Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(6): 1754-1766, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31901078

RESUMEN

Ten-eleven translocation-2 (TET2) is a member of the methylcytosine dioxygenase family of enzymes and has been implicated in cancer and aging because of its role as a global epigenetic modifier. TET2 has a large N-terminal domain and a catalytic C-terminal region. Previous reports have demonstrated that the TET2 catalytic domain remains active independently of the N-terminal domain. As such, the function of the N terminus of this large protein remains poorly characterized. Here, using yeast two-hybrid screening, co-immunoprecipitation, and several biochemical assays, we found that several isoforms of the 14-3-3 family of proteins bind TET2. 14-3-3 proteins bound TET2 when it was phosphorylated at Ser-99. In particular, we observed that AMP-activated protein kinase-mediated phosphorylation at Ser-99 promotes TET2 stability and increases global DNA 5-hydroxymethylcytosine levels. The interaction of 14-3-3 proteins with TET2 protected the Ser-99 phosphorylation, and disruption of this interaction both reduced TET2 phosphorylation and decreased TET2 stability. Furthermore, we noted that protein phosphatase 2A can interact with TET2 and dephosphorylate Ser-99. Collectively, these results provide detailed insights into the role of the TET2 N-terminal domain in TET2 regulation. Moreover, they reveal the dynamic nature of TET2 protein regulation that could have therapeutic implications for disease states resulting from reduced TET2 levels or activity.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Dioxigenasas , Células HEK293 , Humanos , Ratones , Fosforilación , Unión Proteica , Isoformas de Proteínas/metabolismo
2.
Mol Cell ; 51(2): 236-48, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23747014

RESUMEN

The tricarboxylic acid cycle enzyme fumarate hydratase (FH) has been identified as a tumor suppressor in a subset of human renal cell carcinomas. Human FH-deficient cancer cells display high fumarate concentration and ROS levels along with activation of HIF-1. The underlying mechanisms by which FH loss increases ROS and HIF-1 are not fully understood. Here, we report that glutamine-dependent oxidative citric acid cycle metabolism is required to generate fumarate and increase ROS and HIF-1 levels. Accumulated fumarate directly bonds the antioxidant glutathione in vitro and in vivo to produce the metabolite succinated glutathione (GSF). GSF acts as an alternative substrate to glutathione reductase to decrease NADPH levels and enhance mitochondrial ROS and HIF-1 activation. Increased ROS also correlates with hypermethylation of histones in these cells. Thus, fumarate serves as a proto-oncometabolite by binding to glutathione which results in the accumulation of ROS.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Fumaratos/metabolismo , Glutatión/metabolismo , Neoplasias Renales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Carcinoma de Células Renales/patología , Cromatografía Liquida , Fumarato Hidratasa/antagonistas & inhibidores , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Glutatión Reductasa/metabolismo , Histona Demetilasas/metabolismo , Histonas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Immunoblotting , Neoplasias Renales/patología , NADP/metabolismo , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Consumo de Oxígeno , ARN Interferente Pequeño/genética , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Células Tumorales Cultivadas
3.
Prostate ; 78(16): 1311-1320, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30051493

RESUMEN

Prostate cancer (PCa) is one of the most frequently diagnosed cancers among men. Many molecular changes have been detailed during PCa progression. The gene encoding the transcription factor ERG shows recurrent rearrangement, resulting in the overexpression of ERG in the majority of prostate cancers. Overexpression of ERG plays a critical role in prostate oncogenesis and development of metastatic disease. Among the downstream effectors of ERG, Frizzled family member FZD4 has been shown to be a target of ERG. Frizzled-8 (FZD8) has been shown to be involved in PCa bone metastasis. In the present study, we show that the expression of FZD8 is directly correlated with ERG expression in PCa. Furthermore, we show that ERG directly targets and activates FZD8 by binding to its promoter. This activation is specific to ETS transcription factor ERG and not ETV1. We propose that ERG overexpression in PCa leads to induction of Frizzled family member FZD8, which is known to activate the Wnt pathway. Taken together, these findings uncover a novel mechanism for PCa metastasis, and indicate that FZD8 may represent a potential therapeutic target for PCa.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Próstata/metabolismo , Neoplasias de la Próstata/genética , Receptores de Superficie Celular/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Masculino , Regiones Promotoras Genéticas , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores de Superficie Celular/metabolismo , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
4.
J Biol Chem ; 289(35): 24691-9, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25028521

RESUMEN

Inactivating mutations of the gene encoding the tricarboxylic acid cycle enzyme fumarate hydratase (FH) have been linked to an aggressive variant of hereditary kidney cancer (hereditary leiomyomatosis and renal cell cancer). These tumors accumulate markedly elevated levels of fumarate. Fumarate is among a growing list of oncometabolites identified in cancers with mutations of genes involved in intermediary metabolism. FH-deficient tumors are notable for their pronounced accumulation of the transcription factor hypoxia inducible factor-1α (HIF-1α) and aggressive behavior. To date, HIF-1α accumulation in hereditary leiomyomatosis and renal cell cancer tumors is thought to result from fumarate-dependent inhibition of prolyl hydroxylases and subsequent evasion from von Hippel-Lindau-dependent degradation. Here, we demonstrate a novel mechanism by which fumarate promotes HIF-1α mRNA and protein accumulation independent of the von Hippel-Lindau pathway. Here we demonstrate that fumarate promotes p65 phosphorylation and p65 accumulation at the HIF-1α promoter through non-canonical signaling via the upstream Tank binding kinase 1 (TBK1). Consistent with these data, inhibition of the TBK1/p65 axis blocks HIF-1α accumulation in cellular models of FH loss and markedly reduces cell invasion of FH-deficient RCC cancer cells. Collectively, our data demonstrate a novel mechanism by which pseudohypoxia is promoted in FH-deficient tumors and identifies TBK1 as a novel putative therapeutic target for the treatment of aggressive fumarate-driven tumors.


Asunto(s)
Hipoxia de la Célula , Fumaratos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Secuencia de Bases , Línea Celular , Inmunoprecipitación de Cromatina , Cartilla de ADN , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosforilación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
bioRxiv ; 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38352483

RESUMEN

Pseudouridylation is a prevalent post-transcriptional RNA modification that impacts many aspects of RNA biology and function. The conversion of uridine to pseudouridine (Ψ) is catalyzed by the family of pseudouridine synthases (PUSs). Development of robust methods to determine PUS-dependent regulation of Ψ location and stoichiometry in low abundant mRNA is essential for biological and functional understanding of pseudouridylation. Here, we present a framework, NanoPsiPy, for identifying Ψ sites and quantify their levels in poly-A RNA at single-nucleotide resolution using direct RNA long-read Nanopore sequencing, based on the observation that Ψ can cause characteristic U-to-C basecalling errors in Nanopore direct RNA sequencing data. Our method was able to detect low and high stoichiometric Ψ sites in human mRNA. We validated our method by transcriptome-wide quantitative profiling of PUS7-dependent Ψ sites in poly-A RNA from a MYCN -amplified neuroblastoma cell line. We identified 8,625 PUS7-dependent Ψ sites in 1,246 mRNAs that encode proteins involved primarily in ribosome biogenesis, translation, and mitochondrial energy metabolism. Our work provides the first example of using direct RNA long-read Nanopore sequencing for transcriptome-wide quantitative profiling of mRNA pseudouridylation regulated by a PUS. We envision that our method will facilitate functional interrogation of PUSs in biological and pathological processes.

6.
J Clin Invest ; 134(13)2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38743486

RESUMEN

Tumor cells are known to undergo considerable metabolic reprogramming to meet their unique demands and drive tumor growth. At the same time, this reprogramming may come at a cost with resultant metabolic vulnerabilities. The small molecule l-2-hydroxyglutarate (l-2HG) is elevated in the most common histology of renal cancer. Similarly to other oncometabolites, l-2HG has the potential to profoundly impact gene expression. Here, we demonstrate that l-2HG remodels amino acid metabolism in renal cancer cells through combined effects on histone methylation and RNA N6-methyladenosine. The combined effects of l-2HG result in a metabolic liability that renders tumors cells reliant on exogenous serine to support proliferation, redox homeostasis, and tumor growth. In concert with these data, high-l-2HG kidney cancers demonstrate reduced expression of multiple serine biosynthetic enzymes. Collectively, our data indicate that high-l-2HG renal tumors could be specifically targeted by strategies that limit serine availability to tumors.


Asunto(s)
Glutaratos , Neoplasias Renales , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Glutaratos/metabolismo , Humanos , Animales , Ratones , Línea Celular Tumoral , Serina/metabolismo , Epigenoma , Transcriptoma , Histonas/metabolismo , Histonas/genética , Regulación Neoplásica de la Expresión Génica , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Epigénesis Genética , Adenosina/análogos & derivados
7.
Cancer Res ; 83(13): 2248-2261, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37057874

RESUMEN

High-risk neuroblastoma exhibits transcriptional activation of the mevalonate pathway that produces cholesterol and nonsterol isoprenoids. A better understanding of how this metabolic reprogramming contributes to neuroblastoma development could help identify potential prevention and treatment strategies. Here, we report that both the cholesterol and nonsterol geranylgeranyl-pyrophosphate branches of the mevalonate pathway are critical to sustain neuroblastoma cell growth. Blocking the mevalonate pathway by simvastatin, a cholesterol-lowering drug, impeded neuroblastoma growth in neuroblastoma cell line xenograft, patient-derived xenograft (PDX), and TH-MYCN transgenic mouse models. Transcriptional profiling revealed that the mevalonate pathway was required to maintain the FOXM1-mediated transcriptional program that drives mitosis. High FOXM1 expression contributed to statin resistance and led to a therapeutic vulnerability to the combination of simvastatin and FOXM1 inhibition. Furthermore, caffeine synergized with simvastatin to inhibit the growth of neuroblastoma cells and PDX tumors by blocking statin-induced feedback activation of the mevalonate pathway. This function of caffeine depended on its activity as an adenosine receptor antagonist, and the A2A adenosine receptor antagonist istradefylline, an add-on drug for Parkinson's disease, could recapitulate the synergistic effect of caffeine with simvastatin. This study reveals that the FOXM1-mediated mitotic program is a molecular statin target in cancer and identifies classes of agents for maximizing the therapeutic efficacy of statins, with implications for treatment of high-risk neuroblastoma. SIGNIFICANCE: Caffeine treatment and FOXM1 inhibition can both enhance the antitumor effect of statins by blocking the molecular and metabolic processes that confer statin resistance, indicating potential combination therapeutic strategies for neuroblastoma. See related commentary by Stouth et al., p. 2091.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Neuroblastoma , Ratones , Animales , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Cafeína/farmacología , Ácido Mevalónico/metabolismo , Simvastatina/farmacología , Colesterol , Ratones Transgénicos , Neuroblastoma/tratamiento farmacológico , Antagonistas de Receptores Purinérgicos P1 , Suplementos Dietéticos , Proteína Forkhead Box M1/genética
8.
Urol Oncol ; 41(8): 358.e17-358.e24, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37301680

RESUMEN

INTRODUCTION: Despite modern advances in surgical and perioperative technologies, management of renal cell carcinoma (RCC) with tumor thrombus (TT) is a morbid procedure that necessitates careful patient selection. It is not known whether established prognostic models for metastatic RCC are suitable prognostic tools for more immediate perioperative outcomes in patients with RCC with TT. We evaluated if established risk models for cytoreductive nephrectomy, as a potential extension of their purpose-built use, are associated with immediate perioperative outcomes in patients undergoing nephrectomy and tumor thrombectomy. METHODS: Perioperative outcomes of patients who underwent radical nephrectomy and tumor thrombectomy for RCC were compared to presences of established predictors of long-term outcomes from prior risk models individually and as stratified by risk grouping (International Metastatic Renal-Cell Carcinoma Database Consortium [IMDC], Memorial Sloan Kettering Cancer Center [MSKCC], M.D. Anderson Cancer Center [MDACC], and Moffitt Cancer Center [MCC]). Wilcoxon rank-sum test or the Kruskal-Wallis test compared continuous variables and the chi-square test or Fisher's exact test compared categorical variables. RESULTS: Fifty-five patients were analyzed with 17 (30.9%) being cytoreductive. Eighteen (32.7%) patients had a level III or higher TT. Individually, preoperative variables were inconsistently associated with perioperative outcomes. Poorer risk patients per the IMDC model had more major postoperative complications (Clavien-Dindo grade≥3, P = 0.008). For the MSKCC model, poorer risk patients had increased intraoperative estimated blood loss (EBL), longer length of stay (LOS), more major postoperative complications, and more likely to discharge to a rehabilitation facility (P < 0.05). Less favorable risk patients per MDACC model had increased LOS (P = 0.038). Poorer risk patients per the MCC model had increased EBL, LOS, major postoperative complications, and 30-day hospital readmissions (P < 0.05). CONCLUSION: Overall, cytoreductive risks models were heterogeneously associated with perioperative outcomes in patients undergoing nephrectomy and tumor thrombectomy. Of available models, the MCC model is associated with more perioperative outcomes including EBL, LOS, major postoperative complications, and readmissions within 30 days when compared to the IMDC, MSKCC, and MDACC models.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Trombosis , Humanos , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Pronóstico , Trombectomía/métodos , Trombosis/cirugía , Nefrectomía/métodos , Complicaciones Posoperatorias/cirugía , Estudios Retrospectivos , Vena Cava Inferior/patología
9.
Front Endocrinol (Lausanne) ; 13: 932286, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36133305

RESUMEN

2-Hydroxyglutarate (2HG) overproducing tumors arise in a number of tissues, including the kidney. The tumorigenesis resulting from overproduced 2HG has been attributed to the ability of 2HG alter gene expression by inhibiting α-ketoglutarate (αKG)-dependent dioxygenases, including Ten-eleven-Translocation (TET) enzymes. Genes that regulate cellular differentiation are reportedly repressed, blocking differentiation of mesenchymal cells into myocytes, and adipocytes. In this report, the expression of the enzyme responsible for L2HG degradation, L-2HG dehydrogenase (L2HGDH), is knocked down, using lentiviral shRNA, as well as siRNA, in primary cultures of normal Renal Proximal Tubule (RPT) cells. The knockdown (KD) results in increased L-2HG levels, decreased demethylation of 5mC in genomic DNA, and increased methylation of H3 Histones. Consequences include reduced tubulogenesis by RPT cells in matrigel, and reduced expression of molecular markers of differentiation, including membrane transporters as well as HNF1α and HNF1ß, which regulate their transcription. These results are consistent with the hypothesis that oncometabolite 2HG blocks RPT differentiation by altering the methylation status of chromatin in a manner that impedes the transcriptional events required for normal differentiation. Presumably, similar alterations are responsible for promoting the expansion of renal cancer stem-cells, increasing their propensity for malignant transformation.


Asunto(s)
Dioxigenasas , Histonas , Diferenciación Celular/genética , Cromatina , Dioxigenasas/metabolismo , Epigénesis Genética , Glutaratos , Histonas/metabolismo , Ácidos Cetoglutáricos/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Oxidorreductasas/metabolismo , ARN Interferente Pequeño
10.
JCI Insight ; 6(22)2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34609963

RESUMEN

Mounting evidence points to alterations in mitochondrial metabolism in renal cell carcinoma (RCC). However, the mechanisms that regulate the TCA cycle in RCC remain uncharacterized. Here, we demonstrate that loss of TCA cycle enzyme expression is retained in RCC metastatic tissues. Moreover, proteomic analysis demonstrates that reduced TCA cycle enzyme expression is far more pronounced in RCC relative to other tumor types. Loss of TCA cycle enzyme expression is correlated with reduced expression of the transcription factor PGC-1α, which is also lost in RCC tissues. PGC-1α reexpression in RCC cells restores the expression of TCA cycle enzymes in vitro and in vivo and leads to enhanced glucose carbon incorporation into TCA cycle intermediates. Mechanistically, TGF-ß signaling, in concert with histone deacetylase 7 (HDAC7), suppresses TCA cycle enzyme expression. Our studies show that pharmacologic inhibition of TGF-ß restores the expression of TCA cycle enzymes and suppresses tumor growth in an orthotopic model of RCC. Taken together, this investigation reveals a potentially novel role for the TGF-ß/HDAC7 axis in global suppression of TCA cycle enzymes in RCC and provides insight into the molecular basis of altered mitochondrial metabolism in this malignancy.


Asunto(s)
Ciclo del Ácido Cítrico/inmunología , Perfilación de la Expresión Génica/métodos , Histona Desacetilasas/metabolismo , Neoplasias Renales/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Humanos , Ratones , Transfección
11.
J Urol ; 183(5): 1719-24, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20299057

RESUMEN

PURPOSE: We examined outcomes in patients with recurrent or de novo renal lesions treated with repeat partial nephrectomy on a solitary kidney. MATERIALS AND METHODS: We reviewed the records of patients who underwent nephron sparing surgery at the National Cancer Institute from 1989 to 2008. Patients were included in analysis if they underwent repeat partial nephrectomy on a solitary kidney. Perioperative, functional and oncological outcomes were assessed. Functional outcomes were evaluated using the Modification of Diet in Renal Disease equation for the estimated glomerular filtration rate. Oncological efficacy was examined by the need for subsequent repeat renal surgery and the development of metastatic disease. RESULTS: A total of 25 patients were included in the analysis. A median of 4 tumors were resected. Median estimated blood loss was 2,400 ml and median operative time was 8.5 hours. Perioperative complications occurred in 52% of patients, including 1 death and the loss of 3 renal units. There was a decrease in the estimated glomerular filtration rate at followup visit 1 within 3 months after surgery but at 1-year followup the difference was not significant (p <0.01 and 0.12, respectively). Surgical intervention was recommended in 8 patients (38%) for recurrent or de novo tumors at a median of 36 months. The average metastasis-free survival rate in the cohort was 95% at 57 months (median 50, range 3 to 196). CONCLUSIONS: Repeat partial nephrectomy in patients with solitary kidney is a high risk alternative. The complication rate is high and there is a modest decrease in renal function but most patients remain free of dialysis with acceptable oncological outcomes at intermediate followup.


Asunto(s)
Carcinoma de Células Renales/cirugía , Neoplasias Renales/cirugía , Nefrectomía/métodos , Adulto , Anciano , Carcinoma de Células Renales/fisiopatología , Femenino , Tasa de Filtración Glomerular , Humanos , Complicaciones Intraoperatorias , Riñón/fisiopatología , Riñón/cirugía , Pruebas de Función Renal , Neoplasias Renales/fisiopatología , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/cirugía , Complicaciones Posoperatorias , Recuperación de la Función , Reoperación , Tasa de Supervivencia , Resultado del Tratamiento
12.
Urol Ann ; 12(2): 172-175, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32565657

RESUMEN

Prostate cancer is the most common noncutaneous malignancy in American men. Its lymphatic drainage is very well established throughout literature. We report the case of a 72-year-old Caucasian male with elevated serum prostate-specific antigen and biopsy-confirmed high-risk prostate cancer who underwent multiparametric magnetic resonance imaging (MRI) for staging and treatment planning. The imaging revealed suspicious lymph nodes in the left ischiorectal and right obturator fossae that were biopsy confirmed as metastatic prostate adenocarcinoma. Herein, we present the divergence from the well-established lymphatic drainage of prostate cancer and the role of MRI in detecting this prostate cancer site of spread.

13.
J Exp Med ; 217(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32251515

RESUMEN

Analysis of transcriptomic data demonstrates extensive epigenetic gene silencing of the transcription factor PRDM16 in renal cancer. We show that restoration of PRDM16 in RCC cells suppresses in vivo tumor growth. RNaseq analysis reveals that PRDM16 imparts a predominantly repressive effect on the RCC transcriptome including suppression of the gene encoding semaphorin 5B (SEMA5B). SEMA5B is a HIF target gene highly expressed in RCC that promotes in vivo tumor growth. Functional studies demonstrate that PRDM16's repressive properties, mediated by physical interaction with the transcriptional corepressors C-terminal binding proteins (CtBP1/2), are required for suppression of both SEMA5B expression and in vivo tumor growth. Finally, we show that reconstitution of RCC cells with a PRDM16 mutant unable to bind CtBPs nullifies PRDM16's effects on both SEMA5B repression and tumor growth suppression. Collectively, our data uncover a novel epigenetic basis by which HIF target gene expression is amplified in kidney cancer and a new mechanism by which PRDM16 exerts its tumor suppressive effects.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Renales/genética , Factores de Transcripción/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Colforsina/farmacología , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Renales/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Fenotipo , Regiones Promotoras Genéticas/genética , Rosiglitazona/farmacología , Semaforinas/genética , Semaforinas/metabolismo , Transcripción Genética/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Insect Biochem Mol Biol ; 127: 103493, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33157229

RESUMEN

The oncometabolite L-2-hydroxyglutarate (L-2HG) is considered an abnormal product of central carbon metabolism that is capable of disrupting chromatin architecture, mitochondrial metabolism, and cellular differentiation. Under most circumstances, mammalian tissues readily dispose of this compound, as aberrant L-2HG accumulation induces neurometabolic disorders and promotes renal cell carcinomas. Intriguingly, Drosophila melanogaster larvae were recently found to accumulate high L-2HG levels under normal growth conditions, raising the possibility that L-2HG plays a unique role in insect metabolism. Here we explore this hypothesis by analyzing L-2HG levels in 18 insect species. While L-2HG was present at low-to-moderate levels in most of these species (<100 pmol/mg; comparable to mouse liver), dipteran larvae exhibited a tendency to accumulate high L-2HG concentrations (>100 pmol/mg), with the mosquito Aedes aegypti, the blow fly Phormia regina, and three representative Drosophila species harboring concentrations that exceed 1 nmol/mg - levels comparable to those measured in mutant mice that are unable to degrade L-2HG. Overall, our findings suggest that one of the largest groups of animals on earth commonly generate high concentrations of an oncometabolite during juvenile growth, hint at a role for L-2HG in the evolution of dipteran development, and raise the possibility that L-2HG metabolism could be targeted to restrict the growth of key disease vectors and agricultural pests.


Asunto(s)
Aedes/metabolismo , Calliphoridae/metabolismo , Drosophila/metabolismo , Glutaratos/metabolismo , Aedes/crecimiento & desarrollo , Animales , Calliphoridae/crecimiento & desarrollo , Drosophila/crecimiento & desarrollo , Larva/crecimiento & desarrollo , Larva/metabolismo
15.
Dis Model Mech ; 13(11)2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-32928875

RESUMEN

L-2-hydroxyglutarate (L-2HG) is an oncometabolite found elevated in renal tumors. However, this molecule might have physiological roles that extend beyond its association with cancer, as L-2HG levels are elevated in response to hypoxia and during Drosophila larval development. L-2HG is known to be metabolized by L-2HG dehydrogenase (L2HGDH), and loss of L2HGDH leads to elevated L-2HG levels. Despite L2HGDH being highly expressed in the kidney, its role in renal metabolism has not been explored. Here, we report our findings utilizing a novel CRISPR/Cas9 murine knockout model, with a specific focus on the role of L2HGDH in the kidney. Histologically, L2hgdh knockout kidneys have no demonstrable histologic abnormalities. However, GC-MS metabolomics demonstrates significantly reduced levels of the TCA cycle intermediate succinate in multiple tissues. Isotope labeling studies with [U-13C] glucose demonstrate that restoration of L2HGDH in renal cancer cells (which lowers L-2HG) leads to enhanced incorporation of label into TCA cycle intermediates. Subsequent biochemical studies demonstrate that L-2HG can inhibit the TCA cycle enzyme α-ketoglutarate dehydrogenase. Bioinformatic analysis of mRNA expression data from renal tumors demonstrates that L2HGDH is co-expressed with genes encoding TCA cycle enzymes as well as the gene encoding the transcription factor PGC-1α, which is known to regulate mitochondrial metabolism. Restoration of PGC-1α in renal tumor cells results in increased L2HGDH expression with a concomitant reduction in L-2HG levels. Collectively, our analyses provide new insight into the physiological role of L2HGDH as well as mechanisms that promote L-2HG accumulation in disease states.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Riñón/enzimología , Oxidorreductasas de Alcohol/genética , Animales , Encéfalo/enzimología , Encéfalo/patología , Sistemas CRISPR-Cas/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Ciclo del Ácido Cítrico , Fertilidad , Regulación Neoplásica de la Expresión Génica , Glutaratos/metabolismo , Heterocigoto , Riñón/patología , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Análisis de Flujos Metabólicos , Metaboloma , Metabolómica , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Ácido Succínico/metabolismo
16.
Matrix Biol ; 89: 43-58, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31982456

RESUMEN

The transcriptional events that promote invasive and metastatic phenotypes in renal cell carcinoma (RCC) remain poorly understood. Here we report that the decreased expression of peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (PGC1α) and the increased expression of several genes encoding collagen family members are associated with RCC tumor progression. PGC1α restoration attenuates invasive phenotypes and suppresses tumor progression in vivo. In contrast, collagens produced by RCC cells promote invasive and migratory phenotypes. PGC1α restoration suppresses the expression of collagens and tumor phenotypes via the induction of miR-29a. Furthermore, decreased collagens via the PGC1α/miR-29a axis suppresses collagen-mediated activation of discoidin domain receptor 1 (DDR1)/ERK signaling. In turn, the suppression of collagen/DDR1 signaling by PGC1α leads to decreased levels of the known EMT regulators SNAIL1 and 2. Collectively, our results demonstrate a novel role for PGC1α in the regulation of proinvasive SNAIL proteins.


Asunto(s)
Carcinoma de Células Renales/patología , Colágeno/metabolismo , Neoplasias Renales/patología , MicroARNs/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Factores de Transcripción de la Familia Snail/genética , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Ratones , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Estabilidad Proteica , Factores de Transcripción de la Familia Snail/metabolismo
17.
Semin Ultrasound CT MR ; 30(4): 315-25, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19711643

RESUMEN

Recent advances in molecular biology and cytogenetics have provided unique insights into the ontogeny, pathogenesis, and biological behavior of renal cell carcinoma. Renal cell carcinoma is now known to be a polymorphic malignant neoplasm consisting of several histologic subtypes demonstrating different biological profiles. Clear cell renal carcinoma, the most common histologic subtype, is predominantly associated with mutations involving the von Hippel-Lindau gene and elaboration of vascular and somatic growth factors. Clear cell renal cell carcinoma is thus typically hypervascular at imaging. By contrast, papillary renal cell carcinoma, the second most common subtype, is frequently hypovascular. Current molecular data on the biology of renal neoplasms have shown important diagnostic, therapeutic, and prognostic implications. Comprehensive knowledge of molecular pathways of carcinogenesis of renal cancers has allowed design of rational treatment protocols and posttreatment surveillance algorithms, thereby permitting optimal patient management.


Asunto(s)
Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/genética , Neoplasias Renales/diagnóstico , Neoplasias Renales/genética , Imagen por Resonancia Magnética/métodos , Tomografía Computarizada por Rayos X/métodos , Medios de Contraste , Predisposición Genética a la Enfermedad/genética , Humanos , Aumento de la Imagen/métodos , Riñón/diagnóstico por imagen , Riñón/patología
18.
Mol Cancer Res ; 17(1): 84-96, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30131446

RESUMEN

The Cancer Genome Atlas (TCGA) and other large-scale genomic data pipelines have been integral to the current understanding of the molecular events underlying renal cell carcinoma (RCC). These data networks have focused mostly on primary RCC, which often demonstrates indolent behavior. However, metastatic disease is the major cause of mortality associated with RCC and data sets examining metastatic tumors are sparse. Therefore, a more comprehensive analysis of gene expression and DNA methylome profiling of metastatic RCC in addition to primary RCC and normal kidney was performed. Integrative analysis of the methylome and transcriptome identified over 30 RCC-specific genes whose mRNA expression inversely correlated with promoter methylation, including several known targets of hypoxia inducible factors. Notably, genes encoding several metabolism-related proteins were identified as differentially regulated via methylation including hexokinase 2, aldolase C, stearoyl-CoA desaturase, and estrogen-related receptor-γ (ESRRG), which has a known role in the regulation of nuclear-encoded mitochondrial metabolism genes. Several gene expression changes could portend prognosis in the TCGA cohort. Mechanistically, ESRRG loss occurs via DNA methylation and histone repressive silencing mediated by the polycomb repressor complex 2. Restoration of ESRRG in RCC lines suppresses migratory and invasive phenotypes independently of its canonical role in mitochondrial metabolism. IMPLICATIONS: Collectively, these data provide significant insight into the biology of aggressive RCC and demonstrate a novel role for DNA methylation in the promotion of HIF signaling and invasive phenotypes in renal cancer.


Asunto(s)
Epigenómica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Renales/genética , Progresión de la Enfermedad , Humanos , Neoplasias Renales/patología , Metástasis de la Neoplasia
19.
J Urol ; 180(1): 89-93; discussion 93, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18485404

RESUMEN

PURPOSE: Despite the proven efficacy of nephron sparing surgery, patients with hereditary renal cancer remain at risk for tumor recurrence. Management options for recurrent tumors include completion nephrectomy, ablation and repeat partial nephrectomy. We examine the feasibility and outcomes of repeat partial nephrectomy performed on the same renal unit. MATERIALS AND METHODS: We retrospectively reviewed the records of 51 attempted repeat partial nephrectomy procedures in 47 patients from 1992 to 2006. Demographic information as well as intraoperative, perioperative and renal functional outcome data were collected. Comparison of preoperative and postoperative renal function was performed using the 2-tailed t test. RESULTS: Major perioperative complications or reoperations occurred in 10 of 51 (19.6%) cases that included 1 perioperative mortality (1.9%). In cases of successful repeat partial nephrectomy there was a statistically significant increase in postoperative serum creatinine (1.35 vs 1.16 mg/dl, p <0.05), and a significant decrease in creatinine clearance (84.6 vs 95.3 ml per minute, p = 0.05) and renogram split function (52.3% vs 54.8%, p <0.05). Two patients required long-term hemodialysis (3.9%). Of the 51 renal units 10 (19.6%) required subsequent operations for additional local recurrence or de novo tumor formations with a median time to subsequent surgery of 50 months. Of 47 patients 46 are alive at a median followup of 56 months. CONCLUSIONS: Repeat partial nephrectomy is technically feasible. Although there is a statistically significant decrease in postoperative renal functional studies, most patients retained sufficient function to avoid hemodialysis. Repeat partial nephrectomy may provide acceptable oncological control despite the anticipated development of locally recurrent or de novo tumors.


Asunto(s)
Neoplasias Renales/cirugía , Recurrencia Local de Neoplasia/cirugía , Nefrectomía/métodos , Adulto , Anciano , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reoperación , Estudios Retrospectivos , Resultado del Tratamiento
20.
J Urol ; 179(1): 67-70, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17997447

RESUMEN

PURPOSE: Patients with von Hippel-Lindau disease may require repeat partial nephrectomies to avoid the need for renal replacement therapy. We evaluated outcomes in patients who underwent third or fourth partial nephrectomies on the same kidney, which we call salvage partial nephrectomy. MATERIALS AND METHODS: We retrospectively reviewed the charts of patients who underwent surgery at the National Cancer Institute from 1999 to 2006. We identified 11 of 13 patients in whom salvage partial nephrectomy was performed. Surgical outcomes were assessed by renal preservation as well as intraoperative and postoperative complications. Functional outcomes were assessed by comparing serum creatinine, 24-hour urine creatinine clearance, and nuclear renal scan before and after salvage partial nephrectomy. RESULTS: All patients were alive and had no radiographic evidence of metastatic disease at a median followup of 25 months (range 3 to 83). Major perioperative complications occurred in 6 of 13 patients (46%) and loss of a renal unit occurred in 3 (23%). None of the patients in whom renal units were preserved required dialysis, although there was an upward trend in serum creatinine and a downward trend in creatinine clearance at postoperative followup (p = 0.07 and 0.08, respectively). CONCLUSIONS: Although salvage partial nephrectomy is technically demanding and it has a high complication rate, it allows many patients to avoid dialysis. We believe that this experience can be used as a reference for surgeons and patients when considering the risks and benefits of salvage partial nephrectomy.


Asunto(s)
Neoplasias Renales/genética , Neoplasias Renales/cirugía , Nefrectomía/métodos , Enfermedad de von Hippel-Lindau/cirugía , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Terapia Recuperativa , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA