Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(24)2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34948109

RESUMEN

The innate and adaptive immunities have been documented to participate in the pathogenesis of nephrotoxic acute kidney injury (AKI); however, the mechanisms controlling these processes have yet to be established. In our cisplatin-induced AKI mouse model, we show pathological damage to the kidneys, with the classical markers elevated, consistent with the response to cisplatin treatment. Through assessments of the components of the immune system, both locally and globally, we demonstrate that the immune microenvironment of injured kidneys was associated with an increased infiltration of CD4+ T cells and macrophages concomitant with decreased Treg cell populations. Our cell-based assays and animal studies further show that cisplatin exposure downregulated the protein levels of programmed death-ligand 1 (PD-L1), an immune checkpoint protein, in primary renal proximal tubular epithelial cells, and that these inhibitions were dose-dependent. After orthotopic delivery of PD-L1 gene into the kidneys, cisplatin-exposed mice displayed lower levels of both serum urea nitrogen and creatinine upon PD-L1 expression. Our data suggest a renoprotective effect of the immune checkpoint protein, and thereby provide a novel therapeutic strategy for cisplatin-induced AKI.


Asunto(s)
Lesión Renal Aguda , Antígeno B7-H1 , Cisplatino/efectos adversos , Células Epiteliales/metabolismo , Técnicas de Transferencia de Gen , Túbulos Renales Proximales/metabolismo , Regulación hacia Arriba , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/terapia , Animales , Antígeno B7-H1/biosíntesis , Antígeno B7-H1/genética , Linfocitos T CD4-Positivos/metabolismo , Cisplatino/farmacología , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Ratones
2.
Brief Bioinform ; 18(2): 312-320, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-26896791

RESUMEN

With expanded access to, and decreased costs of, mass spectrometry, investigators are collecting and analyzing multiple biological matrices from the same subject such as serum, plasma, tissue and urine to enhance biomarker discoveries, understanding of disease processes and identification of therapeutic targets. Commonly, each biological matrix is analyzed separately, but multivariate methods such as MANOVAs that combine information from multiple biological matrices are potentially more powerful. However, mass spectrometric data typically contain large amounts of missing values, and imputation is often used to create complete data sets for analysis. The effects of imputation on multiple biological matrix analyses have not been studied. We investigated the effects of seven imputation methods (half minimum substitution, mean substitution, k-nearest neighbors, local least squares regression, Bayesian principal components analysis, singular value decomposition and random forest), on the within-subject correlation of compounds between biological matrices and its consequences on MANOVA results. Through analysis of three real omics data sets and simulation studies, we found the amount of missing data and imputation method to substantially change the between-matrix correlation structure. The magnitude of the correlations was generally reduced in imputed data sets, and this effect increased with the amount of missing data. Significant results from MANOVA testing also were substantially affected. In particular, the number of false positives increased with the level of missing data for all imputation methods. No one imputation method was universally the best, but the simple substitution methods (Half Minimum and Mean) consistently performed poorly.


Asunto(s)
Espectrometría de Masas , Algoritmos , Teorema de Bayes , Análisis por Conglomerados , Análisis de los Mínimos Cuadrados
3.
BMC Nephrol ; 20(1): 66, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30803434

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease and is characterized by gradual cyst growth and expansion, increase in kidney volume with an ultimate decline in kidney function leading to end stage renal disease (ESRD). Given the decades long period of stable kidney function while cyst growth occurs, it is important to identify those patients who will progress to ESRD. Recent data from our and other laboratories have demonstrated that metabolic reprogramming may play a key role in cystic epithelial proliferation resulting in cyst growth in ADPKD. Height corrected total kidney volume (ht-TKV) accurately reflects cyst burden and predicts future loss of kidney function. We hypothesize that specific plasma metabolites will correlate with eGFR and ht-TKV early in ADPKD, both predictors of disease progression, potentially indicative of early physiologic derangements of renal disease severity. METHODS: To investigate the predictive role of plasma metabolites on eGFR and/or ht-TKV, we used a non-targeted GC-TOF/MS-based metabolomics approach on hypertensive ADPKD patients in the early course of their disease. Patient data was obtained from the HALT-A randomized clinical trial at baseline including estimated glomerular filtration rate (eGFR) and measured ht-TKV. To identify individual metabolites whose intensities are significantly correlated with eGFR and ht-TKV, association analyses were performed using linear regression with each metabolite signal level as the primary predictor variable and baseline eGFR and ht-TKV as the continuous outcomes of interest, while adjusting for covariates. Significance was determined by Storey's false discovery rate (FDR) q-values to correct for multiple testing. RESULTS: Twelve metabolites significantly correlated with eGFR and two triglycerides significantly correlated with baseline ht-TKV at FDR q-value < 0.05. Specific significant metabolites, including pseudo-uridine, indole-3-lactate, uric acid, isothreonic acid, and creatinine, have been previously shown to accumulate in plasma and/or urine in both diabetic and cystic renal diseases with advanced renal insufficiency. CONCLUSIONS: This study identifies metabolic derangements in early ADPKD which may be prognostic for ADPKD disease progression. CLINICAL TRIAL: HALT Progression of Polycystic Kidney Disease (HALT PKD) Study A; Clinical www.clinicaltrials.gov identifier: NCT00283686; first posted January 30, 2006, last update posted March 19, 2015.


Asunto(s)
Riñón , Riñón Poliquístico Autosómico Dominante , Insuficiencia Renal , Adulto , Creatinina/sangre , Progresión de la Enfermedad , Femenino , Humanos , Indoles/sangre , Riñón/metabolismo , Riñón/patología , Pruebas de Función Renal/métodos , Pruebas de Función Renal/estadística & datos numéricos , Estudios Longitudinales , Masculino , Tamaño de los Órganos , Gravedad del Paciente , Riñón Poliquístico Autosómico Dominante/sangre , Riñón Poliquístico Autosómico Dominante/diagnóstico , Riñón Poliquístico Autosómico Dominante/fisiopatología , Valor Predictivo de las Pruebas , Pronóstico , Seudouridina/sangre , Insuficiencia Renal/sangre , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/etiología , Ácido Úrico/sangre
4.
Am J Physiol Renal Physiol ; 315(6): F1855-F1868, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30280600

RESUMEN

Research into metabolic reprogramming in cancer has become commonplace, yet this area of research has only recently come of age in nephrology. In light of the parallels between cancer and autosomal dominant polycystic kidney disease (ADPKD), the latter is currently being studied as a metabolic disease. In clear cell renal cell carcinoma (RCC), which is now considered a metabolic disease, we and others have shown derangements in the enzyme arginosuccinate synthase 1 (ASS1), resulting in RCC cells becoming auxotrophic for arginine and leading to a new therapeutic paradigm involving reducing extracellular arginine. Based on our earlier finding that glutamine pathways are reprogrammed in ARPKD, and given the connection between arginine and glutamine synthetic pathways via citrulline, we investigated the possibility of arginine reprogramming in ADPKD. We now show that, in a remarkable parallel to RCC, ASS1 expression is reduced in murine and human ADPKD, and arginine depletion results in a dose-dependent compensatory increase in ASS1 levels as well as decreased cystogenesis in vitro and ex vivo with minimal toxicity to normal cells. Nontargeted metabolomics analysis of mouse kidney cell lines grown in arginine-deficient versus arginine-replete media suggests arginine-dependent alterations in the glutamine and proline pathways. Thus, depletion of this conditionally essential amino acid by dietary or pharmacological means, such as with arginine-degrading enzymes, may be a novel treatment for this disease.


Asunto(s)
Arginina/metabolismo , Proliferación Celular , Metabolismo Energético , Riñón/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Animales , Arginina/deficiencia , Arginina/farmacología , Argininosuccinato Sintasa/genética , Argininosuccinato Sintasa/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Femenino , Predisposición Genética a la Enfermedad , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Metabolómica/métodos , Ratones Noqueados , Fenotipo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Transducción de Señal , Canales Catiónicos TRPP/deficiencia , Canales Catiónicos TRPP/genética
5.
Bioinformatics ; 33(1): 17-25, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27592710

RESUMEN

MOTIVATION: High through-put mass spectrometry (MS) is now being used to profile small molecular compounds across multiple biological sample types from the same subjects with the goal of leveraging information across biospecimens. Multivariate statistical methods that combine information from all biospecimens could be more powerful than the usual univariate analyses. However, missing values are common in MS data and imputation can impact between-biospecimen correlation and multivariate analysis results. RESULTS: We propose two multivariate two-part statistics that accommodate missing values and combine data from all biospecimens to identify differentially regulated compounds. Statistical significance is determined using a multivariate permutation null distribution. Relative to univariate tests, the multivariate procedures detected more significant compounds in three biological datasets. In a simulation study, we showed that multi-biospecimen testing procedures were more powerful than single-biospecimen methods when compounds are differentially regulated in multiple biospecimens but univariate methods can be more powerful if compounds are differentially regulated in only one biospecimen. AVAILABILITY AND IMPLEMENTATION: We provide R functions to implement and illustrate our method as supplementary information CONTACT: sltaylor@ucdavis.eduSupplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Espectrometría de Masas/métodos , Neoplasias/metabolismo , Programas Informáticos , Animales , Glicómica/métodos , Humanos , Metabolómica/métodos , Ratones , Análisis Multivariante
6.
Kidney Int ; 91(3): 524-526, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28202165

RESUMEN

Due to their common pathogenesis and parallel proliferative signaling pathways, the cystic diseases have been recently studied in the context of cancer biology. The present study continues this paradigm by identifying signal transducer and activator of transcription (STAT5) and growth hormone (GH) as potentially modifiable pathways in polycystic kidney disease. GH, which is a potent activator of STAT5, has the additional possibility of being a biomarker, as well as providing a potential mechanism of action of somatostatin analogs in clinical trials.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Factor de Transcripción STAT5 , Hormona del Crecimiento , Humanos , Transducción de Señal
7.
Kidney Int ; 92(4): 922-933, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28545714

RESUMEN

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a common hereditary renal disease with no currently available targeted therapies. Based on the established connection between ß-catenin signaling and renal ciliopathies, and on data from our and other laboratories showing striking similarities of this disease and cancer, we evaluated the use of an orally bioavailable small molecule, KPT-9274 (a dual inhibitor of the protein kinase PAK4 and nicotinamide phosphoribosyl transferase), for treatment of ADPKD. Treatment of PKD-derived cells with this compound not only reduces PAK4 steady-state protein levels and regulates ß-catenin signaling, but also inhibits nicotinamide phosphoribosyl transferase, the rate-limiting enzyme in a key NAD salvage pathway. KPT-9274 can attenuate cellular proliferation and induce apoptosis associated with a decrease in active (phosphorylated) PAK4 and ß-catenin in several Pkd1-null murine cell lines, with a less pronounced effect on the corresponding phenotypically normal cells. Additionally, KPT-9274 shows inhibition of cystogenesis in an ex vivo model of cyclic AMP-induced cystogenesis as well as in the early stage Pkd1flox/flox:Pkhd1-Cre mouse model, the latter showing confirmation of specific anti-proliferative, apoptotic, and on-target effects. NAD biosynthetic attenuation by KPT-9274, while critical for highly proliferative cancer cells, does not appear to be important in the slower growing cystic epithelial cells during cystogenesis. KPT-9274 was not toxic in our ADPKD animal model or in other cancer models. Thus, this small molecule inhibitor could be evaluated in a clinical trial as a viable therapy of ADPKD.


Asunto(s)
Acrilamidas/farmacología , Aminopiridinas/farmacología , Citocinas/metabolismo , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Quinasas p21 Activadas/metabolismo , Acrilamidas/uso terapéutico , Aminopiridinas/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células Epiteliales , Femenino , Humanos , Riñón/citología , Masculino , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Fosforilación , Riñón Poliquístico Autosómico Dominante/patología , Receptores de Superficie Celular/genética , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPP/genética , beta Catenina/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(30): 11127-32, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024195

RESUMEN

Prostaglandins derived from the cyclooxygenase (COX) pathway and epoxyeicosatrienoic acids (EETs) from the cytochrome P450/soluble epoxide hydrolase (sEH) pathway are important eicosanoids that regulate angiogenesis and tumorigenesis. COX-2 inhibitors, which block the formation of prostaglandins, suppress tumor growth, whereas sEH inhibitors, which increase endogenous EETs, stimulate primary tumor growth and metastasis. However, the functional interactions of these two pathways in cancer are unknown. Using pharmacological inhibitors as probes, we show here that dual inhibition of COX-2 and sEH synergistically inhibits primary tumor growth and metastasis by suppressing tumor angiogenesis. COX-2/sEH dual pharmacological inhibitors also potently suppress primary tumor growth and metastasis by inhibiting tumor angiogenesis via selective inhibition of endothelial cell proliferation. These results demonstrate a critical interaction of these two lipid metabolism pathways on tumorigenesis and suggest dual inhibition of COX-2 and sEH as a potential therapeutic strategy for cancer therapy.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Ciclooxigenasa 2/metabolismo , Epóxido Hidrolasas , Proteínas de Neoplasias/farmacología , Neoplasias Experimentales , Animales , Antineoplásicos , Sinergismo Farmacológico , Epóxido Hidrolasas/antagonistas & inhibidores , Epóxido Hidrolasas/metabolismo , Masculino , Ratones , Metástasis de la Neoplasia , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología
9.
Proc Natl Acad Sci U S A ; 111(30): E3062-71, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024194

RESUMEN

Tumor suppressor p53 plays an important role in mediating growth inhibition upon telomere dysfunction. Here, we show that loss of the p53 target gene cyclin-dependent kinase inhibitor 1A (CDKN1A, also known as p21(WAF1/CIP1)) increases apoptosis induction following telomerase inhibition in a variety of cancer cell lines and mouse xenografts. This effect is highly specific to p21, as loss of other checkpoint proteins and CDK inhibitors did not affect apoptosis. In telomerase, inhibited cell loss of p21 leads to E2F1- and p53-mediated transcriptional activation of p53-upregulated modulator of apoptosis, resulting in increased apoptosis. Combined genetic or pharmacological inhibition of telomerase and p21 synergistically suppresses tumor growth. Furthermore, we demonstrate that simultaneous inhibition of telomerase and p21 also suppresses growth of tumors containing mutant p53 following pharmacological restoration of p53 activity. Collectively, our results establish that inactivation of p21 leads to increased apoptosis upon telomerase inhibition and thus identify a genetic vulnerability that can be exploited to treat many human cancers containing either wild-type or mutant p53.


Asunto(s)
Apoptosis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Neoplasias Experimentales/metabolismo , Telomerasa/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Ratones , Ratones Desnudos , Mutación , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Telomerasa/genética , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/genética
10.
J Am Soc Nephrol ; 27(8): 2227-37, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26961346

RESUMEN

Kidney cancer, or renal cell carcinoma (RCC), is a disease of increasing incidence that is commonly seen in the general practice of nephrology. However, RCC is under-recognized by the nephrology community, such that its presence in curricula and research by this group is lacking. In the most common form of RCC, clear cell renal cell carcinoma (ccRCC), inactivation of the von Hippel-Lindau tumor suppressor is nearly universal; thus, the biology of ccRCC is characterized by activation of hypoxia-relevant pathways that lead to the associated paraneoplastic syndromes. Therefore, RCC is labeled the internist's tumor. In light of this characterization and multiple other metabolic abnormalities recently associated with ccRCC, it can now be viewed as a metabolic disease. In this review, we discuss the basic biology, pathology, and approaches for treatment of RCC. It is important to distinguish between kidney confinement and distant spread of RCC, because this difference affects diagnostic and therapeutic approaches and patient survival, and it is important to recognize the key interplay between RCC, RCC therapy, and CKD. Better understanding of all aspects of this disease will lead to optimal patient care and more recognition of an increasingly prevalent nephrologic disease, which we now appropriately label the nephrologist's tumor.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Fenómenos Biológicos , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/terapia , Humanos , Neoplasias Renales/diagnóstico , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/terapia , Nefrólogos
11.
Proc Natl Acad Sci U S A ; 110(16): 6530-5, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23553837

RESUMEN

Epidemiological and preclinical evidence supports that omega-3 dietary fatty acids (fish oil) reduce the risks of macular degeneration and cancers, but the mechanisms by which these omega-3 lipids inhibit angiogenesis and tumorigenesis are poorly understood. Here we show that epoxydocosapentaenoic acids (EDPs), which are lipid mediators produced by cytochrome P450 epoxygenases from omega-3 fatty acid docosahexaenoic acid, inhibit VEGF- and fibroblast growth factor 2-induced angiogenesis in vivo, and suppress endothelial cell migration and protease production in vitro via a VEGF receptor 2-dependent mechanism. When EDPs (0.05 mg · kg(-1) · d(-1)) are coadministered with a low-dose soluble epoxide hydrolase inhibitor, EDPs are stabilized in circulation, causing ~70% inhibition of primary tumor growth and metastasis. Contrary to the effects of EDPs, the corresponding metabolites derived from omega-6 arachidonic acid, epoxyeicosatrienoic acids, increase angiogenesis and tumor progression. These results designate epoxyeicosatrienoic acids and EDPs as unique endogenous mediators of an angiogenic switch to regulate tumorigenesis and implicate a unique mechanistic linkage between omega-3 and omega-6 fatty acids and cancers.


Asunto(s)
Carcinoma Pulmonar de Lewis/prevención & control , Transformación Celular Neoplásica/efectos de los fármacos , Ácidos Docosahexaenoicos/metabolismo , Compuestos Epoxi/farmacología , Ácidos Grasos Omega-3/química , Ácidos Grasos Insaturados/farmacología , Metástasis de la Neoplasia/prevención & control , Neovascularización Patológica/prevención & control , Animales , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Epóxido Hidrolasas/antagonistas & inhibidores , Compuestos Epoxi/metabolismo , Ácidos Grasos Insaturados/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Microscopía
12.
Clin J Sport Med ; 26(5): 417-22, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26657822

RESUMEN

OBJECTIVE: Examine whether the acute kidney injury (AKI) commonly observed among ultramarathon participants places the individual at risk for subsequent AKI of worse magnitude. DESIGN: Observational. SETTING: Western States Endurance Run. PARTICIPANTS: Race finishers with postrace blood studies. INDEPENDENT VARIABLE: Acute kidney injury after 1 race. MAIN OUTCOME MEASURES: Extent of AKI in subsequent race. RESULTS: Among 627 finishes in which serum creatinine values were known, 36.2% met "risk" or "injury" criterion with this group characterized by having faster finish times, greater body weight loss during the race, and higher postrace serum creatine kinase and urea nitrogen concentrations when compared with those not meeting the criteria. We identified 38 runners who had undergone postrace blood analyses at multiple races among which 16 (42.1%) met the risk or injury criterion at the first race. Of those 16 runners, 12 (75%) met the criteria at a subsequent race, an incidence that was higher (P = 0.0026) than the overall 36.2% incidence. For most (56.2%) of the 16 runners meeting the criteria at the first race, the subsequent race caused less increase in serum creatinine concentration and decrement in estimated glomerular filtration rate than the first race. CONCLUSIONS: Mild AKI is common in 161-km ultramarathons, but there was no evidence that previous AKI caused greater renal dysfunction from a subsequent exercise stimulus of similar magnitude. This offers some reassurance to runners and their physicians that mild to moderate AKI in the setting of an ultramarathon is not cumulative or without complete recovery of kidney function when stressed.


Asunto(s)
Lesión Renal Aguda/etiología , Carrera/fisiología , Lesión Renal Aguda/sangre , Lesión Renal Aguda/diagnóstico , Adulto , Biomarcadores/sangre , Creatinina/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
13.
Am J Physiol Cell Physiol ; 308(11): C890-8, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810260

RESUMEN

Kidney cancer [renal cell carcinoma (RCC)] is the sixth-most-common cancer in the United States, and its incidence is increasing. The current progression-free survival for patients with advanced RCC rarely extends beyond 1-2 yr due to the development of therapeutic resistance. We previously identified peroxisome proliferator-activating receptor-α (PPARα) as a potential therapeutic target for this disease and showed that a specific PPARα antagonist, GW6471, induced apoptosis and cell cycle arrest at G0/G1 in RCC cell lines associated with attenuation of cell cycle regulatory proteins. We now extend that work and show that PPARα inhibition attenuates components of RCC metabolic reprogramming, capitalizing on the Warburg effect. The specific PPARα inhibitor GW6471, as well as a siRNA specific to PPARα, attenuates the enhanced fatty acid oxidation and oxidative phosphorylation associated with glycolysis inhibition, and PPARα antagonism also blocks the enhanced glycolysis that has been observed in RCC cells; this effect did not occur in normal human kidney epithelial cells. Such cell type-specific inhibition of glycolysis corresponds with changes in protein levels of the oncogene c-Myc and has promising clinical implications. Furthermore, we show that treatment with GW6471 results in RCC tumor growth attenuation in a xenograft mouse model, with minimal obvious toxicity, a finding associated with the expected on-target effects on c-Myc. These studies demonstrate that several pivotal cancer-relevant metabolic pathways are inhibited by PPARα antagonism. Our data support the concept that targeting PPARα, with or without concurrent inhibition of glycolysis, is a potential novel and effective therapeutic approach for RCC that targets metabolic reprogramming in this tumor.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/tratamiento farmacológico , Oxazoles/farmacología , PPAR alfa/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Tirosina/análogos & derivados , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Ratones , Ratones Desnudos , Especificidad de Órganos , PPAR alfa/genética , PPAR alfa/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Tirosina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Am J Physiol Renal Physiol ; 309(6): F492-8, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26155843

RESUMEN

Since polycystic kidney disease (PKD) was first noted over 30 years ago to have neoplastic parallels, there has been a resurgent interest in elucidating neoplasia-relevant pathways in PKD. Taking a nontargeted metabolomics approach in the B6(Cg)-Cys1(cpk/)J (cpk) mouse model of recessive PKD, we have now characterized metabolic reprogramming in these tissues, leading to a glutamine-dependent TCA cycle shunt toward total 2-hydroxyglutarate (2-HG) production in cpk compared with B6 wild-type kidney tissue. After confirmation of increased 2-HG expression in immortalized collecting duct cpk cells as well as in human autosomal recessive PKD tissue using targeted analysis, we show that the increase in 2-HG is likely due to glutamine-sourced α-ketoglutarate. In addition, cpk cells require exogenous glutamine for growth such that inhibition of glutaminase-1 decreases cell viability as well as proliferation. This study is a demonstration of the striking parallels between recessive PKD and cancer metabolism. Our data, once confirmed in other PKD models, suggest that future therapeutic approaches targeting this pathway, such as using glutaminase inhibitors, have the potential to open novel treatment options for renal cystic disease.


Asunto(s)
Creatina Quinasa/genética , Glutamina/metabolismo , Glutaratos/metabolismo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Animales , Células Cultivadas , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Glutaminasa/antagonistas & inhibidores , Humanos , Lactante , Recién Nacido , Masculino , Metabolómica , Ratones , Modelos Genéticos
15.
Clin Nephrol ; 83(4): 235-42, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24931911

RESUMEN

BACKGROUND: Proper acute management of exercise-associated hyponatremia (EAH) has been known for decades, yet this information has not been uniformly implemented. Since treatment of EAH with isotonic fluids can result in delayed recovery and death, it is important that proper acute management in the field and hospital be utilized. CASE REPORT: We describe a participant of the 161-km Western States Endurance Run (WSER) who presented with seizure after dropping out at 145 km. He had gained 2.2% of his initial weight by 126 km from using sodium supplements and drinking copious volumes of fluids. He was treated promptly in the field for presumed EAH with two intravenous boluses of 100 mL of 3% hypertonic saline and showed rapid improvement in neurologic status. His recovery was then delayed with the use of high volumes of intravenous isotonic fluids, apparently for concern over his mild exertional rhabdomyolysis. CONCLUSIONS: Symptomatic EAH should be acutely managed with hypertonic saline, whereas treatment with high volumes of isotonic fluids may delay recovery and has even resulted in deaths from cerebral edema. Concern over central pontine myelinolysis from rapid correction of hyponatremia in EAH is unsupported. Furthermore, the exertional rhabdomyolysis often associated with EAH, and the concern over progression to acute kidney failure, should not dictate initial treatment.


Asunto(s)
Ejercicio Físico , Hiponatremia/terapia , Rabdomiólisis/terapia , Humanos , Masculino , Persona de Mediana Edad , Solución Salina Hipertónica/uso terapéutico , Sodio en la Dieta/administración & dosificación
16.
Int J Mol Sci ; 16(5): 11609-28, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-26006237

RESUMEN

Phosphorylation of H2AX on Ser139 (γH2AX) after exposure to ionizing radiation produces nuclear foci that are detectable by immunofluorescence microscopy. These so-called γH2AX foci have been adopted as quantitative markers for DNA double-strand breaks. High numbers of spontaneous γH2AX foci have also been reported for some human solid tumor-derived cell lines, but the molecular mechanism(s) for this response remains elusive. Here we show that cancer cells (e.g., HCT116; MCF7) that constitutively express detectable levels of p21WAF1 (p21) exhibit low numbers of γH2AX foci (<3/nucleus), whereas p21 knockout cells (HCT116p21-/-) and constitutively low p21-expressing cells (e.g., MDA-MB-231) exhibit high numbers of foci (e.g., >50/nucleus), and that these foci are not associated with apoptosis. The majority (>95%) of cells within HCT116p21-/- and MDA-MB-231 cultures contain high levels of phosphorylated p53, which is localized in the nucleus. We further show an inverse relationship between γH2AX foci and nuclear accumulation of WIP1, an oncogenic phosphatase. Our studies suggest that: (i) p21 deficiency might provide a selective pressure for the emergence of apoptosis-resistant progeny exhibiting genomic instability, manifested as spontaneous γH2AX foci coupled with phosphorylation and nuclear accumulation of p53; and (ii) p21 might contribute to positive regulation of WIP1, resulting in dephosphorylation of γH2AX.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Roturas del ADN de Doble Cadena , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Fosfoproteínas Fosfatasas/genética , Apoptosis , Línea Celular Tumoral , Reparación del ADN , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Inestabilidad Genómica , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias/metabolismo , Fosforilación , Proteína Fosfatasa 2C
17.
Am J Physiol Renal Physiol ; 307(11): F1179-86, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25234309

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is a progressive, proliferative renal disease. Kidneys from ADPKD patients are characterized by the presence of cysts that are marked by enhanced proliferation and apoptosis of renal tubular epithelial cells. Current treatment of this disease is supportive, as there are few if any clinically validated targeted therapeutics. Given the parallels between cystic disease and cancer, and in light of our findings of the efficacy of the nuclear transport inhibitors in kidney cancer, which has similarities to ADPKD, we asked whether such inhibitors show utility in ADPKD. In this study, we tested selective inhibitors of nuclear export (SINE) in two human ADPKD cell lines and in an in vivo mouse model of ADPKD. After effective downregulation of a nuclear exporter, exportin 1 (XPO1), with KPT-330, both cell lines showed dose-dependent inhibition of cell proliferation through G0/G1 arrest associated with downregulation of CDK4, with minimal apoptosis. To analyze mechanisms of CDK4 decrease by XPO1 inhibition, localization of various XPO1 target proteins was examined, and C/EBPß was found to be localized in the nucleus by XPO1 inhibition, resulting in an increase of C/EBPα, which activates degradation of CDK4. Furthermore, inhibition of XPO1 with the parallel inhibitor KPT-335 attenuated cyst growth in vivo in the PKD1 mutant mouse model Pkd1(v/v). Thus, inhibition of nuclear export by KPT-330, which has shown no adverse effects in renal serum chemistries and urinalyses in animal models, and which is already in phase 1 trials for cancers, will be rapidly translatable to human ADPKD.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quistes/patología , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/patología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Humanos , Hidrazinas/farmacología , Carioferinas/antagonistas & inhibidores , Ratones , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Triazoles/farmacología , Proteína Exportina 1
18.
Anticancer Drugs ; 25(4): 433-46, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24525589

RESUMEN

In the current work, we carried out a mechanistic study on the cytotoxicity of two compounds, trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-N-methyl-benzamide (t-AUCMB) and trans-N-methyl-4-{4-[3-(4-trifluoromethoxy-phenyl)-ureido]-cyclohexyloxy}-benzamide (t-MTUCB), that are structurally similar to sorafenib. These compounds show strong cytotoxic responses in various cancer cell lines, despite significant differences in the induction of apoptotic events such as caspase activation and lactate dehydrogenase release in hepatoma cells. Both compounds induce autophagosome formation and LC3I cleavage, but there was little observable effect on mTORC1 or the downstream targets, S6K1 and 4E-binding protein. In addition, there was an increase in the activity of upstream signaling through the IRS1/PI3K/Akt-signaling pathway, suggesting that, unlike sorafenib, both compounds induce mammalian target of rapamycin (mTOR)-independent autophagy. The autophagy observed correlates with mitochondrial membrane depolarization, apoptosis-inducing factor release, and oxidative stress-induced glutathione depletion. However, there were no observable changes in the endoplasmic reticulum-stress markers such as binding immunoglobulin protein, inositol-requiring enzyme-α, phosphorylated eukaryotic initiation factor 2, and the lipid peroxidation marker, 4-hydroxynonenal, suggesting endoplasmic reticulum-independent oxidative stress. Finally, these compounds do not have the multikinase inhibitory activity of sorafenib, which may be reflected in their difference in the ability to halt cell cycle progression compared with sorafenib. Our findings indicate that both compounds have anticancer effects comparable with sorafenib in multiple cell lines, but they induce significant differences in apoptotic responses and appear to induce mTOR-independent autophagy. t-AUCMB and t-MTUCB represent novel chemical probes that are capable of inducing mTOR-independent autophagy and apoptosis to differing degrees, and may thus be potential tools for further understanding the link between these two cellular stress responses.


Asunto(s)
Adamantano/análogos & derivados , Antineoplásicos/farmacología , Benzamidas/farmacología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Urea/análogos & derivados , Adamantano/farmacología , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Membranas Mitocondriales/fisiología , Niacinamida/farmacología , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/metabolismo , Sorafenib , Urea/farmacología
19.
Clin Chem ; 59(1): 138-46, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23150057

RESUMEN

BACKGROUND: Metabolomics, the study of all metabolites produced in the body, which often includes flora and drug metabolites, is the omics approach that can be considered most closely related to a patient's phenotype. Metabolomics has a great and largely untapped potential in the field of oncology, and the analysis of the cancer metabolome to identify biofluid markers and novel druggable targets can now be undertaken in many research laboratories. CONTENT: The cancer metabolome has been used to identify and begin to evaluate potential biomarkers and therapeutic targets in a variety of malignancies, including breast, prostate, and kidney cancer. We discuss the several standard techniques for metabolite separation and identification, with their potential problems and drawbacks. Validation of biomarkers and targets may entail intensive use of labor and technology and generally requires a large number of study participants as well as laboratory validation studies. The field of pharmacometabolomics, in which specific therapies are chosen on the basis of a patient's metabolomic profile, has shown some promise in the translation of metabolomics into the arena of personalized medicine. SUMMARY: The relatively new approach using metabolomics has just begun to enter the mainstream of cancer diagnostics and therapeutics. As this field advances, metabolomics will take its well-deserved place next to genomics, transcriptomics, and proteomics in both clinical and basic research in oncology.


Asunto(s)
Metaboloma , Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Diagnóstico Precoz , Humanos , Neoplasias/diagnóstico , Neoplasias/radioterapia , Medicina de Precisión
20.
J Urol ; 189(6): 2317-26, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23079374

RESUMEN

PURPOSE: Renal cell carcinoma often presents asymptomatically and patients are commonly diagnosed at the metastatic stage, when treatment options are limited and survival is poor. Since progression-free survival using current therapy for metastatic renal cell carcinoma is only 1 to 2 years and existing drugs are associated with a high resistance rate, new pharmacological targets are needed. We identified and evaluated the nuclear exporter protein CRM1 as a novel potential therapy for renal cell carcinoma. MATERIALS AND METHODS: We tested the efficacy of the CRM1 inhibitors KPT-185 and 251 in several renal cell carcinoma cell lines and in a renal cell carcinoma xenograft model. Apoptosis and cell cycle arrest were quantified and localization of p53 family proteins was assessed using standard techniques. RESULTS: KPT-185 attenuated CRM1 and showed increased cytotoxicity in renal cell carcinoma cells in vitro with evidence of increased apoptosis as well as cell cycle arrest. KPT-185 caused p53 and p21 to remain primarily in the nucleus in all renal cell carcinoma cell lines, suggesting that the mechanism of action of these compounds depends on tumor suppressor protein localization. Furthermore, when administered orally in a high grade renal cell carcinoma xenograft model, the bioavailable CRM1 inhibitor KPT-251 significantly inhibited tumor growth in vivo with the expected on target effects and no obvious toxicity. CONCLUSIONS: The CRM1 inhibitor protein family is a novel therapeutic target for renal cell carcinoma that deserves further intensive investigation for this and other urological malignancies.


Asunto(s)
Acrilatos/farmacología , Antineoplásicos/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Carioferinas/efectos de los fármacos , Neoplasias Renales/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Triazoles/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Línea Celular Tumoral/efectos de los fármacos , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Carioferinas/genética , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Distribución Aleatoria , Receptores Citoplasmáticos y Nucleares/genética , Sensibilidad y Especificidad , Tasa de Supervivencia , Carga Tumoral/efectos de los fármacos , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA