Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(15): 3918-3923, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29572430

RESUMO

TP53 (p53) is a tumor suppressor whose functions are lost or altered in most malignancies. p53 homozygous knockout (p53-/-) mice uniformly die of spontaneous malignancy, typically T-cell lymphoma. RALBP1 (RLIP76, Rlip) is a stress-protective, mercapturic acid pathway transporter protein that also functions as a Ral effector involved in clathrin-dependent endocytosis. In stark contrast to p53-/- mice, Rlip-/- mice are highly resistant to carcinogenesis. We report here that partial Rlip deficiency induced by weekly administration of an Rlip-specific phosphorothioate antisense oligonucleotide, R508, strongly inhibited spontaneous as well as benzo(a)pyrene-induced carcinogenesis in p53-/- mice. This treatment effectively prevented large-scale methylomic and transcriptomic abnormalities suggestive of inflammation found in cancer-bearing p53-/- mice. The remarkable efficiency with which Rlip deficiency suppresses spontaneous malignancy in p53-/- mice has not been observed with any previously reported pharmacologic or genetic intervention. These findings are supported by cross-breeding experiments demonstrating that hemizygous Rlip deficiency also reduces the spontaneous malignancy phenotype of p53+/- mice. Rlip is found on the cell surface, and antibodies directed against Rlip were found to inhibit growth and promote apoptosis of cell lines as effectively as Rlip siRNA. The work presented here investigates several features, including oxidative DNA damage of the Rlip-p53 association in malignant transformation, and offers a paradigm for the mechanisms of tumor suppression by p53 and the prospects of suppressing spontaneous malignancy in hereditary cancer syndromes such as Li-Fraumeni.


Assuntos
Proteínas Ativadoras de GTPase/deficiência , Neoplasias/genética , Neoplasias/prevenção & controle , Proteína Supressora de Tumor p53/genética , Animais , Apoptose , Feminino , Proteínas Ativadoras de GTPase/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/metabolismo
4.
J Biol Chem ; 286(27): 24089-100, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21576240

RESUMO

The development of intratumoral hypoxia, a hallmark of rapidly progressing solid tumors, renders tumor cells resistant to chemotherapy and radiation therapy. We have recently shown that inhibition of aldose reductase (AR), an enzyme that catalyzes the reduction of lipid aldehydes and their glutathione conjugates, prevents human colon cancer cell growth in culture as well as in nude mouse xenografts by inhibiting the NF-κB-dependent activation of oxidative stress-mediated inflammatory and carcinogenic markers. However, the role of AR in mediating hypoxic stress signals is not known. We therefore investigated the molecular mechanisms by which AR inhibition prevents the hypoxia-induced human colon cancer cells growth and invasion. Our results indicate that AR inhibition by the pharmacological inhibitor fidarestat or ablation by AR-specific siRNA prevents hypoxia-induced proliferation of HT29, SW480, and Caco-2 colon cancer cells. Furthermore, hypoxia-induced increase in the level of HIF-1α in colon cancer cells was significantly decreased by AR inhibition. During hypoxic conditions, treatment of HT29 cells with the AR inhibitor fidarestat significantly decreased the expression of vascular endothelial growth factor, a down target of HIF-1α, at both mRNA and protein levels and also prevented the activation of PI3K/AKT, GSK3ß, Snail, and lysyl oxidase. Furthermore, inhibition of hypoxia-induced HIF-1α protein accumulation by AR inhibition was abolished in the presence of MG132, a potent inhibitor of the 26 S proteasome. In addition, AR inhibition also prevented the hypoxia-induced inflammatory molecules such as Cox-2 and PGE2 and expression of extracellular matrix proteins such as MMP2, vimentin, uPAR, and lysyl oxidase 2. In conclusion, our results indicate that AR mediates hypoxic signals, leading to tumor progression and invasion.


Assuntos
Aldeído Redutase/metabolismo , Neoplasias do Colo/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/genética , Células CACO-2 , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Dinoprostona/genética , Dinoprostona/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imidazolidinas/farmacologia , Leupeptinas/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma , Proteína-Lisina 6-Oxidase/genética , Proteína-Lisina 6-Oxidase/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Fator A de Crescimento do Endotélio Vascular/genética , Vimentina/genética , Vimentina/metabolismo
5.
Carcinogenesis ; 32(8): 1259-67, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21642355

RESUMO

Colon cancer is the third most common cause of cancer and is the second leading cause of cancer deaths in the USA. Although inhibition of aldose reductase (AR) is known to prevent human colon cancer cell growth in nude mice xenografts, the role of AR in the regulation of cancer metastasis is not known. We now demonstrate the mechanisms by which AR regulates colon cancer metastasis in vitro and in vivo. Inhibition of AR prevented the epidermal growth factor (EGF) or fibroblast growth factor (FGF)-induced migration and invasion of human colon cancer (HT29; KM20) cells by >70% and also inhibited (>80%) the adhesion of the cancer cells to endothelial cells. Treatment of endothelial cells with AR inhibitors significantly (∼85%) downregulated the EGF or FGF-induced expression of Inter-Cellular Adhesion Molecule-1, Vascular cell adhesion molecule-1 and vascular endothelial-cadherin. Furthermore, liver metastasis of green fluorescent protein-labeled KM20 cells injected into the spleen of athymic nude mice was significantly (>65%) prevented by AR inhibitor, fidarestat or ARsiRNA delivered systemically into the mice. Similar results were observed with HT29 cells. AR inhibition or ablation also prevented (70-90%) the increase in the levels of matrix metalloproteinase-2, cyclin D1, CD31, CD34 and the activation of nuclear factor-kappa-binding protein in metastatic liver. Thus, our results indicate that AR regulates cancer cell adhesion, invasion and migration events which initiate metastasis and therefore, AR inhibition could be a novel therapeutic approach for the prevention of colon cancer metastasis.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Movimento Celular/efeitos dos fármacos , Neoplasias do Colo/enzimologia , Neoplasias do Colo/prevenção & controle , Imidazolidinas/uso terapêutico , Neoplasias Hepáticas Experimentais/enzimologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Aldeído Redutase/genética , Aldeído Redutase/metabolismo , Animais , Western Blotting , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/patologia , Humanos , Técnicas Imunoenzimáticas , Neoplasias Hepáticas Experimentais/secundário , Masculino , Camundongos , Camundongos Nus , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Angiogenesis ; 14(2): 209-21, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21409599

RESUMO

We have recently shown that aldose reductase (AR, EC 1.1.1.21) a nicotinamide adenine dinucleotide phosphate-dependent aldo-keto reductase, known to be involved in oxidative stress-signaling, prevents human colon cancer cell growth in culture as well as in nude mice xenografts. Inhibition of AR also prevents azoxymethane-induced aberrant crypt foci formation in mice. In order to understand the chemopreventive mechanism(s) of AR inhibition in colon cancer, we have investigated the role of AR in the mediation of angiogenic signals in vitro and in vivo models. Our results show that inhibition of AR significantly prevented the VEGF- and FGF -induced proliferation and expression of proliferative marker Ki67 in the human umbilical vein endothelial cells (HUVEC). Further, AR inhibition or ablation with siRNA prevented the VEGF- and FGF -induced invasion and migration in HUVEC. AR inhibition also prevented the VEGF- and FGF- induced secretion/expression of IL-6, MMP2, MMP9, ICAM, and VCAM. The anti-angiogenic feature of AR inhibition in HUVEC was associated with inactivation of PI3 K/AKT and NF-κB (p65) and suppression of VEGF receptor 2 protein levels. Most importantly, matrigel plug model of angiogenesis in rats showed that inhibition of AR prevented infiltration of blood cells, invasion, migration and formation of capillary like structures, and expression of blood vessels markers CD31 and vWF. Thus, our results demonstrate that AR inhibitors could be novel agents to prevent angiogenesis.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Neovascularização Patológica/enzimologia , Aldeído Redutase/metabolismo , Indutores da Angiogênese/farmacologia , Animais , Capilares/efeitos dos fármacos , Capilares/crescimento & desenvolvimento , Moléculas de Adesão Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno , Combinação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Imidazolidinas/farmacologia , Interleucina-6/metabolismo , Laminina , Metaloproteinases da Matriz/metabolismo , Camundongos , NF-kappa B/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteoglicanas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Veias Umbilicais/citologia
7.
Respir Res ; 12: 145, 2011 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-22054012

RESUMO

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.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Asma/enzimologia , Asma/prevenção & controle , Imidazolidinas/administração & dosagem , Pólen , Rinite Alérgica Sazonal/enzimologia , Rinite Alérgica Sazonal/prevenção & controle , Aldeído Redutase/deficiência , Aldeído Redutase/metabolismo , Ambrosia/química , Animais , Camundongos , Camundongos Knockout , Extratos Vegetais , Resultado do Tratamento
8.
J Immunol ; 183(7): 4723-32, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19752229

RESUMO

Airway inflammation induced by reactive oxygen species-mediated activation of redox-sensitive transcription factors is the hallmark of asthma, a prevalent chronic respiratory disease. In various cellular and animal models, we have recently demonstrated that, in response to multiple stimuli, aldose reductase (AR) regulates the inflammatory signals mediated by NF-kappaB. Because NF-kappaB-mediated inflammation is a major characteristic of asthma pathogenesis, we have investigated the effect of AR inhibition on NF-kappaB and various inflammatory markers in cellular and animal models of asthma using primary human small airway epithelial cells and OVA-sensitized/challenged C57BL/6 mice, respectively. We observed that pharmacological inhibition or genetic ablation of AR by small interfering RNA prevented TNF-alpha- as well as LPS-induced apoptosis; reactive oxygen species generation; synthesis of inflammatory markers IL-6, IL-8, and PGE(2); and activation of NF-kappaB and AP-1 in small airway epithelial cells. In OVA-challenged mice, we observed that administration of an AR inhibitor markedly reduced airway hyperresponsiveness, IgE levels, eisonophils infiltration, and release of Th2 type cytokines in the airway. Our results indicate that AR inhibitors may offer a novel therapeutic approach to treat inflammatory airway diseases such as asthma.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Asma/imunologia , Asma/prevenção & controle , Citocinas/antagonistas & inibidores , Mediadores da Inflamação/antagonistas & inibidores , Ovalbumina/toxicidade , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Células Th2/imunologia , Aldeído Redutase/fisiologia , Animais , Asma/enzimologia , Asma/patologia , Brônquios/enzimologia , Brônquios/imunologia , Brônquios/metabolismo , Brônquios/patologia , Células Cultivadas , Galinhas , Citocinas/biossíntese , Citocinas/genética , Citotoxicidade Imunológica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/farmacologia , Mucosa Respiratória/enzimologia , Mucosa Respiratória/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Th2/enzimologia , Células Th2/metabolismo
9.
Am J Physiol Renal Physiol ; 298(6): F1393-404, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20335317

RESUMO

Renal-specific oxido-reductase/myoinositol oxygenase (RSOR/MIOX) is expressed in renal tubules. It catabolizes myo-inositol and its expression is increased in diabetic mice and in LLC-PK(1) cells under high-glucose ambience. Aldose reductase (AR) is another aldo-keto reductase that is expressed in renal tubules. It regulates the polyol pathway and plays an important role in glucose metabolism, osmolyte regulation, and ECM pathobiology via the generation of advanced glycation end products, reactive oxygen species, and activation of transforming growth factor (TGF)-beta. In view of the similarities between AR and RSOR/MIOX, the pathobiology of RSOR/MIOX and some of the cellular pathways affected by its overexpression were investigated. An increased expression of fibronectin was noted by transfection of LLC-PK(1) cells with pcDNA3.1-RSOR/MIOX. Similar changes were observed in LLC-PK(1) cells under high-glucose ambience, and they were notably lessened by RSOR/MIOX-small interfering (si) RNA treatment. The changes in tubulointerstitial fibronectin expression were also observed in the kidneys of db/db mice having high levels of RSOR. The pcDNA3.1-RSOR/MIOX transfectants had an increased NADH/NAD(+) ratio, PKC and TGF-beta activity, Raf1:Ras association, and p-ERK phosphorylation. These changes were significantly reduced by the inhibitors of PKC, aldose reductase, Ras farnesylation, and MEK1. Similar increases in various the above-noted parameters were observed under high-glucose ambience. Such changes were partially reversed with RSOR-siRNA treatment. Expression of E-cadherin and vimentin paralleled in cells overexpressing RSOR/MIOX or subjected to high-glucose ambience. These studies suggest that RSOR/MIOX modulates various downstream pathways affected by high-glucose ambience, and conceivably it plays a role in the pathobiology of tubulointerstitium in diabetic nephropathy.


Assuntos
Aldeído Redutase/metabolismo , Nefropatias Diabéticas/enzimologia , Inositol Oxigenase/metabolismo , Túbulos Renais/enzimologia , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/genética , Animais , Caderinas/metabolismo , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibronectinas/metabolismo , Fibrose , Glucose/metabolismo , Inositol Oxigenase/antagonistas & inibidores , Inositol Oxigenase/genética , Túbulos Renais/patologia , Células LLC-PK1 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NAD/metabolismo , Fosforilação , Proteína Quinase C-alfa/metabolismo , Prenilação de Proteína , Transporte Proteico , Interferência de RNA , Transdução de Sinais , Suínos , Transfecção , Fator de Crescimento Transformador beta1/metabolismo , Vimentina/metabolismo , Quinases raf/metabolismo , Proteínas ras/metabolismo
10.
Exp Eye Res ; 90(5): 555-63, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20138035

RESUMO

Chronic inflammatory diseases such as autoimmune and bacterial infections are associated with an elevated risk of ocular inflammation. Ciliary epithelial cells that play an important role in maintaining aqueous humor dynamics and homeostasis of anterior segment of eye are continuously exposed to inflammatory markers during infections and injury. Lipopolysacchharide (LPS), a Gram-negative bacterial endotoxin, dysregulates aqueous humor (AqH) homeostasis by inducing inflammatory changes. We have investigated how inhibition of a polyol pathway enzyme, aldose reductase (AR), alters LPS-induced inflammatory changes in human non-pigmented ciliary epithelial cells (hNPECs). The stimulation of hNPECs with LPS (1 microg/ml) caused increased secretion of inflammatory markers such as PGE(2) and NO in the culture medium as well as increased expression of COX-2 and iNOS proteins in cell extracts. LPS also increased phosphorylation of MAPKs (ERK1/2) and SAPK/JNK and activation of redox-sensitive transcription factors NF-kappaB and AP-1 in hNPECs and inhibition of AR by zopolrestat and sorbinil ameliorated these changes. Further, LPS-induced decrease in the expression of Na/K-ATPase in hNPECs was restored by AR inhibitors. Similar results were observed in ciliary bodies of LPS-injected rats. Taken together, our results suggest that AR plays an important role in the LPS-induced inflammatory changes in hNPECs and that inhibition of AR could be a novel therapeutic approach for ocular inflammation.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Corpo Ciliar/citologia , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Transdução de Sinais , Aldeído Redutase/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Benzotiazóis/farmacologia , Western Blotting , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Células Epiteliais/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Imidazolidinas/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo , Fosforilação , Ftalazinas/farmacologia , Ratos , Ratos Endogâmicos Lew , Fator de Transcrição AP-1/metabolismo , Transfecção
11.
Arch Environ Contam Toxicol ; 58(1): 123-30, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19557474

RESUMO

Malathion [S-(1,2-dicarboethoxyethyl) O, O-dimethyl phosphorodithioate] is a widely used organophosphorus insecticide throughout the world. However, limited efforts have made to study its genotoxic effect in different fish tissues. The present investigation was aimed to assess the genotoxic potential of the pesticide to the freshwater teleost fish Channa punctatus at sublethal concentrations using the micronucleus test and comet assay. Initially, the 96-h LC50 value of commercial-grade malathion (50% EC) was determined as 5.93 ppm in a semistatic system. Based on LC50, three test concentrations (viz. sublethal I, sublethal II, and sublethal III) were determined to be 1.48, 0.74, and 0.59 ppm, respectively, and the fish specimens were exposed to these concentrations. Tissue samplings were done on days 0, 1, 3, 7, 15, 22 and 29 of malathion exposure for assessment of the induction of micronuclei (MN) frequency and DNA damage. The MN formation in the peripheral blood cells was found to be significantly higher (p < 0.05) in the treated specimens at all sampling intervals compared to the control. The MN frequency reached maximum on days 3 and 7 at sublethal I and II concentrations, respectively, followed by a nonlinear decline with the progression of the experiment. Similarly, significant effects (p < 0.05) of both concentration and time of exposure were observed on DNA damage in the gill, kidney, and lymphocytes. All of the tissues exhibited a concentration-dependent increase in DNA damage up to day 3, followed by a nonlinear decrease with the duration of exposure. A comparison of the extent of DNA damage among the tissues showed the sensitivity of gill tissue to malathion.


Assuntos
Ensaio Cometa/métodos , Dano ao DNA , Inseticidas/toxicidade , Malation/toxicidade , Testes para Micronúcleos/métodos , Perciformes/genética , Animais , Água Doce
12.
Carcinogenesis ; 30(5): 799-807, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19028703

RESUMO

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.


Assuntos
Aldeído Redutase/deficiência , Azoximetano/toxicidade , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/prevenção & controle , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/prevenção & controle , Aldeído Redutase/metabolismo , Animais , Neoplasias do Colo/patologia , Primers do DNA , Peroxidação de Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/fisiologia , Lesões Pré-Cancerosas/patologia , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Endocrinology ; 150(1): 63-74, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18772236

RESUMO

Chronic low-grade inflammation has emerged as a key contributor to the cardiovascular complications of diabetes, however, the mechanisms by which diabetes increases inflammation remain poorly understood. Here, we report that exposure to high glucose (HG) stimulates ectodomain shedding of TNF-alpha from rat aortic smooth muscle cells in culture. Our results show that exposure to HG decreases membrane-associated TNF-alpha. This decrease in unprocessed TNF-alpha was prevented by the aldose reductase (AR) inhibitor sorbinil and AR small interference RNA. Treatment with HG, but not equimolar mannitol or 3-O-methyl glucose, resulted in phosphorylation and activation of TNF-alpha converting enzyme (TACE) (ADAM17), which were attenuated by sorbinil or AR-specific small interference RNA. HG-induced TACE phosphorylation and TNF-alpha processing were also prevented by TNF-alpha protease inhibitor-1, an inhibitor of TACE. Inhibition of protein kinase C (PKC)-delta by rottlerin prevented HG-induced TACE activation and the accumulation of unprocessed TNF-alpha. Treatment with sorbinil decreased elevated levels of circulating TNF-alpha in streptozotocin-treated diabetic rats. Sorbinil treatment also decreased the expression of TNF-alpha, matrix metalloproteinase-2, matrix metalloproteinase-9, and increased tissue inhibitor of metalloproteinase-3 in vascular smooth muscle cells treated with HG and in balloon-injured carotid arteries of diabetic rats. These results indicate that HG-induced TNF-alpha shedding could be attributed to TACE activation, which is regulated, in part, by PKC-delta and AR. Therefore, inhibition of TACE by TNF-alpha protease inhibitor-1, or pharmacological inhibition of PKC-delta or AR may represent useful strategies for treating vascular inflammation associated with diabetes.


Assuntos
Proteínas ADAM/metabolismo , Aldeído Redutase/metabolismo , Glucose/farmacologia , Músculo Liso Vascular/enzimologia , Proteína Quinase C-delta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína ADAM17 , Animais , Dipeptídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Inflamação/prevenção & controle , Cinética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Fosforilação , Ratos
14.
Endocr Rev ; 26(3): 380-92, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15814847

RESUMO

Aldose reductase (AR) is widely expressed aldehyde-metabolizing enzyme. The reduction of glucose by the AR-catalyzed polyol pathway has been linked to the development of secondary diabetic complications. Although treatment with AR inhibitors has been shown to prevent tissue injury in animal models of diabetes, the clinical efficacy of these drugs remains to be established. Recent studies suggest that glucose may be an incidental substrate of AR, which appears to be more adept in catalyzing the reduction of a wide range of aldehydes generated from lipid peroxidation. Moreover, inhibition of the enzyme has been shown to increase inflammation-induced vascular oxidative stress and prevent myocardial protection associated with the late phase of ischemic preconditioning. On the basis of these studies, several investigators have ascribed an important antioxidant role to the enzyme. Additionally, ongoing work indicates that AR is a critical component of intracellular signaling, and inhibition of the enzyme prevents high glucose-, cytokine-, or growth factor-induced activation of protein kinase C and nuclear factor-kappa-binding protein. Thus, treatment with AR inhibitors prevents vascular smooth muscle cell growth and endothelial cell apoptosis in culture and inflammation and restenosis in vivo. Additional studies indicate that the antioxidant and signaling roles of AR are interlinked and that AR regulates protein kinase C and nuclear factor-kappaB via redox-sensitive mechanisms. These data underscore the need for reevaluating anti-AR interventions for the treatment of diabetic complications. Potentially, the development of newer drugs that selectively inhibit AR-mediated glucose metabolism and signaling, without affecting aldehyde detoxification, may be useful in preventing inflammation associated with the development of diabetic complications, particularly micro- and macrovascular diseases.


Assuntos
Aldeído Redutase/metabolismo , Complicações do Diabetes/metabolismo , Aldeído Redutase/antagonistas & inibidores , Animais , Catarata/complicações , Catarata/metabolismo , Complicações do Diabetes/enzimologia , Humanos , Imidazolidinas/farmacologia , Modelos Moleculares , Naftalenos/farmacologia , Oxirredução , Estresse Oxidativo/fisiologia , Transdução de Sinais/fisiologia
15.
Cytokine ; 48(3): 170-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19660963

RESUMO

AIM: Increased production of cytokines and chemokines in serum and tissues upon oxidative stress caused by severe systemic infections are the major cause of sepsis. Aldose reductase (AR) known to mediate oxidative stress-induced NF-kappaB activation and transcription of cytokines and chemokines are the main mediator of bacterial endotoxin-induced inflammatory response. Our aim is to investigate the effect of AR inhibitors on the prevention of inflammatory cytokines in the cecum ligation and puncture (CLP) model of polymicrobial sepsis which closely mimics the sepsis syndrome in humans. RESULTS: Mice were rendered septic by CLP in the absence and presence of AR inhibitor, sorbinil. The levels of cytokines, chemokines and other inflammatory markers in the plasma, peritoneal fluid and heart of mice were significantly inhibited by sorbinil. Inhibition of AR also prevented CLP-induced COX-2, iNOS and HMGB-1 in heart, kidney and spleen. CONCLUSIONS: Our results showed that the inhibition of AR significantly prevented the polymicrobial sepsis-induced increase in inflammatory markers and thus indicate the use of AR inhibitors as anti-inflammatory agents.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Anti-Inflamatórios/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Síndrome de Resposta Inflamatória Sistêmica/prevenção & controle , Aldeído Redutase/metabolismo , Animais , Citocinas/análise , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Imidazolidinas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia
16.
Exp Eye Res ; 89(4): 494-502, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19524565

RESUMO

Cadmium (Cd), released from cigarette smoke and metal industrial activities, is known to accumulate in human body organs including retina and is particularly higher in retinal tissues of age-related macular degeneration (AMD) eyes compared to non-AMD eyes. We have determined the cytotoxic effects of Cd on human retinal pigment epithelial (RPE) cells. Upon Cd treatment, there was a dose- and time-dependent decline in ARPE-19 cell viability as well as early apoptotic changes such as altered mitochondrial membrane potential (MMP) and Cytochrome C release in cytosol. Depletion of GSH by buthionine-[S,R]-sulfoximine (BSO) resulted in increased Cd toxicity in ARPE-19 cells. Cadmium also caused reactive oxygen species (ROS) generation and activation of mitogen-activated protein kinases (MAPKs) pathway including c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase 1/2 (Erk1/2), and p38 in ARPE-19 cells. Antioxidants such as N-acetylcysteine (NAC) significantly reduced Cd-induced toxicity. These results indicate that elevated ROS-induced activation of the MAPK signaling pathway could be associated with Cd-induced RPE cell apoptosis, one of the major contributing factors in AMD. The toxic effects of Cd on ARPE-19 cells indicate that environmental heavy metals such as Cd could be important potential factors in RPE cells death associated retinal diseases particularly related to smoking.


Assuntos
Apoptose/efeitos dos fármacos , Cloreto de Cádmio/toxicidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Epitélio Pigmentado da Retina/patologia , Acetilcisteína/farmacologia , Western Blotting , Butionina Sulfoximina/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocromos c/metabolismo , Citosol/enzimologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/toxicidade , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Fatores de Tempo
17.
J Toxicol Environ Health A ; 72(9): 577-84, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19296407

RESUMO

Earlier studies showed that human lens ALDH1A1 plays a critical role in protection against oxidative stress-induced cytotoxicity in human lens epithelial cells (HLEC), and opacification of rat and mouse lens. The complete coding sequence of ALDH1A1 was cloned from human lens cDNA library by using PCR methods and expressed it in Escherichia coli. The cloned human lens ALDH1A1 cDNA encodes a 501-amino-acid protein (molecular mass = 54.8 kD) that is 100% identical to human liver ALDH1A1 and shares significant identity with the same isozyme from other tissues and species. The purified recombinant human lens ALDH1A1 exhibited optimal catalytic activity at pH 8 and preferred NAD(+) as cofactor and specifically catalyzed the oxidation of toxic lipid aldehydes such as 4-hydroxynonenal (HNE; K(m) = 4.8 microM) and malonaldehyde (K(m) MDA = 3.5 microM). Citral, disulfiram, and cyanamide were found to inhibit human lens ALDH1A1 at IC50 values of 55, 101, and 22610 microM, respectively, whereas diethylstilbestrol (DES) was found to be an activator (EC(50), 1.3 microM). Further, modification of recombinant human lens ALDH1A1 with nitric oxide donors such as S-nitroso-N-acetylpenicillamine (SNAP) and S-nitrosoglutathione (GSNO) significantly inhibited the enzyme activity. It therefore appears that activation of ALDH1A1, which efficiently catalyzes the detoxification of lipid-derived toxic aldehydes, and/or prevention of its oxidative modification may be novel therapeutic interventions against oxidative stress-induced lens pathologies.


Assuntos
Aldeído Desidrogenase/metabolismo , Catarata/enzimologia , Cristalino/enzimologia , Estresse Oxidativo , Aldeído Desidrogenase/genética , Família Aldeído Desidrogenase 1 , Catarata/etiologia , Humanos , Oxirredução , Proteínas Recombinantes/metabolismo , Retinal Desidrogenase
18.
J Mol Endocrinol ; 63(1): 11-25, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30986766

RESUMO

Although hyperglycemia-mediated death and dysfunction of endothelial cells have been reported to be a major cause of diabetes associated vascular complications, the mechanisms through which hyperglycemia cause endothelial dysfunction is not well understood. We have recently demonstrated that aldose reductase (AR, AKR1B1) is an obligatory mediator of oxidative and inflammatory signals induced by growth factors, cytokines and hyperglycemia. However, the molecular mechanisms by which AR regulates hyperglycemia-induced endothelial dysfunction is not well known. In this study, we have investigated the mechanism(s) by which AR regulates hyperglycemia-induced endothelial dysfunction. Incubation of human umbilical vein endothelial cells (HUVECs) with high glucose (HG) decreased the cell viability and inhibition of AR prevented it. Further, AR inhibition prevented the HG-induced ROS generation and expression of BCL-2, BAX and activation of Caspase-3 in HUVECs. AR inhibition also prevented the adhesion of THP-1 monocytes on HUVECs, expression of iNOS and eNOS and adhesion molecules ICAM-1 and VCAM-1 in HG-treated HUVECs. Further, AR inhibition restored the HG-induced depletion of SIRT1 in HUVECs and increased the phosphorylation of AMPKα1 along-with a decrease in phosphorylation of mTOR in HG-treated HUVECs. Fidarestat decreased SIRT1 expression in HUVECs pre-treated with specific SIRT1 inhibitor but not with the AMPKα1 inhibitor. Similarly, knockdown of AR in HUVECs by siRNA prevented the HG-induced HUVECs cell death, THP-1 monocyte adhesion and SIRT1 depletion. Furthermore, fidarestat regulated the phosphorylation of AMPKα1 and mTOR, and expression of SIRT1 in STZ-induced diabetic mice heart and aorta tissues. Collectively, our data suggest that AR regulates hyperglycemia-induced endothelial death and dysfunction by altering the ROS/SIRT1/AMPKα1/mTOR pathway.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Aldeído Redutase/metabolismo , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Sirtuína 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Aldeído Redutase/antagonistas & inibidores , Animais , Caspase 3/genética , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glucose/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Imidazolidinas/farmacologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 1/genética , Células THP-1 , Serina-Treonina Quinases TOR/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
19.
FEBS Lett ; 582(17): 2609-14, 2008 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-18586034

RESUMO

We tested the hypothesis that activation of the polyol pathway and protein kinase C (PKC) during diabetes is due to loss of NO. Our results show that after 4 weeks of streptozotocin-induced diabetes, treatment with L-arginine restored NO levels and prevented tissue accumulation of sorbitol in mice, which was accompanied by an increase in glutathiolation of aldose reductase. L-Arginine treatment decreased superoxide generation in the aorta, total PKC activity and PKC-beta(II) phosphorylation in the heart, and the plasma levels of triglycerides and soluble ICAM. These data suggest that increasing NO bioavailability by L-arginine corrects the major biochemical abnormalities of diabetes.


Assuntos
Aortite/prevenção & controle , Arginina/uso terapêutico , Diabetes Mellitus Tipo 1/complicações , Cardiopatias/prevenção & controle , Hiperglicemia/complicações , Óxido Nítrico/biossíntese , Aldeído Redutase/antagonistas & inibidores , Animais , Aortite/etiologia , Aortite/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animais de Doenças , Glutationa/metabolismo , Cardiopatias/etiologia , Cardiopatias/metabolismo , Hiperglicemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Polímeros/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Sorbitol/metabolismo , Superóxidos/metabolismo
20.
Curr Cancer Drug Targets ; 18(9): 905-911, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28786349

RESUMO

BACKGROUND: Recent epidemiological and experimental studies have shown that obesity is a major risk factor for Colorectal Cancer (CRC). Regular intake of high fat-containing diet can promote obesity and metabolic syndrome by increasing the insulin resistance and inflammatory response which contribute to carcinogenesis. Previously, we have shown that inhibition of polyol pathway enzyme aldose reductase (AR) prevents carcinogens- and inflammatory growth factorsinduced CRC. However, the effect of AR inhibition on a high-fat diet (HFD)-induced formation of intestinal polyps in Apc-deficient Min (multiple intestinal neoplasia; ApcMin/+) mice is not known. METHODS: We examined the effect of AR inhibitor, fidarestat on the HFD-induced formation of preneoplastic intestinal polyps in ApcMin/+ mice which is an excellent model of colon cancer. RESULTS: APCMin/+ mice fed for 12 weeks of HFD caused a significant increase in the formation of polyps in the small and large intestines and fidarestat given along with the HFD prevented the number of intestinal polyps. Fidarestat also decreased the size of the polyps in the intestines of HFDtreated APC Min mice. Further, the expression levels of beta-catenin, PCNA, PKC-ß2, P-AKT, Pp65, COX-2, and iNOS in the small and large intestines of HFD-treated mice significantly increased, and AR inhibitor prevented it. CONCLUSION: Our results thus suggest that fidarestat could be used as a potential chemopreventive drug for intestinal cancers due to APC gene mutations.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Aldeído Redutase/antagonistas & inibidores , Neoplasias do Colo/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Inibidores Enzimáticos/farmacologia , Imidazolidinas/farmacologia , Pólipos Intestinais/prevenção & controle , Animais , Neoplasias do Colo/enzimologia , Neoplasias do Colo/etiologia , Feminino , Pólipos Intestinais/enzimologia , Pólipos Intestinais/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa