Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Endocrinol ; 63(1): 11-25, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30986766

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aldehído Reductasa/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Sirtuina 1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Aldehído Reductasa/antagonistas & inhibidores , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Imidazolidinas/farmacología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 1/genética , Células THP-1 , Serina-Treonina Quinasas TOR/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
4.
Proc Natl Acad Sci U S A ; 115(15): 3918-3923, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29572430

RESUMEN

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.


Asunto(s)
Proteínas Activadoras de GTPasa/deficiencia , Neoplasias/genética , Neoplasias/prevención & control , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis , Femenino , Proteínas Activadoras de GTPasa/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/metabolismo , Neoplasias/fisiopatología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo
5.
Biochem Pharmacol ; 150: 181-190, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29458045

RESUMEN

Despite doxorubicin (Dox) being one of the most widely used chemotherapy agents for breast, blood and lung cancers, its use in colon cancer is limited due to increased drug resistance and severe cardiotoxic side effects that increase mortality associated with its use at high doses. Therefore, better adjuvant therapies are warranted to improve the chemotherapeutic efficacy and to decrease cardiotoxicity. We have recently shown that aldose reductase inhibitor, fidarestat, increases the Dox-induced colon cancer cell death and reduces cardiomyopathy. However, the efficacy of fidarestat in the prevention of Dox-induced endothelial dysfunction, a pathological event critical to cardiovascular complications, is not known. Here, we have examined the effect of fidarestat on Dox-induced endothelial cell toxicity and dysfunction in vitro and in vivo. Incubation of human umbilical vein endothelial cells (HUVECs) with Dox significantly increased the endothelial cell death, and pre-treatment of fidarestat prevented it. Further, fidarestat prevented the Dox-induced oxidative stress, formation of reactive oxygen species (ROS) and activation of Caspase-3 in HUVECs. Fidarestat also prevented Dox-induced monocyte adhesion to HUVECs and expression of ICAM-1 and VCAM-1. Fidarestat pre-treatment to HUVECs restored the Dox-induced decrease in the Nitric Oxide (NO)-levels and eNOS expression. Treatment of HUVECs with Dox caused a significant increase in the activation of NF-κB and expression of various inflammatory cytokines and chemokines which were prevented by fidarestat pre-treatment. Most importantly, fidarestat prevented the Dox-induced mouse cardiac cell hypertrophy and expression of eNOS, iNOS, and 3-Nitrotyrosine in the aorta tissues. Further, fidarestat blunted the Dox-induced expression of various inflammatory cytokines and chemokines in vivo. Thus, our results suggest that by preventing Dox-induced endothelial cytotoxicity and dysfunction, AR inhibitors could avert cardiotoxicity associated with anthracycline chemotherapy.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Imidazolidinas/farmacología , Mediadores de Inflamación/antagonistas & inhibidores , Aldehído Reductasa/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Relación Dosis-Respuesta a Droga , Doxorrubicina/antagonistas & inhibidores , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
6.
Curr Cancer Drug Targets ; 18(9): 905-911, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28786349

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Aldehído Reductasa/antagonistas & inhibidores , Neoplasias del Colon/prevención & control , Dieta Alta en Grasa/efectos adversos , Inhibidores Enzimáticos/farmacología , Imidazolidinas/farmacología , Pólipos Intestinales/prevención & control , Animales , Neoplasias del Colon/enzimología , Neoplasias del Colon/etiología , Femenino , Pólipos Intestinales/enzimología , Pólipos Intestinales/etiología , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Cancer Lett ; 411: 57-63, 2017 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-28986187

RESUMEN

Although we have shown earlier that aldose reductase (AR) inhibitors prevent colorectal cancer cell (CRC) growth in culture as well as in nude mice xenografts, the mechanism(s) is not well understood. In this study, we have investigated how AR inhibition prevents CRC growth by regulating the mitochondrial biogenesis via Nrf2/HO-1 pathway. Incubation of CRC cells such as SW-480, HT29, and HCT116 with AR inhibitor, fidarestat that non-covalently binds to the enzyme, increases the expression of Nrf2. Further, fidarestat augmented the EGF-induced expression of Nrf2 in CRC cells. Fidarestat also increased the Nrf2 -DNA binding activity as well as expression of HO-1 and NQO1 and activation of SOD and catalase in SW480 cells. Similarly, in nude mice xenograft tumor tissues, Nrf2 and HO-1 levels were significantly higher in fidarestat-treated mice compared to controls. Further, stimulation of CRC cells with EGF in the presence of fidarestat increased the mRNA levels of PGC-1α, Nrf1 and TFAM and protein levels of PGC-1α, TFAM and COX-IV and decreased the mitochondrial DNA damage as measured by 8-hydroxy-2'-deoxyguanosine levels. AR inhibitor also modulated the phosphorylations of AMPK and mTOR and expression of p53 in EGF-treated cells. Collectively, our results indicate that AR inhibitor prevents CRC growth by increasing mitochondrial biogenesis via increasing the expression of Nrf2/HO-1/AMPK/p53 and decreasing the mitochondrial DNA damage.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Neoplasias del Colon/tratamiento farmacológico , Hemo-Oxigenasa 1/metabolismo , Imidazolidinas/farmacología , Mitocondrias/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Aldehído Reductasa/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células HCT116 , Células HT29 , Humanos , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Biogénesis de Organelos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Endocrinology ; 158(10): 3661-3675, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28938395

RESUMEN

Despite recent studies that show oxidative stress-generated reactive oxygen species (ROS) regulate NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated innate immune response in various diabetic complications, the mechanism by which ROS activate innate immune response is not well understood. We have shown previously that aldose reductase (AR), besides reducing glucose, reduces lipid aldehydes and their glutathione conjugates and participates in various oxidative stress-induced inflammatory pathways. To understand the role of AR in ROS-induced innate immune response, we have investigated the mechanism(s) by which AR activates hyperglycemia-induced NLRP3 inflammsome-initiated innate immune response in Thp1 monocytes and in streptozotocin (STZ)-induced diabetic mice. In Thp1 monocytes, inhibition or ablation of AR prevented high-glucose-induced activation of NLRP3 inflammasome and caspase-1 and release of the innate immune cytokines interleukin (IL)-1ß and IL-18. AR inhibition in Thp1 cells also prevented the high-glucose-induced generation of ROS, influx of Ca2+, efflux of K+, and activation of Lyn, Syk, and PI3K. Furthermore, the AR inhibitor fidarestat prevented the expression of NLRP inflammasome components in STZ-induced diabetic mouse heart and aorta, and also prevented the release of various cytokines in the serum. Collectively, our data suggest that AR regulates hyperglycemia-induced NLRP3 inflammasome-mediated innate immune response by altering the ROS/Lyn/Syk/PI3K/Ca2+/K+ signals.


Asunto(s)
Aldehído Reductasa/genética , Diabetes Mellitus Experimental/inmunología , Hiperglucemia/inmunología , Inmunidad Innata/inmunología , Inflamasomas/inmunología , Monocitos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Estrés Oxidativo/inmunología , Aldehído Reductasa/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Supervivencia Celular , Citocinas/inmunología , Diabetes Mellitus Experimental/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Hiperglucemia/metabolismo , Immunoblotting , Interleucina-1beta/inmunología , Masculino , Ratones , Monocitos/metabolismo , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Diabetes Res ; 2017: 6785852, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28740855

RESUMEN

We have shown earlier that pretreatment of cultured cells with aldose reductase (AR) inhibitors prevents hyperglycemia-induced mitogenic and proinflammatory responses. However, the effects of AR inhibitors on Nrf2-mediated anti-inflammatory responses have not been elucidated yet. We have investigated how AR inhibitor fidarestat protects high glucose- (HG-) induced cell viability changes by increasing the expression of Nrf2 and its dependent phase II antioxidant enzymes. Fidarestat pretreatment prevents HG (25 mM)-induced Thp1 monocyte viability. Further, treatment of Thp1 monocytes with fidarestat caused a time-dependent increase in the expression as well as the DNA-binding activity of Nrf2. In addition, fidarestat augmented the HG-induced Nrf2 expression and activity and also upregulated the expression of Nrf2-dependent proteins such as hemeoxygenase-1 (HO1) and NQO1 in Thp1 cells. Similarly, treatment with AR inhibitor also induced the expression of Nrf2 and HO1 in STZ-induced diabetic mice heart and kidney tissues. Further, AR inhibition increased the HG-induced expression of antioxidant enzymes such as SOD and catalase and activation of AMPK-α1 in Thp1 cells. Our results thus suggest that pretreatment with AR inhibitor prepares the monocytes against hyperglycemic stress by overexpressing the Nrf2-dependent antioxidative proteins.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Diabetes Mellitus Experimental/metabolismo , Hiperglucemia/metabolismo , Imidazolidinas/farmacología , Monocitos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Catalasa/metabolismo , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Corazón/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Monocitos/metabolismo , Miocardio/metabolismo , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo
10.
Sci Rep ; 7(1): 3182, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28600556

RESUMEN

Anthracycline drugs such as doxorubicin (DOX) and daunorubicin remain some of the most active wide-spectrum and cost-effective drugs in cancer therapy. However, colorectal cancer (CRC) cells are inherently resistant to anthracyclines which at higher doses cause cardiotoxicity. Our recent studies indicate that aldose reductase (AR) inhibitors such as fidarestat inhibit CRC growth in vitro and in vivo. Here, we show that treatment of CRC cells with fidarestat increases the efficacy of DOX-induced death in HT-29 and SW480 cells and in nude mice xenografts. AR inhibition also results in higher intracellular accumulation of DOX and decreases the expression of drug transporter proteins MDR1, MRP1, and ABCG2. Further, fidarestat also inhibits DOX-induced increase in troponin-I and various inflammatory markers in the serum and heart and restores cardiac function in mice. These results suggest that fidarestat could be used as adjuvant therapy to enhance DOX sensitivity of CRC cells and to reduce DOX-associated cardiotoxicity.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Cardiotoxicidad/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Imidazolidinas/administración & dosificación , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Aldehído Reductasa/genética , Animales , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Doxorrubicina/efectos adversos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Ratones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas de Neoplasias/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Free Radic Biol Med ; 111: 235-243, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27794453

RESUMEN

Glutathione-S-Transferases (GSTs) have primarily been thought to be xenobiotic metabolizing enzymes that protect cells from toxic drugs and environmental electrophiles. However, in last three decades, these enzymes have emerged as the regulators of oxidative stress-induced signaling and toxicity. 4-Hydroxy-trans 2-nonenal (HNE) an end-product of lipid peroxidation, has been shown to be a major determinant of oxidative stress-induced signaling and toxicity. HNE is involved in signaling pathways, including apoptosis, proliferation, modulation of gene expression, activation of transcription factors/repressors, cell cycle arrest, and differentiation. In this article, available evidence for a major role of GSTs in the regulation of HNE-mediated cell signaling processes through modulation of the intracellular levels of HNE is discussed.


Asunto(s)
Aldehídos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Glutatión Transferasa/metabolismo , Estrés Oxidativo/genética , Transducción de Señal/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Proteínas Co-Represoras , Regulación de la Expresión Génica , Glutatión Transferasa/genética , Factores de Transcripción del Choque Térmico/genética , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Peroxidación de Lípido , Chaperonas Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
13.
Cancer Lett ; 355(1): 141-7, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25218594

RESUMEN

Type-2 diabetes and obesity-related metabolic abnormalities are major risk factors for the development of colon cancer. In the present study, we examined the effects of polyol pathway enzyme aldose reductase (AR) inhibitor, fidarestat, on the development of azoxymethane (AOM)-induced colonic premalignant lesions in C57BL/KsJ-db/db obese mice. Our results indicate that fidarestat given in the drinking water caused a significant reduction in the total number of colonic premalignant lesions in the AOM treated obese mice. Further, the expression levels of PKC-ß2, AKT, COX-2 and iNOS in the colonic mucosa of AOM-treated mice were significantly decreased by fidarestat. The serum levels of IL-1α, IP-10, MIG, TNF-α and VEGF are significantly suppressed in AOM + fidarestat treated obese mice. Fidarestat also decreased the expression of COX-2, iNOS, XIAP, survivin, ß-catenin and NF-κB in high glucose-treated HT29 colon cancer cells. In conclusion, our results indicate that fidarestat inhibits the development of colonic premalignant lesions in an obesity-related colon cancer and is chemopreventive to colorectal carcinogenesis in obese individuals.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Anticarcinógenos/farmacología , Azoximetano , Colon/efectos de los fármacos , Neoplasias del Colon/prevención & control , Diabetes Mellitus Tipo 2/complicaciones , Inhibidores Enzimáticos/farmacología , Imidazolidinas/farmacología , Mucosa Intestinal/efectos de los fármacos , Obesidad/complicaciones , Lesiones Precancerosas/prevención & control , Aldehído Reductasa/metabolismo , Animales , Colon/enzimología , Colon/patología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Células HT29 , Humanos , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Masculino , Ratones Endogámicos C57BL , Obesidad/enzimología , Obesidad/genética , Estrés Oxidativo/efectos de los fármacos , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Transducción de Señal/efectos de los fármacos
14.
Curr Med Chem ; 21(2): 230-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23848536

RESUMEN

Metastable aldehydes produced by lipid peroxidation act as 'toxic second messengers' that extend the injurious potential of free radicals. 4-hydroxy 2-nonenal (HNE), a highly toxic and most abundant stable end product of lipid peroxidation, has been implicated in the tissue damage, dysfunction, injury associated with aging and other pathological states such as cancer, Alzheimer, diabetes, cardiovascular and inflammatory complications. Further, HNE has been considered as a oxidative stress marker and it act as a secondary signaling molecule to regulates a number of cell signaling pathways. Biological activity of HNE depends on its intracellular concentration, which can differentially modulate cell death, growth and differentiation. Therefore, the mechanisms responsible for maintaining the intracellular levels of HNE are most important, not only in the defense against oxidative stress but also in the pathophysiology of a number of disease processes. In this review, we discussed the significance of HNE in mediating various disease processes and how regulation of its metabolism could be therapeutically effective.


Asunto(s)
Aldehídos/metabolismo , Progresión de la Enfermedad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Peroxidación de Lípido , Estructura Molecular , Neoplasias/metabolismo , Neoplasias/patología
15.
Eur J Cancer ; 49(15): 3311-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23827854

RESUMEN

Inhibition of polyol pathway enzyme aldose reductase (AR) has been shown to prevent colon cancer cells growth in culture and in nude mice xenografts. However, the role of AR in the mediation of growth factor-induced colon cancer cells growth is not well understood. In this study, we have investigated how AR inhibition prevents tumour growth via regulation of microRNA (miR)-21-mediated programmed cell death 4 (PDCD4) expression in colon cancer cells in in vitro and in vivo. Treatment of colon cancer cells (HT29, SW480 and Caco-2) with epidermal growth factor (EGF) caused increased expression of miR-21 and inhibition of AR prevented it. Further, AR inhibition also increased PDCD4, a putative target of miR-21 in human colon cancer cells. Inhibition of AR also prevented EGF-induced phosphorylation of PDCD4. Treatment of HT29 cells with AR inhibitor, fidarestat, prevented the EGF-induced phosphorylation of mammalian target of rapamycin (mTOR), regulatory associated protein of mTOR (Raptor), eukaryotic initiation factor 4E (eIF4E), p70 S6 kinase (S6K) and eukaryotic initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) and increased the phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK). Similarly, in nude mice xenograft tissues, PDCD4 and 4E-BP1 levels were significantly higher in AR inhibitor-treated mice compared to controls. Collectively, these results indicate that AR inhibition prevents growth factor-induced colon cancer growth by down-regulating miR-21 expression and increasing PDCD4 levels through the reactive oxygen species (ROS)/AMPK/mTOR/AP1/4E-BP1 pathway.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/biosíntesis , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Imidazolidinas/farmacología , MicroARNs/metabolismo , Proteínas de Unión al ARN/biosíntesis , Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Células CACO-2 , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Células HT29 , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , MicroARNs/genética , Fosforilación , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Free Radic Biol Med ; 63: 280-90, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23732517

RESUMEN

One of the major problems associated with the chemotherapy of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) that selectively kills tumor cells is decreased drug resistance. This warranted the development of safe novel pharmacological agents that could sensitize the tumor cells to TRAIL. Herein, we examined the role of aldose reductase (AR) in sensitizing cancer cells to TRAIL and potentiating TRAIL-induced apoptosis of human colon cancer cells. We demonstrate that AR inhibition potentiates TRAIL-induced cytotoxicity in cancer cells by upregulation of both death receptor (DR)-5 and DR4. Knockdown of DR5 and DR4 significantly (>85%) reduced the sensitizing effect of the AR inhibitor fidarestat on TRAIL-induced apoptosis. Further, AR inhibition also downregulates cell survival proteins (Bcl-xL, Bcl-2, survivin, XIAP, and FLIP) and upregulates the expression of proapoptotic proteins such as Bax and alters mitochondrial membrane potential, leading to cytochrome c release, caspases-3 activation, and PARP cleavage. We found that AR inhibition regulates AKT/PI3K-dependent activation of forkhead transcription factor FOXO3a. Knockdown of FOXO3a significantly (>80%) abolished AR inhibition-induced upregulation of DR5 and DR4 and apoptosis in colon cancer cells. Overall, our results show that fidarestat potentiates TRAIL-induced apoptosis through downregulation of cell survival proteins and upregulation of death receptors via activation of the AKT/FOXO3a pathway.


Asunto(s)
Aldehído Reductasa/metabolismo , Apoptosis/genética , Neoplasias del Colon/metabolismo , Factores de Transcripción Forkhead/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Aldehído Reductasa/antagonistas & inhibidores , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Inhibidores Enzimáticos/administración & dosificación , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/metabolismo , Receptores de Muerte Celular/genética , Receptores de Muerte Celular/metabolismo , Regulación hacia Arriba/efectos de los fármacos
17.
Free Radic Biol Med ; 65: 15-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23770200

RESUMEN

Aldose reductase (AR), a glucose-metabolizing enzyme, reduces lipid aldehydes and their glutathione conjugates with more than 1000-fold efficiency (Km aldehydes 5-30 µM) relative to glucose. Acrolein, a major endogenous lipid peroxidation product as well as a component of environmental pollutants and cigarette smoke, is known to be involved in various pathologies including atherosclerosis, airway inflammation, COPD, and age-related disorders, but the mechanism of acrolein-induced cytotoxicity is not clearly understood. We have investigated the role of AR in acrolein-induced cytotoxicity in primary human small airway epithelial cells (SAECs). Exposure of SAECs to varying concentrations of acrolein caused cell death in a concentration- and time-dependent manner. AR inhibition by fidarestat prevented the low-dose (5-10 µM) but not the high-dose (>10 µM) acrolein-induced SAEC death. AR inhibition protected SAECs from low-dose (5 µM) acrolein-induced cellular reactive oxygen species (ROS). Inhibition of acrolein-induced apoptosis by fidarestat was confirmed by decreased condensation of nuclear chromatin, DNA fragmentation, comet tail moment, and annexin V fluorescence. Further, fidarestat inhibited acrolein-induced translocation of the proapoptotic proteins Bax and Bad from the cytosol to the mitochondria and that of Bcl2 and BclXL from the mitochondria to the cytosol. Acrolein-induced cytochrome c release from mitochondria was also prevented by AR inhibition. The mitogen-activated protein kinases (MAPKs), such as extracellular signal-regulated kinases 1 and 2, stress-activated protein kinase/c-Jun NH2-terminal kinase, and p38MAPK, and c-Jun were transiently activated in airway epithelial cells by acrolein in a concentration- and time-dependent fashion, which was significantly prevented by AR inhibition. These results suggest that AR inhibitors could prevent acrolein-induced cytotoxicity in the lung epithelial cells.


Asunto(s)
Acroleína/toxicidad , Aldehído Reductasa/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/enzimología , Apoptosis/efectos de los fármacos , Western Blotting , Bronquios/efectos de los fármacos , Bronquios/enzimología , Células Cultivadas , Ensayo Cometa , Daño del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Imidazolidinas/farmacología , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Transfección
18.
PLoS One ; 8(2): e57442, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23460857

RESUMEN

BACKGROUND: Long-term and unresolved airway inflammation and airway remodeling, characteristic features of chronic asthma, if not treated could lead to permanent structural changes in the airways. Aldose reductase (AR), an aldo-sugar and lipid aldehyde metabolizing enzyme, mediates allergen-induced airway inflammation in mice, but its role in the airway remodeling is not known. In the present study, we have examined the role of AR on airway remodeling using ovalbumin (OVA)-induced chronic asthma mouse model and cultured human primary airway epithelial cells (SAECs) and mouse lung fibroblasts (mLFs). METHODS: Airway remodeling in chronic asthma model was established in mice sensitized and challenged twice a week with OVA for 6 weeks. AR inhibitor, fidarestat, was administered orally in drinking water after first challenge. Inflammatory cells infiltration in the lungs and goblet cell metaplasia, airway thickening, collagen deposition and airway hyper-responsiveness (AHR) in response to increasing doses of methacholine were assessed. The TGFß1-induced epithelial-mesenchymal transition (EMT) in SAECs and changes in mLFs were examined to investigate AR-mediated molecular mechanism(s) of airway remodeling. RESULTS: In the OVA-exposed mice for 6 wks inflammatory cells infiltration, levels of inflammatory cytokines and chemokines, goblet cell metaplasia, collagen deposition and AHR were significantly decreased by treatment with AR inhibitor, fidarestat. Further, inhibition of AR prevented TGFß1-induced altered expression of E-cadherin, Vimentin, Occludin, and MMP-2 in SAECs, and alpha-smooth muscle actin and fibronectin in mLFs. Further, in SAECs, AR inhibition prevented TGFß1- induced activation of PI3K/AKT/GSK3ß pathway but not the phosphorylation of Smad2/3. CONCLUSION: Our results demonstrate that allergen-induced airway remodeling is mediated by AR and its inhibition blocks the progression of remodeling via inhibiting TGFß1-induced Smad-independent and PI3K/AKT/GSK3ß-dependent pathway.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Aldehído Reductasa/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Hipersensibilidad/enzimología , Hipersensibilidad/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Aldehído Reductasa/metabolismo , Animales , Asma/tratamiento farmacológico , Asma/enzimología , Biomarcadores/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipersensibilidad/patología , Imidazolidinas/farmacología , Imidazolidinas/uso terapéutico , Inflamación/patología , Pulmón/enzimología , Pulmón/patología , Metaplasia , Ratones , Moco/metabolismo , Ovalbúmina , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
19.
Antioxid Redox Signal ; 18(11): 1249-62, 2013 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22978663

RESUMEN

AIMS: We have shown earlier that inhibition of aldose reductase (AR), an oxidative stress-response protein, prevents colon cancer cell growth in vitro and in vivo. Changes in microribonucleic acid (miR) expression can contribute to cancer by modulating the functional expression of critical genes involved in cancer growth and metastasis. However, the molecular mechanisms by which AR regulates miR expression and their dependent mitogenic effects in cancer cells are not known. Therefore, we investigated how AR regulates growth factor-induced expression of miRs and growth of colon cancer cells. RESULTS: Inhibition of AR significantly downregulated growth factor-induced miR-21 expression in human colon cancer cells, HT29, SW480, and Caco-2. Further, AR inhibition also increased phosphatase and tensin homolog (PTEN) (a direct target of miR-21) and forkhead box O3A (FOXO3a) in colon cancer cells. Our results obtained with HT29 cells ablated with FOXO3a siRNA showed increased activator protein-1 (AP-1) activation and miR-21 expression, indicating that FOXO3a represses miR-21 via AP-1 inactivation. Inhibition of AR also prevented the epidermal growth factor-induced phosphorylation of phosphatidylinositol 3-kinase (PI3K), serine/threonine kinase (AKT), c-Jun, c-Fos, PTEN, and FOXO3a, and deoxyribonucleic acid (DNA)-binding activity of AP-1. More importantly, in human colon adenocarcinoma xenograft tissues, miR-21 expression was lower, and PTEN and FOXO3a levels were significantly higher in AR inhibitor-treated mice compared to controls. INNOVATION: These findings demonstrate a novel role of AR in the regulation of miR-21 and its target PTEN in growth factor-induced colon cancer cell growth. CONCLUSIONS: Collectively, these results show a novel role of AR in mediation of growth factor-induced colon cancer growth by modulating miR-21, PTEN, and FOXO3a expression through reactive oxygen species (ROS)/PI3K/AKT/AP-1.


Asunto(s)
Aldehído Reductasa/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Factores de Transcripción Forkhead/metabolismo , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Aldehído Reductasa/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Factor de Transcripción AP-1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Diabetes Complications ; 26(5): 369-77, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22658411

RESUMEN

OBJECTIVE: Since diabetes-induced vascular endothelial growth factor (VEGF) is implicated in retinal angiogenesis, we aimed to examine the role of aldose reductase (AR) in VEGF-induced human retinal endothelial cells (HREC) growth and tube formation. MATERIALS AND METHODS: HRECs were stimulated with VEGF and cell-growth was determined by MTT assay. AR inhibitor, fidarestat, to block the enzyme activity and AR siRNA to ablate AR gene expression in HREC were used to investigate the role of AR in neovascularization using cell-migration and tube formation assays. Various signaling intermediates and angiogenesis markers were assessed by Western blot analysis. Immuno-histochemical analysis of diabetic rat eyes was performed to examine VEGF expression in the retinal layer. RESULTS: Stimulation of primary HREC with VEGF caused increased cell growth and migration, and AR inhibition with fidarestat or ablation with siRNA significantly prevented it. VEGF-induced tube formation in HREC was also significantly prevented by fidarestat. Treatment of HREC with VEGF also increased the expression of VCAM, AR, and phosphorylation and activation of Akt and p38-MAP kinase, which were prevented by fidarestat. VEGF-induced expression of VEGFRII in HREC was also prevented by AR inhibition or ablation. CONCLUSIONS: Our results indicate that inhibition of AR in HREC prevents tube formation by inhibiting the VEGF-induced activation of the Akt and p38-MAPK pathway and suggest a mediatory role of AR in ocular neovascularization generally implicated in retinopathy and AMD.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Retinopatía Diabética/prevención & control , Inhibidores Enzimáticos/farmacología , Imidazolidinas/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Biomarcadores/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Imidazolidinas/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Terapia Molecular Dirigida , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Endogámicas F344 , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...