Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Cancer ; 148(12): 3032-3040, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33521927

RESUMEN

Proteasome inhibitor (PI) therapy has improved the survival of multiple myeloma (MM) patients. However, inevitably, primary or acquired resistance to PIs leads to disease progression; resistance mechanisms are unclear. Obesity is a risk factor for MM mortality. Oxidized LDL (OxLDL), a central mediator of atherosclerosis that is elevated in metabolic syndrome (co-occurrence of obesity, insulin resistance, dyslipidemia and hypertension), has been linked to an increased risk of solid cancers and shown to stimulate pro-oncogenic/survival signaling. We hypothesized that OxLDL is a mediator of chemoresistance and evaluated its effects on MM cell killing by PIs. OxLDL potently suppressed the ability of the boronic acid-based PIs bortezomib (BTZ) and ixazomib, but not the epoxyketone-based PI carfilzomib, to kill human MM cell lines and primary cells. OxLDL suppressed BTZ-induced inhibition of proteasome activity and induction of pro-apoptotic signaling. These cytoprotective effects were abrogated when lipid hydroperoxides (LOOHs) associated with OxLDL were enzymatically reduced. We also demonstrated the presence of OxLDL in the MM bone marrow microenvironment as well as numerous granulocytes and monocytes capable of cell-mediated LDL oxidation through myeloperoxidase. Our findings suggest that OxLDL may be a potent mediator of boronic acid-based PI resistance, particularly for MM patients with metabolic syndrome, given their elevated systemic levels of OxLDL. LDL cholesterol-lowering therapy to reduce circulating OxLDL, and pharmacologic targeting of LOOH levels or resistance pathways induced by the modified lipoprotein, could deepen the response to these important agents and offer clinical benefit to MM patients with metabolic syndrome.


Asunto(s)
Resistencia a Antineoplásicos , Lipoproteínas LDL/metabolismo , Mieloma Múltiple/metabolismo , Inhibidores de Proteasoma/farmacología , Compuestos de Boro/farmacología , Bortezomib/farmacología , Línea Celular Tumoral , Glicina/análogos & derivados , Glicina/farmacología , Granulocitos/metabolismo , Humanos , Peróxidos Lipídicos/metabolismo , Monocitos/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Oligopéptidos/farmacología , Inhibidores de Proteasoma/uso terapéutico
2.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33920782

RESUMEN

The role of tumor protein 63 (TP63) in regulating insulin receptor substrate 1 (IRS-1) and other downstream signal proteins in diabetes has not been characterized. RNAs extracted from kidneys of diabetic mice (db/db) were sequenced to identify genes that are involved in kidney complications. RNA sequence analysis showed more than 4- to 6-fold increases in TP63 expression in the diabetic mice's kidneys, compared to wild-type mice at age 10 and 12 months old. In addition, the kidneys from diabetic mice showed significant increases in TP63 mRNA and protein expression compared to WT mice. Mouse proximal tubular cells exposed to high glucose (HG) for 48 h showed significant decreases in IRS-1 expression and increases in TP63, compared to cells grown in normal glucose (NG). When TP63 was downregulated by siRNA, significant increases in IRS-1 and activation of AMP-activated protein kinase (AMPK (p-AMPK-Th172)) occurred under NG and HG conditions. Moreover, activation of AMPK by pretreating the cells with AICAR resulted in significant downregulation of TP63 and increased IRS-1 expression. Ad-cDNA-mediated over-expression of tuberin resulted in significantly decreased TP63 levels and upregulation of IRS-1 expression. Furthermore, TP63 knockdown resulted in increased glucose uptake, whereas IRS-1 knockdown resulted in a decrease in the glucose uptake. Altogether, animal and cell culture data showed a potential role of TP63 as a new candidate gene involved in regulating IRS-1 that may be used as a new therapeutic target to prevent kidney complications in diabetes.


Asunto(s)
Nefropatías Diabéticas/genética , Transactivadores/genética , Regulación hacia Arriba/genética , Adenilato Quinasa/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Glucemia/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/sangre , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Túbulos Renales Proximales/patología , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Transactivadores/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/farmacología , Regulación hacia Arriba/efectos de los fármacos
3.
FASEB J ; 33(1): 373-387, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29975570

RESUMEN

Chronic exposure of tubular renal cells to high glucose contributes to tubulointerstitial changes in diabetic nephropathy. In the present study, we identified a new fibrosis gene called galectin-1 (Gal-1), which is highly expressed in tubular cells of kidneys of type 1 and type 2 diabetic mouse models. Gal-1 protein and mRNA expression showed significant increase in kidney cortex of heterozygous Akita+/- and db/db mice compared with wild-type mice. Mouse proximal tubular cells exposed to high glucose showed significant increase in phosphorylation of Akt and Gal-1. We cloned Gal-1 promoter and identified the transcription factor AP4 as binding to the Gal-1 promoter to up-regulate its function. Transfection of cells with plasmid carrying mutations in the binding sites of AP4 to Gal-1 promoter resulted in decreased protein function of Gal-1. In addition, inhibition of Gal-1 by OTX-008 showed significant decrease in p-Akt/AP4 and protein-promoter activity of Gal-1 and fibronectin. Moreover, down-regulation of AP4 by small interfering RNA resulted in a significant decrease in protein expression and promoter activity of Gal-1. We found that kidney of Gal-1-/- mice express very low levels of fibronectin protein. In summary, Gal-1 is highly expressed in kidneys of type 1 and 2 diabetic mice, and AP4 is a major transcription factor that activates Gal-1 under hyperglycemia. Inhibition of Gal-1 by OTX-008 blocks activation of Akt and prevents accumulation of Gal-1, suggesting a novel role of Gal-1 inhibitor as a possible therapeutic target to treat renal fibrosis in diabetes.-Al-Obaidi, N., Mohan, S., Liang, S., Zhao, Z., Nayak, B. K., Li, B., Sriramarao, P., Habib, S. L. Galectin-1 is a new fibrosis protein in type 1 and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Fibrosis/metabolismo , Galectina 1/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Animales , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Fibronectinas/metabolismo , Fibrosis/etiología , Fibrosis/patología , Glucosa/administración & dosificación , Células HEK293 , Humanos , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Regiones Promotoras Genéticas
4.
Carcinogenesis ; 40(6): 782-790, 2019 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-30508035

RESUMEN

Kidneys are one of the main dose-limiting organs in radiotherapeutic procedures of lower abdomen. Likewise, the threat of exposure of radiosensitive organs such as kidneys in warfare or radiation accidents among military personal or due to terrorist activities in general public is of increasing concern. These events warrant the need for appropriate animal models to study the acute and chronic effects of low- and high-dose rate radiation exposures. In this study, for the first time, we validated Tsc2+/- mouse model to study whether radiation accelerates carcinogenesis in kidneys. Tsc2+/- mice at increasing age groups at 8 and 10 months were exposed to repeated doses of gamma radiation (0.4 Gy × 5) and assessed for aggravated kidney tumor formation at 2 months post-irradiation. Animals from irradiated group showed a significant increase in numbers of bilateral, multifocal tumors compared with mock-irradiated animals. Intra-glomerular reactive oxygen species (ROS) levels measured by dihydroethidium florescence showed significant increases in ROS production in irradiated Tsc2+/- mice compared with non-irradiated animals. Similarly, selective hematological parameters and glomerular filtration rate were further reduced significantly in irradiated Tsc2+/- mice. Tsc2 protein, tuberin in irradiated mice, however, remains at the same reduced levels as that of the mock-irradiated heterozygous Tsc2 mice. The results indicate that radiation alters kidney homeostatic function and influences high spontaneous incidence of renal cell carcinoma in this rodent model. Repurposing of Tsc2+/- mice model will, therefore, provide a unique opportunity to study acute and delayed effects of radiation in the development of kidney cancers.


Asunto(s)
Neoplasias Renales/radioterapia , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Glomérulos Renales/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/patología , Ratones , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética
5.
Postepy Dermatol Alergol ; 34(6): 526-534, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29422816

RESUMEN

The aim of this paper was to collect currently available data related to the use of stem cells in aesthetic dermatology and plastic surgery based on a systemic review of experimental and clinical applications. We found that the use of stem cells is very promising but the current state of art is still not effective. This situation is connected with not fully known mechanisms of cell interactions, possible risks and side effects. We think that there is a big need to create and conduct different studies which could resolve problems of stem cells use for implementation into aesthetic dermatology and plastic surgery.

6.
Cancer Sci ; 107(8): 1092-100, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27278252

RESUMEN

Reactive oxygen species (ROS) are an important endogenous source of DNA damage and oxidative stress in all cell types. Deficiency in tuberin resulted in increased oxidative DNA damage in renal cells. In this study, the role of tuberin in the regulating of ROS and NADPH oxidases was investigated. Formation of ROS and activity of NADPH oxidases were significantly higher in mouse embryonic fibroblasts and in primary culture of rat renal proximal tubular epithelial tuberin-deficient cells compared to wild-type cells. In addition, expression of NADPH oxidase (Nox)1, Nox2, and Nox4 (Nox isoforms) was higher in mouse embryonic fibroblasts and renal proximal tubular epithelial tuberin-deficient cells compared to wild-type cells. Furthermore, activity levels of NADPH oxidases and protein expression of all Nox isoforms were higher in the renal cortex of rat deficient in tuberin. However, treatment of tuberin-deficient cells with rapamycin showed significant decrease in protein expression of all Nox. Significant increase in protein kinase C ßII expression was detected in tuberin-deficient cells, whereas inhibition of protein kinase C ßII by bisindolylmaleimide I resulted in decreased protein expression of all Nox isoforms. In addition, treatment of mice deficient in tuberin with rapamycin resulted in significant decrease in all Nox protein expression. Moreover, protein and mRNA expression of all Nox were highly expressed in tumor kidney tissue of patients with tuberous sclerosis complex compared to control kidney tissue of normal subjects. These data provide the first evidence that tuberin plays a novel role in regulating ROS generation, NADPH oxidase activity, and Nox expression that may potentially be involved in development of kidney tumor in patients with tuberous sclerosis complex.


Asunto(s)
Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Esclerosis Tuberosa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Corteza Renal/enzimología , Corteza Renal/metabolismo , Masculino , Ratones , NADPH Oxidasa 1 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia
7.
Am J Physiol Cell Physiol ; 308(8): C673-83, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25652452

RESUMEN

Endothelial nitric oxide (NO) synthase (eNOS) is the predominant isoform that generates NO in the blood vessels. Many different regulators, including heat shock protein 90 (Hsp90), govern eNOS function. Hsp90-dependent phosphorylation of eNOS is a critical event that determines eNOS activity. In our earlier study we demonstrated an inhibitor-κB kinase-ß (IKKß)-Hsp90 interaction in a high-glucose environment. In the present study we further define the putative binding domain of IKKß on Hsp90. Interestingly, IKKß binds to the middle domain of Hsp90, which has been shown to interact with eNOS to stimulate its activity. This new finding suggests a tighter regulation of eNOS activity than was previously assumed. Furthermore, addition of purified recombinant IKKß to the eNOS-Hsp90 complex reduces the eNOS-Hsp90 interaction and eNOS activity, indicating a competition for Hsp90 between eNOS and IKKß. The pathophysiological relevance of the IKKß-Hsp90 interaction has also been demonstrated using in vitro vascular endothelial growth factor-mediated signaling and an Ins2(Akita) in vivo model. Our study further defines the preferential involvement of α- vs. ß-isoforms of Hsp90 in the IKKß-eNOS-Hsp90 interaction, even though both Hsp90α and Hsp90ß stimulate NO production. These studies not only reinforce the significance of maintaining a homeostatic balance of eNOS and IKKß within the cell system that regulates NO production, but they also confirm that the IKKß-Hsp90 interaction is favored in a high-glucose environment, leading to impairment of the eNOS-Hsp90 interaction, which contributes to endothelial dysfunction and vascular complications in diabetes.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Quinasa I-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Aorta/citología , Sitios de Unión , Bovinos , Células Cultivadas , Diabetes Mellitus/patología , Células Endoteliales/enzimología , Glucosa/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Humanos , Quinasa I-kappa B/genética , Insulina/metabolismo , Ratones , Ratones Endogámicos NOD , Óxido Nítrico/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Lab Invest ; 95(10): 1092-104, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26214584

RESUMEN

The balance of nitric oxide (NO) versus superoxide generation has a major role in the initiation and progression of endothelial dysfunction. Under conditions of high glucose, endothelial nitric oxide synthase (eNOS) functions as a chief source of superoxide rather than NO. In order to improve NO bioavailability within the vessel wall in type-1 diabetes, we investigated treatment strategies that improve eNOS phosphorylation and NO-dependent vasorelaxation. We evaluated methods to increase the eNOS activity by (1) feeding Ins2(Akita) spontaneously diabetic (type-1) mice with l-arginine in the presence of sepiapterin, a precursor of tetrahydrobiopterin; (2) preventing eNOS/NO deregulation by the inclusion of inhibitor kappa B kinase beta (IKKß) inhibitor, salsalate, in the diet regimen in combination with l-arginine and sepiapterin; and (3) independently increasing eNOS expression to improve eNOS activity and associated NO production through generating Ins2(Akita) diabetic mice that overexpress human eNOS predominantly in vascular endothelial cells. Our results clearly demonstrated that diet supplementation with l-arginine, sepiapterin along with salsalate improved phosphorylation of eNOS and enhanced vasorelaxation of thoracic/abdominal aorta in type-1 diabetic mice. More interestingly, despite the overexpression of eNOS, the in-house generated transgenic eNOS-GFP (TgeNOS-GFP)-Ins2(Akita) cross mice showed an unanticipated effect of reduced eNOS phosphorylation and enhanced superoxide production. Our results demonstrate that enhancement of endogenous eNOS activity by nutritional modulation is more beneficial than increasing the endogenous expression of eNOS by gene therapy modalities.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Suplementos Dietéticos , Endotelio Vascular/metabolismo , Hipoglucemiantes/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Animales , Aorta/citología , Aorta/metabolismo , Aorta/fisiopatología , Arginina/metabolismo , Arginina/uso terapéutico , Bovinos , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Endotelio Vascular/citología , Endotelio Vascular/fisiopatología , Femenino , Heterocigoto , Humanos , Hipoglucemiantes/metabolismo , Insulina/genética , Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/química , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Inhibidores de Proteínas Quinasas/metabolismo , Pterinas/metabolismo , Pterinas/uso terapéutico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Salicilatos/metabolismo , Salicilatos/uso terapéutico , Destete
9.
EMBO J ; 29(17): 2994-3006, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20657550

RESUMEN

p53 mediates DNA damage-induced cell-cycle arrest, apoptosis, or senescence, and it is controlled by Mdm2, which mainly ubiquitinates p53 in the nucleus and promotes p53 nuclear export and degradation. By searching for the kinases responsible for Mdm2 S163 phosphorylation under genotoxic stress, we identified S6K1 as a multifaceted regulator of Mdm2. DNA damage activates mTOR-S6K1 through p38alpha MAPK. The activated S6K1 forms a tighter complex with Mdm2, inhibits Mdm2-mediated p53 ubiquitination, and promotes p53 induction, in addition to phosphorylating Mdm2 on S163. Deactivation of mTOR-S6K1 signalling leads to Mdm2 nuclear translocation, which is facilitated by S163 phosphorylation, a reduction in p53 induction, and an alteration in p53-dependent cell death. These findings thus establish mTOR-S6K1 as a novel regulator of p53 in DNA damage response and likely in tumorigenesis. S6K1-Mdm2 interaction presents a route for cells to incorporate the metabolic/energy cues into DNA damage response and links the aging-controlling Mdm2-p53 and mTOR-S6K pathways.


Asunto(s)
Ciclo Celular , Daño del ADN , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Estrés Fisiológico , Línea Celular , Reparación del ADN , Dimerización , Humanos , Fosforilación , Unión Proteica , Serina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
10.
J Artif Organs ; 17(2): 123-34, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24748421

RESUMEN

The availability of kidney and other organs from matching donors is not enough for many patients on demand for organ transplant. Unfortunately, this situation is not better despite the many of new interesting projects of promoting family, cross or domino transplants. These inexorable global statistics forced medical researchers to find a new potential therapeutic option that would guarantee safety and efficacy for the treatment of ESRD comparable to kidney transplantation. The aim of our review is to summarize the scientific literature that relating to the modern as well as innovative experimental methods and possibilities of kidney regeneration and, in addition, to find whether the regenerative medicine field will be a new hope for curing the patient with renal disease complications. The most important achievements in the field of regenerative medicine of kidney, which were mentioned and described here, are currently cumulated in 4 areas of interest: stem cell-based therapies, neo-kidneys with specially designed scaffolds or cell-seeded matrices, bioartificial kidneys and innovative nanotechnologically bioengineered solutions. Nowadays, we can add some remarks that the regenerative medicine is still insufficient to completely replace current therapy methods used in patients with chronic kidney disease especially with the end-stage renal disease where in many cases kidney transplantation is the only one chance. But we think that development of regenerative medicine especially in the last 20 years brings us more and more closer to solve many of today's problems at the frontier of nephrology and transplantology.


Asunto(s)
Fallo Renal Crónico/terapia , Medicina Regenerativa/tendencias , Órganos Bioartificiales , Humanos , Trasplante de Riñón , Trasplante de Células Madre , Ingeniería de Tejidos
11.
Mol Cancer ; 12: 49, 2013 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-23705901

RESUMEN

BACKGROUND: Deficiency in tuberin results in activation the mTOR pathway and leads to accumulation of cell matrix proteins. The mechanisms by which tuberin regulates fibrosis in kidney angiomyolipomas (AMLs) of tuberous sclerosis patients are not fully known. METHOD: In the present study, we investigated the potential role of tuberin/mTOR pathway in the regulation of cell fibrosis in AML cells and kidney tumor tissue from tuberous sclerosis complex (TSC) patients. RESULTS: AML cells treated with rapamycin shows a significant decrease in mRNA and protein expression as well as in promoter transcriptional activity of alpha-smooth muscle actin (α-SMA) compared to untreated cells. In addition, cells treated with rapamycin significantly decreased the protein expression of the transcription factor YY1. Rapamycin treatment also results in the redistribution of YY1 from the nucleus to cytoplasm in AML cells. Moreover, cells treated with rapamycin resulted in a significant reduce of binding of YY1 to the αSMA promoter element in nuclear extracts of AML cells. Kidney angiomyolipoma tissues from TSC patients showed lower levels of tuberin and higher levels of phospho-p70S6K that resulted in higher levels of mRNA and protein of αSMA expression compared to control kidney tissues. In addition, most of the α-SMA staining was identified in the smooth muscle cells of AML tissues. YY1 was also significantly increased in tumor tissue of AMLs compared to control kidney tissue suggesting that YY1 plays a major role in the regulation of αSMA. CONCLUSIONS: These data comprise the first report to provide one mechanism whereby rapamycin might inhibit the cell fibrosis in kidney tumor of TSC patients.


Asunto(s)
Neoplasias Renales/complicaciones , Neoplasias Renales/patología , Esclerosis Tuberosa/complicaciones , Actinas/genética , Actinas/metabolismo , Línea Celular , Activación Enzimática , Fibrosis , Regulación de la Expresión Génica , Humanos , Espacio Intracelular/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Modelos Biológicos , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
12.
J Am Soc Nephrol ; 23(10): 1652-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22904348

RESUMEN

Exposure of proximal tubular epithelial cells to high glucose contributes to the accumulation of tubulointerstitial and matrix proteins in diabetic nephropathy, but how this occurs is not well understood. We investigated the effect of the signaling molecule tuberin, which modulates the mammalian target of rapamycin pathway, on renal hypertrophy and fibronectin expression. We found that the kidney mass was significantly greater in partially tuberin-deficient (TSC2(+/-) ) diabetic rats than wild-type diabetic rats. Furthermore, TSC2(+/-) rats exhibited significant increases in the basal levels of phospho-tuberin and fibronectin expression in the kidney cortex. Increased levels of phosphorylated tuberin associated with an increase in fibronectin expression in both wild-type and TSC2(+/-) diabetic rats. Treatment with insulin abrogated the diabetes-induced increase in fibronectin expression. In vitro, high glucose enhanced fibronectin expression in TSC2(+/-) primary proximal tubular epithelial cells; both inhibition of Akt and inhibition of the mammalian target of rapamycin could prevent this effect of glucose. In addition, forced expression of tuberin in tuberin-null cells abolished the expression of fibronectin protein. Taken together, these data suggest that tuberin plays a central role in the development of renal hypertrophy and in modulating the production of the matrix protein fibronectin in diabetes.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Fibronectinas/biosíntesis , Proteínas Supresoras de Tumor/deficiencia , Animales , Secuencia de Bases , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/patología , Fibronectinas/genética , Expresión Génica/efectos de los fármacos , Marcación de Gen , Glucosa/metabolismo , Glucosa/farmacología , Hipertrofia , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Modelos Biológicos , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
13.
J Am Soc Nephrol ; 22(2): 262-73, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21289215

RESUMEN

Apoptosis contributes to the development of diabetic nephropathy, but the mechanism by which high glucose (HG) induces apoptosis is not fully understood. Because the tuberin/mTOR pathway can modulate apoptosis, we studied the role of this pathway in apoptosis in type I diabetes and in cultured proximal tubular epithelial (PTE) cells exposed to HG. Compared with control rats, diabetic rats had more apoptotic cells in the kidney cortex. Induction of diabetes also increased phosphorylation of tuberin in association with mTOR activation (measured by p70S6K phosphorylation), inactivation of Bcl-2, increased cytosolic cytochrome c expression, activation of caspase 3, and cleavage of PARP; insulin treatment prevented these changes. In vitro, exposure of PTE cells to HG increased phosphorylation of tuberin and p70S6K, phosphorylation of Bcl-2, expression of cytosolic cytochrome c, and caspase 3 activity. High glucose induced translocation of the caspase substrate YY1 from the cytoplasm to the nucleus and enhanced cleavage of PARP. Pretreatment the cells with the mTOR inhibitor rapamycin reduced the number of apoptotic cells induced by HG and the downstream effects of mTOR activation noted above. Furthermore, gene silencing of tuberin with siRNA decreased cleavage of PARP. These data show that the tuberin/mTOR pathway promotes apoptosis of tubular epithelial cells in diabetes, mediated in part by cleavage of PARP by YY1.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Túbulos Renales Proximales/patología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis , Células Epiteliales/patología , Masculino , Fosforilación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Ratas Long-Evans , Estreptozocina , Proteína 2 del Complejo de la Esclerosis Tuberosa , Factor de Transcripción YY1/metabolismo
15.
Biomedicines ; 10(3)2022 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-35327359

RESUMEN

Innate immunity is critical for immediate recognition and elimination of invading pathogens or defense against cancer cell growth. Dysregulation of innate immune systems is associated with the pathogenesis of different types of inflammatory diseases, including cancer. In addition, the maintenance of innate immune cells' genomic integrity is crucial for the survival of all organisms. Oxidative stress generated from innate immune cells may cause self-inflicted DNA base lesions as well as DNA damage on others neighboring cells, including cancer cells. Oxidative DNA base damage is predominantly repaired by base excision repair (BER). BER process different types of DNA base lesions that are presented in cancer and innate immune cells to maintain genomic integrity. However, mutations in BER genes lead to impaired DNA repair function and cause insufficient genomic integrity. Moreover, several studies have implicated that accumulation of DNA damage leads to chromosomal instability that likely activates the innate immune signaling. Furthermore, dysregulation of BER factors in cancer cells modulate the infiltration of innate immune cells to the tumor microenvironment. In the current review, the role of BER in cancer and innate immune cells and its impact on innate immune signaling within the tumor microenvironment is summarized. This is a special issue that focuses on DNA damage and cancer therapy to demonstrate how BER inhibitor or aberrant repair modulates innate inflammatory response and impact immunotherapy approaches. Overall, the review provides substantial evidence to understand the impact of BER in innate immune response dynamics within the current immune-based therapeutic strategy.

16.
Hum Hered ; 70(2): 97-101, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20606456

RESUMEN

OBJECTIVE: Human 8-oxoguanine glycosylase 1 (OGG1) excises oxidatively damaged promutagenic base 8-oxoguanine, a lesion previously observed in a rat model of type 2 diabetes (T2DM). The objective of the present study is to determine whether genetic variation in OGG1 is associated with type 2 diabetes (T2DM) in a Mexican American cohort. METHODS: Ten SNPs including two tagging SNPs (rs1052133, rs2072668) across the OGG1 gene region were selected from the Hapmap database and genotyped in the entire cohort (n = 670; 29% diabetes; 39 families) by TaqMan assay. Association analyses between the SNPs and T2DM were performed using the measured genotype approach as implemented in the program SOLAR. RESULTS: Of the ten SNPs genotyped, only five were polymorphic. The minor allele frequencies of these 5 SNPs ranged from 1-38%. Of the SNPs examined for association, the Ser(326)Cys (rs1052133) exhibited significant association with T2DM (p = 0.016) after accounting for age and sex effects. Another intronic variant (rs2072668), which was in strong linkage disequilibrium (r(2) = 0.96) with Ser(326)Cys also exhibited significant association with T2DM (p = 0.031). CONCLUSIONS: These results suggest for the first time that the variants in OGG1 could influence diabetes risk in these Mexican American families and support a role for alterations of OGG1 in the pathogenesis of T2DM.


Asunto(s)
ADN Glicosilasas/genética , Diabetes Mellitus Tipo 2/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Americanos Mexicanos/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Femenino , Humanos , Desequilibrio de Ligamiento/genética , Masculino , Texas
17.
FASEB Bioadv ; 3(9): 730-743, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34485841

RESUMEN

Epithelial-mesenchymal transition (EMT) plays an important role in tissue fibrosis following chronic exposure to hyperglycemia. This study investigates the role of chronic diabetes in regulating tuberin/snail/AMPK to enhance EMT and increase renal fibrosis. A new mouse model of db/db/TSC2 +/- was generated by backcrossing db/db mice and TSC2 +/- mice. Wild type (WT), db/db, TSC2 +/- and dbdb/TSC2 +/- mice were sacrificed at ages 6 and 8 months old. Tuberin protein level was significantly decreased in kidneys from diabetic compared to WT mice at both ages. In addition, tuberin and E-cadherin protein levels were significantly decreased in dbdb/TSC2 +/- compared to TSC2 +/- and db/db mice. In contrast, p-PS6K, NFkB, snail, vimentin, fibronectin, and α-SMA protein levels were significantly increased in dbdb/TSC2 +/- compared to db/db and TSC2 +/- mice at ages 6 and 8 months. Both downregulation of AMPK by DN-AMPK and downregulation of tuberin by siRNA resulted in increased NFkB, snail, and fibronectin protein expression and decreased E-cadherin protein expression in mouse primary renal proximal tubular cells. Interestingly, downregulation of snail by siRNA increased tuberin expression via feedback through activation of AMPK and reversed the expression of epithelial proteins such as E-cadherin as well as mesenchymal proteins such as fibronectin, NF-KB, vimentin, and α-SMA in mouse primary renal proximal tubular cells isolated from kidneys of four mice genotypes. The data show that chronic diabetes significantly decreases tuberin expression and that provides strong evidence that tuberin is a major key protein involved in regulating EMT. These data also demonstrated a novel role for snail in regulating of AMPK/tuberin to enhance EMT and renal cell fibrosis in diabetes.

18.
Cell Rep ; 35(5): 109069, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33951440

RESUMEN

mTOR, the sensor of nutrients and growth factors, has important roles in tissue homeostasis and tumorigenesis. However, how mTOR controls gastric epithelial cell turnover and gastric cancer development, a leading malignancy, remains poorly understood. Here, we provide genetic evidence that mTOR activation promotes proliferation and inhibits differentiation of Lgr5+ gastric epithelial progenitors (GEPs) in gastric homeostasis and tumorigenesis. mTOR signaling increases MEK1 and Smad1 expression and enhances activation of MEK1-ERKs and BMP-Smad1 pathways, respectively, in GEPs and gastric tumors. Mek1 deletion or inhibition rescues hyperproliferation, whereas Bmpr1a ablation or inhibition rescues differentiation defects of Tsc1-/- GEPs. Tsc1 deficiency in Lgr5+ GEPs accelerates gastric tumor initiation and development, which require MEK1-ERKs for hyperplasia and BMP-Smad1 for differentiation suppression. These findings reveal how mTOR signaling controls Lgr5+ GEP homeostasis and cancerization and suggest that ERKs and Smad1 signaling can be safely targeted to substitute mTOR inhibitors in gastric cancer therapy.


Asunto(s)
Células Epiteliales/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Neoplasias Gástricas/genética , Serina-Treonina Quinasas TOR/metabolismo , Animales , Carcinogénesis , Proliferación Celular , Homeostasis , Humanos , Ratones , Transducción de Señal , Neoplasias Gástricas/patología
19.
Cancer Res ; 81(12): 3174-3186, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33863779

RESUMEN

Renal cell carcinoma (RCC) mainly originates from renal proximal tubules. Intriguingly, disruption of genes frequently mutated in human RCC samples thus far has only generated RCC originated from other renal tubule parts in mouse models. This hampers our understanding of the pathogenesis of RCC. Here we show that mTOR signaling, often activated in RCC samples, initiates RCC development from renal proximal tubules. Ablation of Tsc1, encoding an mTOR suppressor, in proximal tubule cells led to multiple precancerous renal cysts. mTOR activation increased MEK1 expression and ERK activation, and Mek1 ablation or inhibition diminished cyst formation in Tsc1-deficient mice. mTOR activation also increased MKK6 expression and p38MAPK activation, and ablation of the p38α-encoding gene further enhanced cyst formation and led to RCC with clear cell RCC features. Mechanistically, Tsc1 deletion induced p53 and p16 expression in a p38MAPK-dependent manner, and deleting Tsc1 and Trp53 or Cdkn2a (encoding p16) enhanced renal cell carcinogenesis. Thus, mTOR activation in combination with inactivation of the p38MAPK-p53/p16 pathway drives RCC development from renal proximal tubules. Moreover, this study uncovers previously unidentified mechanisms by which mTOR controls cell proliferation and suggests the MEK-ERK axis to be a potential target for treatment of RCC. SIGNIFICANCE: Mouse modeling studies show that mTOR activation in combination with inactivation of the p38MAPK-p53/p16 axis initiates renal cell carcinoma that mimics human disease, identifying potential therapeutic targets for RCC treatment.


Asunto(s)
Carcinoma de Células Renales/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , MAP Quinasa Quinasa 1/fisiología , Proteína Quinasa 14 Activada por Mitógenos/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Apoptosis , Carcinoma de Células Renales/etiología , Carcinoma de Células Renales/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/etiología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Serina-Treonina Quinasas TOR/genética , Células Tumorales Cultivadas
20.
Carcinogenesis ; 31(11): 2022-30, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20837600

RESUMEN

Tuberin (protein encodes by tuberous sclerosis complex 2, Tsc2) deficiency is associated with the decrease in the DNA repair enzyme 8-oxoG-DNA glycosylase (OGG1) in tumour kidney of tuberous sclerosis complex (TSC) patients. The purpose of this study was to elucidate the mechanisms by which tuberin regulates OGG1. The partial deficiency in tuberin expression that occurs in the renal proximal tubular cells and kidney cortex of the Eker rat is associated with decreased activator protein 4 (AP4) and OGG1 expression. A complete deficiency in tuberin is associated with loss of AP4 and OGG1 expression in kidney tumour from Eker rats and the accumulation of significant levels of 8-oxo-deoxyguanosine. Knockdown of tuberin expression in human renal epithelial cells (HEK293) with small interfering RNA (siRNA) also resulted in a marked decrease in the expression of AP4 and OGG1. In contrast, overexpression of tuberin in HEK293 cells increased the expression of AP4 and OGG1 proteins. Downregulation of AP4 expression using siRNA resulted in a significant decrease in the protein expression of OGG1. Immunoprecipitation studies show that AP4 is associated with tuberin in cells. Gel shift analysis and chromatin immunoprecipitation identified the transcription factor AP4 as a positive regulator of the OGG1 promoter. AP4 DNA-binding activity is significantly reduced in Tsc2(-/-) as compared with Tsc2(+/+) cells. Transcriptional activity of the OGG1 promoter is also decreased in tuberin-null cells compared with wild-type cells. These data indicate a novel role for tuberin in the regulation of OGG1 through the transcription factor AP4. This regulation may be important in the pathogenesis of kidney tumours in patients with TSC disease.


Asunto(s)
ADN Glicosilasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Túbulos Renales Proximales/enzimología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/fisiología , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Reparación del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Técnicas para Inmunoenzimas , Riñón/citología , Riñón/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Túbulos Renales Proximales/citología , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Ratas , Ratas Long-Evans , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 2 del Complejo de la Esclerosis Tuberosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA