mice either with or without an AR null mutation. Mice were evaluated for neurological deficits after 30 min of ischemia and 23.5 hr of reperfusion. Our results showed that the diabetic db/db mice had significantly more severe neurological deficit and larger brain infarct size than the nondiabetic mice. Compared with wild-type db/db mice, the AR(-/-)db/db mice had significantly lower neurological scores, smaller brain infarct areas, and less hemispheric brain swelling. Retinal swelling was also significantly decreased in the AR(-/-)db/db mice. Less swelling in the brain and retina of the AR(-/-)db/db mice correlated with less expression of the water channel aquaporin 4. Taken together, these data clearly show that deletion of AR leads to less severe brain and retinal ischemic injuries in the diabetic db/db mouse. The present study indicates that inhibition of AR in diabetics may protect against damage in the brain and retina following ischemic reperfusion injury.


Asunto(s)
Aldehído Reductasa/metabolismo , Isquemia Encefálica/enzimología , Diabetes Mellitus Tipo 2/complicaciones , Enfermedades de la Retina/enzimología , Accidente Cerebrovascular/enzimología , Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Animales , Acuaporina 4/metabolismo , Glucemia/metabolismo , Encéfalo/enzimología , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Estrés Oxidativo/fisiología , Retina/enzimología , Retina/metabolismo , Retina/patología , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Índice de Severidad de la Enfermedad , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
11.
Carcinogenesis ; 30(5): 799-807, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19028703

RESUMEN

Aldose reductase (AR; EC 1.1.1.21), an nicotinamide adenine dinucleotide phosphate-dependent aldo-keto reductase, has been shown to be involved in oxidative stress signaling initiated by inflammatory cytokines, chemokines and growth factors. Recently, we have shown that inhibition of this enzyme prevents the growth of colon cancer cells in vitro as well as in nude mice xenografts. Herein, we investigated the mediation of AR in the formation of colonic preneoplastic aberrant crypt foci (ACF) using azoxymethane (AOM)-induced colon cancer mice model. Male BALB/c mice were administrated with AOM without or with AR inhibitor, sorbinil and at the end of the protocol, all the mice were euthanized and colons were evaluated for ACF formation. Administration of sorbinil significantly lowered the number of AOM-induced ACF. Similarly, AR-null mice administered with AOM demonstrated significant resistance to ACF formation. Furthermore, inhibition of AR or knockout of AR gene in the mice significantly prevented AOM-induced expression of inducible nitric oxide synthase and cyclooxygenase-2 proteins as well as their messenger RNA. AR inhibition or knockdown also significantly decreased the phosphorylation of protein kinase C (PKC) beta2 and nuclear factor kappa binding protein as well as expression of preneoplastic marker proteins such as cyclin D1 and beta-catenin in mice colons. Our results suggest that AR mediates the formation of ACF in AOM-treated mice and thereby inhibition of AR could provide an effective chemopreventive approach for the treatment of colon cancer.


Asunto(s)
Aldehído Reductasa/deficiencia , Azoximetano/toxicidad , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/prevención & control , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/prevención & control , Aldehído Reductasa/metabolismo , Animales , Neoplasias del Colon/patología , Cartilla de ADN , Peroxidación de Lípido , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/fisiología , Lesiones Precancerosas/patología , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Exp Eye Res ; 87(5): 454-61, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18760274

RESUMEN

Diabetes is a major contributing factor in cataract development. In animal models where cataracts develop within days or weeks of diabetes it is well established that osmotic stress from the accumulation of sorbitol leads to cataract development. This mechanism might explain the rare cases of acute cataract sometimes found in patients with uncontrolled sustained hyperglycemia but cannot account for the vast majority of cataracts that developed after years of diabetes. Thus, a model that can simulate diabetic slow-developing cataract is needed. The contribution of osmotic and oxidative stress in cataract development in sorbitol dehydrogenase (SDH) deficient mice, a model for slow-developing cataract in diabetic patients was determined. Contribution of osmotic stress was assessed by HPLC measurement of sorbitol and by observing the effect of blocking sorbitol accumulation by aldose reductase (AR) null mutation in the SDH deficient mice. Contribution of oxidative stress was assessed by observing the effect of vitamin E treatment and the effect of null mutation of glutathione peroxidase-1 (Gpx-1) on cataract development in these mice. Lenticular sorbitol level was significantly increased in the SDH deficient mice, and blocking sorbitol accumulation by the AR null mutation prevented cataract development, demonstrating the contribution of osmotic stress in cataract development. SDH deficiency did not affect lens oxidative stress status. However, treatment with vitamin E significantly reduced the incidence of cataract, and Gpx-1 deficiency exacerbated cataract development in these mice. Our findings suggest that chronic oxidative stress impaired the osmoregulatory mechanism of the lens. This was not evident until modest increases in lens sorbitol increased the demand of its osmoregulatory function. This osmoregulatory dysfunction model is supported by the fact that the activity of Na+/K+-ATPase, the key regulator of cellular ions and water balance, was dramatically reduced in the precataractous lenses of the SDH deficient mice, and that treatment with vitamin E prevented the loss of Na+/K+-ATPase activity. This osmoregulatory dysfunction model might explain why diabetic patients who control their blood glucose moderately well are still susceptible to develop cataract.


Asunto(s)
Catarata/fisiopatología , Diabetes Mellitus Experimental/complicaciones , Ósmosis/fisiología , Estrés Oxidativo/fisiología , Envejecimiento/metabolismo , Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Aldehído Reductasa/fisiología , Animales , Antioxidantes/metabolismo , Catarata/etiología , Catarata/metabolismo , Catarata/prevención & control , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glutatión/metabolismo , Glutatión Peroxidasa/deficiencia , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/fisiología , L-Iditol 2-Deshidrogenasa/deficiencia , Cristalino/metabolismo , Ratones , Ratones Noqueados , Mutación , ARN Mensajero/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Sorbitol/metabolismo , Vitamina E/uso terapéutico , Glutatión Peroxidasa GPX1
13.
Bioengineered ; 9(1): 233-241, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29865993

RESUMEN

Ethylene glycol (EG) is an important chemical used as antifreeze and a raw material in polyester synthesis. The EG biosynthetic pathway from D-xylose with D-xylonate as key intermediate has some advantages, but showed low EG production. Here, we reconstructed and optimized this pathway in Escherichia coli. In view of the greater intracellular prevalence of NADH, an aldehyde reductase FucO using NADH was employed to convert glycoaldehyde into EG, in replacement of NADPH-dependent reductase YqhD. To suppress the accumulation of by-products acetate and glycolate, two genes arcA and aldA were knocked out. The resultant strain Q2843 produced 72 g/L EG under fed-batch fermentation conditions, with the yield of 0.40 g/g D-xylose and EG productivity of 1.38 g/L/h. The use of NADH-dependent enzyme FucO and by-product elimination significantly improved the performance of EG producing strain, which represented the highest titer, yield and productivity of EG reported so far.


Asunto(s)
Aldehído Reductasa/metabolismo , Escherichia coli/metabolismo , Glicol de Etileno/metabolismo , Ingeniería Metabólica/métodos , Xilosa/metabolismo , Ácido Acético/metabolismo , Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Proteínas de la Membrana Bacteriana Externa/genética , Técnicas de Cultivo Celular por Lotes , Vías Biosintéticas/genética , Clonación Molecular , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Fermentación , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glicolatos/metabolismo , Cinética , NAD/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética
14.
Diabetes ; 55(7): 1946-53, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16804062

RESUMEN

The exaggerated flux through polyol pathway during diabetes is thought to be a major cause of lesions in the peripheral nerves. Here, we used aldose reductase (AR)-deficient (AR(-/-)) and AR inhibitor (ARI)-treated mice to further understand the in vivo role of polyol pathway in the pathogenesis of diabetic neuropathy. Under normal conditions, there were no obvious differences in the innervation patterns between wild-type AR (AR(+/+)) and AR(-/-) mice. Under short-term diabetic conditions, AR(-/-) mice were protected from the reduction of motor and sensory nerve conduction velocities observed in diabetic AR(+/+) mice. Sorbitol levels in the sciatic nerves of diabetic AR(+/+) mice were increased significantly, whereas sorbitol levels in the diabetic AR(-/-) mice were significantly lower than those in diabetic AR(+/+) mice. In addition, signs of oxidative stress, such as increased activation of c-Jun NH(2)-terminal kinase (JNK), depletion of reduced glutathione, increase of superoxide formation, and DNA damage, observed in the sciatic nerves of diabetic AR(+/+) mice were not observed in the diabetic AR(-/-) mice, indicating that the diabetic AR(-/-) mice were protected from oxidative stress in the sciatic nerve. The diabetic AR(-/-) mice also excreted less 8-hydroxy-2'-deoxyguanosine in urine than diabetic AR(+/+) mice. The structural abnormalities observed in the sural nerve of diabetic AR(+/+) mice were less severe in the diabetic AR(-/-) mice, although it was only mildly protected by AR deficiency under short-term diabetic conditions. Signs of oxidative stress and functional and structural abnormalities were also inhibited by the ARI fidarestat in diabetic AR(+/+) nerves, similar to those in diabetic AR(-/-) mice. Taken together, increased polyol pathway flux through AR is a major contributing factor in the early signs of diabetic neuropathy, possibly through depletion of glutathione, increased superoxide accumulation, increased JNK activation, and DNA damage.


Asunto(s)
Aldehído Reductasa/deficiencia , Daño del ADN , Diabetes Mellitus Experimental/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas Motoras/fisiología , Conducción Nerviosa/fisiología , Aldehído Reductasa/genética , Animales , Diabetes Mellitus Experimental/genética , Activación Enzimática , Fructosa/metabolismo , Genes Reporteros , Glucosa/metabolismo , Glutatión/metabolismo , Inositol/metabolismo , Ratones , Ratones Noqueados , Poli Adenosina Difosfato Ribosa/metabolismo , Valores de Referencia , Sorbitol/metabolismo , Superóxidos/metabolismo , Nervio Sural/fisiopatología
15.
Mol Cell Biol ; 20(16): 5840-6, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10913167

RESUMEN

Aldose reductase (ALR2) is thought to be involved in the pathogenesis of various diseases associated with diabetes mellitus, such as cataract, retinopathy, neuropathy, and nephropathy. However, its physiological functions are not well understood. We developed mice deficient in this enzyme and found that they had no apparent developmental or reproductive abnormality except that they drank and urinated significantly more than their wild-type littermates. These ALR2-deficient mice exhibited a partially defective urine-concentrating ability, having a phenotype resembling that of nephrogenic diabetes insipidus.


Asunto(s)
Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Diabetes Insípida Nefrogénica/genética , Ratones Noqueados , Animales , Diabetes Insípida Nefrogénica/etiología , Diabetes Insípida Nefrogénica/metabolismo , Modelos Animales de Enfermedad , Ratones
16.
Neurobiol Aging ; 50: 119-133, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27960106

RESUMEN

Fungicide exposure causes degeneration of dopaminergic neurons and contributes to Parkinson's disease (PD). Benomyl inhibits enzymes responsible for detoxifying the reactive dopamine metabolite 3,4-dihydroxyphenylacetaldehyde. Aldose reductase (AR) is known as tetrahydrobiopterin (BH4) reductase that generates BH4, a cofactor for tyrosine hydroxylase (TH) involved in dopamine synthesis. AR also acts as an aldehyde reductase involved in detoxifying 3,4-dihydroxyphenylacetaldehyde. In PD patients, the level of AR is significantly lower in the cerebellum. To determine if AR deficiency contributes to PD, AR wild-type (AR+/+) and knockout (AR-/-) mice were administrated with 1-methyl-4-phenyl -1,2,3,6- tetrahydropyridine (MPTP). The MPTP-treated AR-/- mice showed more severe behavioral deficits and brain damage than that of AR+/+ mice. Contrary to expectation, under normal or MPTP-treated condition, AR-/- mice showed a significant elevation of BH4 and dopamine in the midbrain, suggesting that either AR does not contribute to BH4 production, or other BH4 synthetic pathways are induced. The AR-/- brain showed upregulation of peroxynitrite, inducible nitric oxide synthase and downregulation of antioxidant enzymes, Cu/Zn superoxide dismutase (SOD) and peroxiredoxin 2 (Prx2), which indicate an increase in oxidative stress. In line with the animal data, pretreating the SH-SY5Y cells with AR inhibitors (Fidarestat or Epalrestat) before MPP+ treatment, increased severe cell death and mitochondrial fragmentation with downregulation of SOD were observed when compared to the MPP+ treatment alone. Cycloxygenase 2 (COX2), which can lead to the oxidation of dopamine, was upregulated in AR-/- brains. Autophagic proteins, beclin-1 and LC3B were also downregulated. The loss of dopaminergic neurons was associated with activation of p-ERK1/2. These findings suggest that AR plays an important role in protecting dopaminergic neuron against neurotoxic metabolites in PD.


Asunto(s)
Aldehído Reductasa/deficiencia , Autofagia , Neuronas Dopaminérgicas/patología , Estrés Oxidativo/fisiología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad de Parkinson/fisiopatología
17.
Oncotarget ; 8(5): 7357-7369, 2017 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-28060768

RESUMEN

The AKR1A1 protein is a member of the aldo-keto reductase superfamily that is responsible for the conversion of D-glucuronate to L-gulonate in the ascorbic acid (vitamin C) synthesis pathway. In a pCAG-eGFP transgenic mouse line that was produced by pronuclear microinjection, the integration of the transgene resulted in a 30-kb genomic DNA deletion, including the Akr1A1 gene, and thus caused the knockout (KO) of the Akr1A1 gene and targeting of the eGFP gene. The Akr1A1 KO mice (Akr1A1eGFP/eGFP) exhibited insufficient serum ascorbic acid levels, abnormal bone development and osteoporosis. Using micro-CT analysis, the results showed that the microarchitecture of the 12-week-old Akr1A1eGFP/eGFP mouse femur was shorter in length and exhibited less cortical bone thickness, enlargement of the bone marrow cavity and a complete loss of the trabecular bone in the distal femur. The femoral head and neck of the proximal femur also showed a severe loss of bone mass. Based on the decreased levels of serum osteocalcin and osteoblast activity in the Akr1A1eGFP/eGFP mice, the osteoporosis might be caused by impaired bone formation. In addition, administration of ascorbic acid to the Akr1A1eGFP/eGFP mice significantly prevented the condition of osteoporotic femurs and increased bone formation. Therefore, through ascorbic acid administration, the Akr1A1 KO mice exhibited controllable osteoporosis and may serve as a novel model for osteoporotic research.


Asunto(s)
Aldehído Reductasa/genética , Deficiencia de Ácido Ascórbico/genética , Fémur/patología , Técnicas de Inactivación de Genes , Osteogénesis , Osteoporosis/genética , Aldehído Reductasa/deficiencia , Animales , Ácido Ascórbico/administración & dosificación , Ácido Ascórbico/sangre , Deficiencia de Ácido Ascórbico/enzimología , Deficiencia de Ácido Ascórbico/patología , Deficiencia de Ácido Ascórbico/prevención & control , Modelos Animales de Enfermedad , Femenino , Fémur/diagnóstico por imagen , Fémur/enzimología , Predisposición Genética a la Enfermedad , Ratones Noqueados , Osteoblastos/enzimología , Osteoblastos/patología , Osteocalcina/sangre , Osteoporosis/enzimología , Osteoporosis/patología , Osteoporosis/prevención & control , Fenotipo , Factores de Tiempo , Microtomografía por Rayos X
18.
J Biosci Bioeng ; 123(3): 333-341, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27865643

RESUMEN

4-Hydroxy-2(or 5)-ethyl-5(or 2)-methyl-3(2H)-furanone (HEMF) is considered a key flavor compound in soy sauce. The compound has a caramel-like aroma and several important physiological activities, such as strong antioxidant activity. Here, we report the identification and characterization of an enzyme involved in the biosynthesis of HEMF in yeast. We fractionated yeast cell-free extract from Saccharomyces cerevisiae using column chromatography and partially purified a fraction with HEMF-forming activity. Peptide mass fingerprinting analysis showed that the partially purified fraction contains aldehyde reductase encoded by YNL134C. This reductase shares low sequence identity with enone oxidoreductase, which is responsible for the formation of 4-hydroxy-2,5-dimethyl-3(2H)-furanone (HDMF) and HEMF in plants. YNL134C was expressed heterologously in Escherichia coli, and the purified protein catalyzed the formation of HEMF from the mixture of Maillard reaction products, acetaldehydes, and NADPH. Multicopy expression in S. cerevisiae resulted in increased HEMF productivity, and gene knockout of YNL134C in S. cerevisiae resulted in decreased HEMF productivity. These data suggest that the translation product of YNL134C is the HEMF-producing enzyme in yeast. Detailed analyses of an intermediate in the enzymatic reaction mixture revealed that HEMF is synthesized from (2E)-2-ethylidene-4-hydroxy-5-methyl-3(2H)-furanone, which formed via Knoevenagel condensation between the acetaldehyde and 4-hydroxy-5-methyl-3(2H)-furanone derived from the Maillard reaction based on ribose and glycine, by YNL134Cp in an NADPH dependent manner. Overall, this study shed light on the molecular basis for the improvement of soy sauce flavor and the biotechnological production of HEMF.


Asunto(s)
Furanos/metabolismo , Oxidorreductasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/metabolismo , Acetaldehído/metabolismo , Aldehído Reductasa/deficiencia , Aldehído Reductasa/genética , Aldehído Reductasa/aislamiento & purificación , Aldehído Reductasa/metabolismo , Extractos Celulares , Escherichia coli/genética , Aromatizantes/química , Glicina/metabolismo , NADP/metabolismo , Oxidorreductasas/deficiencia , Oxidorreductasas/genética , Oxidorreductasas/aislamiento & purificación , Ribosa/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Alimentos de Soja
19.
Diabetes ; 54(11): 3119-25, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16249434

RESUMEN

In 15-month-old db/db mice, signs of diabetic retinopathy, including blood-retinal barrier breakdown, loss of pericytes, neuro-retinal apoptosis, glial reactivation, and proliferation of blood vessels, were evident. These changes in the diabetic retina were associated with increased expression of aldose reductase (AR). To further understand the role of AR in the pathogenesis of diabetic retinopathy, we generated db/db mice with an AR null mutation (AR-/- db/db). AR deficiency led to fewer retinal blood vessels with IgG leakage, suggesting that AR may contribute to blood-retinal barrier breakdown. AR deficiency also prevented diabetes-induced reduction of platelet/endothelial cell adhesion molecule-1 expression and increased expression of vascular endothelial growth factor, which may have contributed to blood-retinal barrier breakdown. In addition, long-term diabetes-induced neuro-retinal stress and apoptosis and proliferation of blood vessels were less prominent in AR-/- db/db mice. These findings indicate that AR is responsible for the early events in the pathogenesis of diabetic retinopathy, leading to a cascade of retinal lesions, including blood-retinal barrier breakdown, loss of pericytes, neuro-retinal apoptosis, glial reactivation, and neovascularization.


Asunto(s)
Aldehído Reductasa/deficiencia , Apoptosis/fisiología , Barrera Hematorretinal/fisiopatología , Diabetes Mellitus/fisiopatología , Retina/citología , Retina/enzimología , Aldehído Reductasa/genética , Animales , Proliferación Celular , Diabetes Mellitus/genética , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Ratones , Neuroglía , Estrés Oxidativo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Factor A de Crecimiento Endotelial Vascular/genética
20.
Mol Neurobiol ; 53(1): 662-676, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25520004

RESUMEN

Inflammatory reactions are the most critical pathological processes occurring after spinal cord injury (SCI). Activated microglia/macrophages have either detrimental or beneficial effects on neural regeneration based on their functional polarized M1/M2 subsets. However, the mechanism of microglia/macrophage polarization to M1/M2 at the injured spinal cord environment remains unknown. In this study, wild-type (WT) or aldose reductase (AR)-knockout (KO) mice were subjected to SCI by a spinal crush injury model. The expression pattern of AR, behavior tests for locomotor activity, and lesion size were assessed at between 4 h and 28 days after SCI. We found that the expression of AR is upregulated in microglia/macrophages after SCI in WT mice. In AR KO mice, SCI led to smaller injury lesion areas compared to WT. AR deficiency-induced microglia/macrophages induce the M2 rather than the M1 response and promote locomotion recovery after SCI in mice. In the in vitro experiments, microglia cell lines (N9 or BV2) were treated with the AR inhibitor (ARI) fidarestat. AR inhibition caused 4-hydroxynonenal (HNE) accumulation, which induced the phosphorylation of the cAMP response element-binding protein (CREB) to promote Arg1 expression. KG501, the specific inhibitor of phosphorylated CREB, could cancel the upregulation of Arg1 by ARI or HNE stimulation. Our results suggest that AR works as a switch which can regulate microglia by polarizing cells to either the M1 or the M2 phenotype under M1 stimulation based on its states of activity. We suggest that inhibiting AR may be a promising therapeutic method for SCI in the future.


Asunto(s)
Aldehído Reductasa/biosíntesis , Polaridad Celular/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Aldehído Reductasa/deficiencia , Animales , Línea Celular , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda