Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Med ; 22(10): 1120-1130, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27571348

RESUMEN

Cachexia represents a fatal energy-wasting syndrome in a large number of patients with cancer that mostly results in a pathological loss of skeletal muscle and adipose tissue. Here we show that tumor cell exposure and tumor growth in mice triggered a futile energy-wasting cycle in cultured white adipocytes and white adipose tissue (WAT), respectively. Although uncoupling protein 1 (Ucp1)-dependent thermogenesis was dispensable for tumor-induced body wasting, WAT from cachectic mice and tumor-cell-supernatant-treated adipocytes were consistently characterized by the simultaneous induction of both lipolytic and lipogenic pathways. Paradoxically, this was accompanied by an inactivated AMP-activated protein kinase (Ampk), which is normally activated in peripheral tissues during states of low cellular energy. Ampk inactivation correlated with its degradation and with upregulation of the Ampk-interacting protein Cidea. Therefore, we developed an Ampk-stabilizing peptide, ACIP, which was able to ameliorate WAT wasting in vitro and in vivo by shielding the Cidea-targeted interaction surface on Ampk. Thus, our data establish the Ucp1-independent remodeling of adipocyte lipid homeostasis as a key event in tumor-induced WAT wasting, and we propose the ACIP-dependent preservation of Ampk integrity in the WAT as a concept in future therapies for cachexia.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos Blancos/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Caquexia/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Neoplasias/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Quinasas Activadas por AMP/farmacología , Adipocitos Blancos/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Caquexia/etiología , Células Cultivadas , Técnicas In Vitro , Lipogénesis/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Neoplasias/complicaciones , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/efectos de los fármacos , Proteína Desacopladora 1/metabolismo
3.
Cell Cycle ; 14(23): 3734-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26654769

RESUMEN

Seven-in-absentia homolog (SIAH) proteins are evolutionary conserved RING type E3 ubiquitin ligases responsible for the degradation of key molecules regulating DNA damage response, hypoxic adaptation, apoptosis, angiogenesis, and cell proliferation. Many studies suggest a tumorigenic role for SIAH2. In breast cancer patients SIAH2 expression levels correlate with cancer aggressiveness and overall patient survival. In addition, SIAH inhibition reduced metastasis in melanoma. The role of SIAH1 in breast cancer is still ambiguous; both tumorigenic and tumor suppressive functions have been reported. Other studies categorized SIAH ligases as either pro- or antimigratory, while the significance for metastasis is largely unknown. Here, we re-evaluated the effects of SIAH1 and SIAH2 depletion in breast cancer cell lines, focusing on migration and invasion. We successfully knocked down SIAH1 and SIAH2 in several breast cancer cell lines. In luminal type MCF7 cells, this led to stabilization of the SIAH substrate Prolyl Hydroxylase Domain protein 3 (PHD3) and reduced Hypoxia-Inducible Factor 1α (HIF1α) protein levels. Both the knockdown of SIAH1 or SIAH2 led to increased apoptosis and reduced proliferation, with comparable effects. These results point to a tumor promoting role for SIAH1 in breast cancer similar to SIAH2. In addition, depletion of SIAH1 or SIAH2 also led to decreased cell migration and invasion in breast cancer cells. SIAH knockdown also controlled microtubule dynamics by markedly decreasing the protein levels of stathmin, most likely via p27(Kip1). Collectively, these results suggest that both SIAH ligases promote a migratory cancer cell phenotype and could contribute to metastasis in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Movimiento Celular/genética , Invasividad Neoplásica/genética , Proteínas Nucleares/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Apoptosis/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Microtúbulos/metabolismo , Metástasis de la Neoplasia/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , Estatmina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Cancer Metab ; 3: 11, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26500770

RESUMEN

BACKGROUND: Numerous studies have demonstrated that functional mitochondria are required for tumorigenesis, suggesting that mitochondrial oxidative phosphorylation (OXPHOS) might be a potential target for cancer therapy. In this study, we investigated the effects of BAY 87-2243, a small molecule that inhibits the first OXPHOS enzyme (complex I), in melanoma in vitro and in vivo. RESULTS: BAY 87-2243 decreased mitochondrial oxygen consumption and induced partial depolarization of the mitochondrial membrane potential. This was associated with increased reactive oxygen species (ROS) levels, lowering of total cellular ATP levels, activation of AMP-activated protein kinase (AMPK), and reduced cell viability. The latter was rescued by the antioxidant vitamin E and high extracellular glucose levels (25 mM), indicating the involvement of ROS-induced cell death and a dependence on glycolysis for cell survival upon BAY 87-2243 treatment. BAY 87-2243 significantly reduced tumor growth in various BRAF mutant melanoma mouse xenografts and patient-derived melanoma mouse models. Furthermore, we provide evidence that inhibition of mutated BRAF using the specific small molecule inhibitor vemurafenib increased the OXPHOS dependency of BRAF mutant melanoma cells. As a consequence, the combination of both inhibitors augmented the anti-tumor effect of BAY 87-2243 in a BRAF mutant melanoma mouse xenograft model. CONCLUSIONS: Taken together, our results suggest that complex I inhibition has potential clinical applications as a single agent in melanoma and also might be efficacious in combination with BRAF inhibitors in the treatment of patients with BRAF mutant melanoma.

5.
EMBO Mol Med ; 5(2): 294-308, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23307490

RESUMEN

In mammals, proper storage and distribution of lipids in and between tissues is essential for the maintenance of energy homeostasis. Here, we show that tumour growth triggers hepatic metabolic dysfunction as part of the cancer cachectic phenotype, particularly by reduced hepatic very-low-density-lipoprotein (VLDL) secretion and hypobetalipoproteinemia. As a molecular cachexia output pathway, hepatic levels of the transcription factor transforming growth factor beta 1-stimulated clone (TSC) 22 D4 were increased in cancer cachexia. Mimicking high cachectic levels of TSC22D4 in healthy livers led to the inhibition of hepatic VLDL release and lipogenic genes, and diminished systemic VLDL levels under both normal and high fat dietary conditions. Liver-specific ablation of TSC22D4 triggered hypertriglyceridemia through the induction of hepatic VLDL secretion. Furthermore, hepatic TSC22D4 expression levels were correlated with the degree of body weight loss and VLDL hypo-secretion in cancer cachexia, and TSC22D4 deficiency rescued tumour cell-induced metabolic dysfunction in hepatocytes. Therefore, hepatic TSC22D4 activity may represent a molecular rationale for peripheral energy deprivation in subjects with metabolic wasting diseases, including cancer cachexia.


Asunto(s)
Caquexia/metabolismo , Hígado/metabolismo , Neoplasias/complicaciones , Factores de Transcripción/metabolismo , Animales , Caquexia/etiología , Humanos , Lipoproteínas VLDL/metabolismo , Hepatopatías/etiología , Hepatopatías/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Factores de Transcripción/genética , Triglicéridos/metabolismo
6.
Biochim Biophys Acta ; 1831(5): 896-904, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23051608

RESUMEN

Epidemiological studies estimate that by the year 2030, 2.16 billion people worldwide will be overweight and 1.12 billion will be obese [1]. Besides its now established function as an endocrine organ, adipose tissue plays a fundamental role as an energy storage compartment. As such, adipose tissue is capable of extensive expansion or retraction depending on the energy balance or disease state of the host, a plasticity that is unparalleled in other organs and - under conditions of excessive energy intake - significantly contributes to the afore mentioned obesity pandemic. Expansion of adipose tissue is driven by both hypertrophy and hyperplasia of adipocytes, which can renew frequently to compensate for cell death. This underlines the importance of adipocyte progenitor cells within the distinct adipose tissue depots to control both energy storage and endocrine functions of adipose tissue. Here we summarize recent findings on the identity and plasticity of adipose stem cells, the involved signaling cascades, and potential clinical implications of these cells for the treatment of metabolic dysfunction in obesity. This article is part of a Special Issue entitled Brown and White Fat: From Signaling to Disease.


Asunto(s)
Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Obesidad/fisiopatología , Células Madre/citología , Animales , Humanos , Transducción de Señal
7.
Cancer Prev Res (Phila) ; 5(4): 536-43, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22262811

RESUMEN

Pharmacoepidemiologic studies provide evidence that use of metformin, a drug commonly prescribed for type II diabetes, is associated with a substantial reduction in cancer risk. Experimental models show that metformin inhibits the growth of certain neoplasms by cell autonomous mechanisms such as activation of AMP kinase with secondary inhibition of protein synthesis or by an indirect mechanism involving reduction in gluconeogenesis leading to a decline in insulin levels and reduced proliferation of insulin-responsive cancers. Here, we show that metformin attenuates paraquat-induced elevations in reactive oxygen species (ROS), and related DNA damage and mutations, but has no effect on similar changes induced by H(2)0(2), indicating a reduction in endogenous ROS production. Importantly, metformin also inhibited Ras-induced ROS production and DNA damage. Our results reveal previously unrecognized inhibitory effects of metformin on ROS production and somatic cell mutation, providing a novel mechanism for the reduction in cancer risk reported to be associated with exposure to this drug.


Asunto(s)
Daño del ADN , Hipoglucemiantes/farmacología , Metformina/farmacología , Especies Reactivas de Oxígeno , Adenilato Quinasa/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 2/genética , Ensayo de Inmunoadsorción Enzimática/métodos , Factor de Crecimiento Epidérmico/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Peróxido de Hidrógeno/metabolismo , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos , Mutagénesis , Mutación , NADP/metabolismo
8.
Cell Cycle ; 10(16): 2770-8, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21811094

RESUMEN

Insulin regulates glucose uptake by normal tissues. Although there is evidence that certain cancers are growth-stimulated by insulin, the possibility that insulin influences tumor glucose uptake as assessed by ( 18) F-2-Fluoro-2-Deoxy-d-Glucose Positron Emission Tomography (FDG-PET) has not been studied in detail. We present a model of diet-induced hyperinsulinemia associated with increased insulin receptor activation in neoplastic tissue and with increased tumor FDG-PET image intensity. Metformin abolished the diet-induced increases in serum insulin level, tumor insulin receptor activation and tumor FDG uptake associated with the high energy diet but had no effect on these measurements in mice on a control diet. These findings provide the first functional imaging correlate of the well-known adverse effect of caloric excess on cancer outcome. They demonstrate that, for a subset of neoplasms, diet and insulin are variables that affect tumor FDG uptake and have implications for design of clinical trials of metformin as an antineoplastic agent.


Asunto(s)
Fluorodesoxiglucosa F18/metabolismo , Insulina/sangre , Metformina/farmacología , Alimentación Animal , Animales , Antineoplásicos/farmacología , Glucemia/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/metabolismo , Radioisótopos de Flúor/análisis , Glucosa/metabolismo , Hiperinsulinismo/inducido químicamente , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Tomografía de Emisión de Positrones , Receptor de Insulina/metabolismo , Transducción de Señal
9.
Prostate ; 70(15): 1628-35, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564323

RESUMEN

BACKGROUND: Plasma insulin concentration is increased in prostate cancer patients during androgen deprivation therapy (ADT) and hyperinsulinemia has been associated with aggressive prostate cancer behavior. To investigate the possible role of castration-induced hyperinsulinemia as a mechanism that may attenuate the beneficial effects of ADT in patients with prostate cancer, a murine model would be useful. We therefore investigated long-term metabolic effects of castration in several mouse models. METHODS: We studied the long-term influence of castration on energy intake, body weight, glucose tolerance, plasma-insulin, plasma insulin-like growth factor-1 (IGF-1), plasma adiponectin, and plasma leptin in C57BL/6, Swiss nu/nu, and CB17 scid mice receiving various diets. In each case, mice were randomized to have either bilateral orchiectomy or a sham operation. RESULTS: Energy intake, body weight, blood glucose levels in glucose tolerance test, plasma insulin, plasma IGF-1, and plasma leptin level in all had a trend to be decreased in castrated as compared to sham operated mice. Plasma adiponectin level was increased in the castrated mice. CONCLUSIONS: The effects of castration on glucose, insulin, and related markers in several mouse models studied does not coincide with clinical observations; further studies in this area will require clinical research and/or the use of alternate models such as the dog.


Asunto(s)
Andrógenos/deficiencia , Glucemia/metabolismo , Insulina/sangre , Neoplasias Hormono-Dependientes/sangre , Neoplasias de la Próstata/sangre , Adiponectina/sangre , Animales , Peso Corporal/fisiología , Modelos Animales de Enfermedad , Ingestión de Energía/fisiología , Prueba de Tolerancia a la Glucosa , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Orquiectomía , Neoplasias de la Próstata/tratamiento farmacológico , Distribución Aleatoria
10.
Endocr Relat Cancer ; 17(2): 351-60, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20228137

RESUMEN

The molecular mechanisms responsible for the association of obesity with adverse colon cancer outcomes are poorly understood. We investigated the effects of a high-energy diet on growth of an in vivo colon cancer model. Seventeen days following the injection of 5x10(5) MC38 colon carcinoma cells, tumors from mice on the high-energy diet were approximately twice the volume of those of mice on the control diet. These findings were correlated with the observation that the high-energy diet led to elevated insulin levels, phosphorylated AKT, and increased expression of fatty acid synthase (FASN) by the tumor cells. Metformin, an antidiabetic drug, leads to the activation of AMPK and is currently under investigation for its antineoplastic activity. We observed that metformin blocked the effect of the high-energy diet on tumor growth, reduced insulin levels, and attenuated the effect of diet on phosphorylation of AKT and expression of FASN. Furthermore, the administration of metformin led to the activation of AMPK, the inhibitory phosphorylation of acetyl-CoA carboxylase, the upregulation of BNIP3 and increased apoptosis as estimated by poly (ADP-ribose) polymerase (PARP) cleavage. Prior work showed that activating mutations of PI3K are associated with increased AKT activation and adverse outcome in colon cancer; our results demonstrate that the aggressive tumor behavior associated with a high-energy diet has similar effects on this signaling pathway. Furthermore, metformin is demonstrated to reverse the effects of the high-energy diet, thus suggesting a potential role for this agent in the management of a metabolically defined subset of colon cancers.


Asunto(s)
Carcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Ingestión de Energía/efectos de los fármacos , Ácido Graso Sintasas/antagonistas & inhibidores , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma/enzimología , Neoplasias del Colon/enzimología , Ácido Graso Sintasas/biosíntesis , Insulina/sangre , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células Tumorales Cultivadas
11.
Cancer Lett ; 289(2): 246-53, 2010 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-19744772

RESUMEN

PTEN loss of function enhances proliferation, but effects on cellular energy metabolism are less well characterized. We used an inducible PTEN expression vector in a PTEN-null glioma cell line to examine this issue. While proliferation of PTEN-positive cells was insensitive to increases in glucose concentration beyond 2.5mM, PTEN-null cells significantly increased proliferation with increasing glucose concentration across the normal physiologic range to approximately 10mM, coinciding with a shift to glycolysis and "glucose addiction". This demonstrates that the impact of loss of function of PTEN is modified by glucose concentration, and may be relevant to epidemiologic results linking hyperglycemia to cancer risk and cancer mortality.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proliferación Celular , Desoxiglucosa/farmacología , Glioma/metabolismo , Glucosa/metabolismo , Glucólisis , Fosfohidrolasa PTEN/metabolismo , Antimetabolitos/farmacología , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Glioma/genética , Glioma/patología , Humanos , Consumo de Oxígeno/efectos de los fármacos , Fosfohidrolasa PTEN/genética , Células Tumorales Cultivadas
12.
Endocr Relat Cancer ; 15(3): 833-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18469156

RESUMEN

We investigated the effects of metformin on the growth of lewis lung LLC1 carcinoma in C57BL/6J mice provided with either a control diet or a high-energy diet, previously reported to lead to weight gain and systemic insulin resistance with hyperinsulinemia. Forty-eight male mice were randomized into four groups: control diet, control diet+metformin, high-energy diet, or high-energy diet+metformin. Following 8 weeks on the experimental diets, selected groups received metformin in their drinking water. Three weeks following the start of metformin treatment, mice were injected with 0.5x10(6) LLC1 cells and tumor growth was measured for 17 days. By day 17, tumors of mice on the high-energy diet were nearly twice the volume of those of mice on the control diet. This effect of diet on tumor growth was significantly attenuated by metformin, but metformin had no effect on tumor growth of the mice on the control diet. Metformin attenuated the increased insulin receptor activation associated with the high-energy diet and also led to increased phosphorylation of AMP kinase, two actions that would be expected to decrease neoplastic proliferation. These experimental results are consistent with prior hypothesis-generating epidemiological studies that suggest that metformin may reduce cancer risk and improve cancer prognosis. Finally, these results contribute to the rationale for evaluation of the anti-neoplastic activity of metformin in hyperinsulinemic cancer patients.


Asunto(s)
Carcinoma Pulmonar de Lewis/patología , Proliferación Celular/efectos de los fármacos , Dieta Aterogénica , Ingestión de Energía/fisiología , Neoplasias Pulmonares/patología , Metformina/farmacología , Algoritmos , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Evaluación Preclínica de Medicamentos , Ingestión de Energía/efectos de los fármacos , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
13.
Biol Reprod ; 72(1): 179-87, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15385415

RESUMEN

One-cell-stage embryos derived from most random-bred and inbred female mice exhibit an in vitro developmental block at the two-cell stage in classical embryo culture media. However, embryos derived from many F1 hybrids develop easily past the two-cell stage under the same conditions. This has given rise to the commonly accepted idea that there exist blocking and nonblocking types of female mice, with only the former being prone to a two-cell block. Recently, culture media have been improved to the point that even embryos prone to the two-cell block will develop past the block in vitro, making it possible to study its etiology. Here, we show that either increased osmolarity or increased glucose/phosphate levels induced the expected two-cell block in random-bred CF1 embryos and the two-cell block at increased osmolarities could be rescued by the organic osmolyte glycine. Surprisingly, one-cell embryos from B6D2F1 (BDF1) F1 hybrid females, considered to be nonblocking, also became blocked at the two-cell stage when osmolarity or glucose/phosphate levels were increased. They were also similarly rescued by glycine from the osmolarity-induced block. The most evident difference was that the purportedly nonblocking embryos became blocked at a higher threshold of osmolarity or glucose/phosphate level than those considered prone to this developmental block. Thus, both blocking and nonblocking embryos actually exhibit a similar two-cell block to development.


Asunto(s)
Desarrollo Embrionario/fisiología , Glucosa/farmacología , Concentración Osmolar , Fosfatos/farmacología , Animales , Animales no Consanguíneos , Fase de Segmentación del Huevo/citología , Relación Dosis-Respuesta a Droga , Técnicas de Cultivo de Embriones/métodos , Desarrollo Embrionario/efectos de los fármacos , Femenino , Fertilización In Vitro , Glicina/farmacología , Ratones , Ratones Endogámicos , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...