Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 107(9): 4153-8, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20160076

RESUMEN

Ataxia-telangiectasia mutated (ATM) is a cellular damage sensor that coordinates the cell cycle with damage-response checkpoints and DNA repair to preserve genomic integrity. However, ATM also has been implicated in metabolic regulation, and ATM deficiency is associated with elevated reactive oxygen species (ROS). ROS has a central role in many physiological and pathophysiological processes including inflammation and chronic diseases such as atherosclerosis and cancer, underscoring the importance of cellular pathways involved in redox homeostasis. We have identified a cytoplasmic function for ATM that participates in the cellular damage response to ROS. We show that in response to elevated ROS, ATM activates the TSC2 tumor suppressor via the LKB1/AMPK metabolic pathway in the cytoplasm to repress mTORC1 and induce autophagy. Importantly, elevated ROS and dysregulation of mTORC1 in ATM-deficient cells is inhibited by rapamycin, which also rescues lymphomagenesis in Atm-deficient mice. Our results identify a cytoplasmic pathway for ROS-induced ATM activation of TSC2 to regulate mTORC1 signaling and autophagy, identifying an integration node for the cellular damage response with key pathways involved in metabolism, protein synthesis, and cell survival.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adenilato Quinasa/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Línea Celular , Proteínas de Unión al ADN/genética , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos , Estrés Oxidativo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas , Serina-Treonina Quinasas TOR , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
2.
Biofouling ; 28(3): 249-56, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22435742

RESUMEN

Settlement and metamorphosis of pediveliger larvae of Mytilus coruscus in response to natural biofilms was investigated in the laboratory. Pediveliger larvae settled and metamorphosed in response to biofilms and post-larval settlement and metamorphosis increased with biofilm age. The activity of the biofilm was positively correlated with biofilm age, dry weight, bacterial density and diatom density, but had no apparent relationship with chlorophyll a concentration. The change in bacterial community composition corresponding to biofilm age may explain differences in the age-dependent inducing activities of biofilms, which in turn may play an important role in larval settlement in this species.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Metamorfosis Biológica/fisiología , Mytilus/fisiología , Animales , Bacterias/genética , Bacterias/crecimiento & desarrollo , Diatomeas/crecimiento & desarrollo , Ecosistema , Larva/fisiología , Factores de Tiempo
3.
J Cell Biol ; 173(2): 279-89, 2006 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-16636147

RESUMEN

Loss of tuberin, the product of TSC2 gene, increases mammalian target of rapamycin (mTOR) signaling, promoting cell growth and tumor development. However, in cells expressing tuberin, it is not known how repression of mTOR signaling is relieved to activate this pathway in response to growth factors and how hamartin participates in this process. We show that hamartin colocalizes with hypophosphorylated tuberin at the membrane, where tuberin exerts its GTPase-activating protein (GAP) activity to repress Rheb signaling. In response to growth signals, tuberin is phosphorylated by AKT and translocates to the cytosol, relieving Rheb repression. Phosphorylation of tuberin at serines 939 and 981 does not alter its intrinsic GAP activity toward Rheb but partitions tuberin to the cytosol, where it is bound by 14-3-3 proteins. Thus, tuberin bound by 14-3-3 in response to AKT phosphorylation is sequestered away from its membrane-bound activation partner (hamartin) and its target GTPase (Rheb) to relieve the growth inhibitory effects of this tumor suppressor.


Asunto(s)
Membrana Celular/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular , Sustancias de Crecimiento/metabolismo , Células HeLa , Humanos , Microscopía Confocal , Modelos Biológicos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/farmacología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/análisis , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas ras/metabolismo
4.
Mol Carcinog ; 49(5): 429-39, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20146253

RESUMEN

The tuberous sclerosis complex 2 (Tsc2) gene product, tuberin, acts as a negative regulator of mTOR signaling, and loss of tuberin function leads to tumors of the brain, skin, kidney, heart, and lungs. Previous studies have shown that loss of tuberin function affects the stability and subcellular localization of the cyclin-dependent kinase inhibitor (CKI) p27, although the mechanism(s) by which tuberin modulates p27 stability has/have not been elucidated. Previous studies have also shown that AMP-activated protein kinase (AMPK), which functions in an energy-sensing pathway in the cell, becomes activated in the absence of tuberin. Here we show that in Tsc2-null tumors and cell lines, AMPK activation correlates with an increase in p27 levels, and inhibition of AMPK signaling decreases p27 levels in these cells. In addition, activation of AMPK led to phosphorylation of p27 at the conserved terminal threonine residue of murine p27 (T197) in both in vitro kinase assays and in cells. Phosphorylation of p27 at T197 led to increased interaction between p27 and 14-3-3 proteins and increased the protein stability of p27. Furthermore, activation of AMPK signaling promoted the interaction between p27 and 14-3-3 proteins and increased the stability of the p27 protein in a manner that was dependent on T197. These data identify a conserved mechanism for the regulation of p27 stability via phosphorylation at the terminal threonine (mT197/hT198) and binding of 14-3-3 proteins, which when AMPK is activated results in stabilization of the p27 protein.


Asunto(s)
Proteínas 14-3-3/metabolismo , Adenilato Quinasa/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Animales , Línea Celular , Humanos , Ratones , Fosforilación , Unión Proteica , Transducción de Señal
5.
Cancer Res ; 62(13): 3766-72, 2002 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12097287

RESUMEN

Unregulated proliferation of mesenchymal cells in leiomyomas, lipomas, hamartomas,and other diseases has been linked to the high mobility group (HMGA) family of DNA architectural proteins. HMGA genes are primarily expressed during embryonal development and silenced in adult tissues but can become reactivated in neoplasia as a result of chromosomal rearrangements. Although the genetic data suggesting a role for HMGA proteins in tumorigenesis are compelling, the biological role of these proteins in mesenchymal proliferation and differentiation is incompletely defined. Uterine myometria and spontaneous leiomyomas from the Eker rat, which carries a germ-line mutation in the tuberous sclerosis complex-2 (Tsc2) tumor suppressor gene, were analyzed for genetic defects in and expression of the Tsc2 and HMGA proteins. Eker leiomyomas exhibited a 50% incidence of loss of the wild-type Tsc2 allele and an almost uniform loss of protein expression, implicating loss of function of the Tsc2 gene in these tumors. Concomitantly, HMGA2 protein, which was completely absent in normal myometria, was expressed in 16 of 19 Eker leiomyomas. HMGA1 was expressed in both leiomyoma and normal myometria. No structural alterations were observed at the HMGA2 locus in either primary rat leiomyomas or leiomyoma-derived cell lines that expressed HMGA2. These data support a role for HMGA2 in the development of smooth muscle neoplasms and suggest HMGA2 expression is a point of convergence between the human disease and the Eker rat model. Furthermore, these data indicate that aberrant HMGA2 expression can result from dysfunction of the Tsc2 tumor suppressor gene, in the absence of structural alterations involving the HMGA2 locus.


Asunto(s)
Genes Supresores de Tumor/fisiología , Proteína HMGA2/biosíntesis , Leiomioma/genética , Proteínas Represoras/genética , Neoplasias Uterinas/genética , Animales , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Proteína HMGA2/genética , Leiomioma/metabolismo , Pérdida de Heterocigocidad , Ratas , Proteínas Represoras/biosíntesis , Translocación Genética , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor , Regulación hacia Arriba , Neoplasias Uterinas/metabolismo
6.
Nephrol Ther ; 2 Suppl 2: S119-22, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17373211

RESUMEN

Research into inherited cancer syndromes that involve defective tumor suppressor genes has increased our understanding of the genetic basis of this disease. Dramatic advances over the past decade have established the tuberous sclerosis complex 2 tumor suppressor gene (TSC2) as a key player in signal transduction pathways involved in the development of cancer. Importantly, the discovery of the functional link between TSC2 and the polycystic kidney disease 1 gene (PKD1) is beginning to build a foundation for understanding the heritable diseases associated with defects in each of these genes, namely, tuberous sclerosis complex and polycystic kidney disease. In this review, we summarize the latest findings about the functions of the TSC2 gene product, tuberin, and their implications for the development of cystic kidney disease.


Asunto(s)
Enfermedades Renales Poliquísticas/genética , Proteínas Supresoras de Tumor/genética , Genes Supresores de Tumor , Humanos , Transducción de Señal , Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa
7.
Cancer Res ; 68(16): 6496-506, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18701472

RESUMEN

Tuberin, the Tsc2 gene product, integrates the phosphatidylinositol 3-kinase/mitogen-activated protein kinase (mitogenic) and LKB1/AMP-activated protein kinase (AMPK; energy) signaling pathways, and previous independent studies have shown that loss of tuberin is associated with elevated AMPK signaling and altered p27 function. In Tsc2-null tumors and tumor-derived cells from Eker rats, we observed elevated AMPK signaling and concordant cytoplasmic mislocalization of p27. Cytoplasmic localization of p27 in Tsc2-null cells was reversible pharmacologically using inhibitors of the LKB1/AMPK pathway, and localization of p27 to the cytoplasm could be induced directly by activating AMPK physiologically (glucose deprivation) or genetically (constitutively active AMPK) in Tsc2-proficient cells. Furthermore, AMPK phosphorylated p27 in vitro on at least three sites including T170 near the nuclear localization signal, and T170 was shown to determine p27 localization in response to AMPK signaling. p27 functions in the nucleus to suppress cyclin-dependent kinase-2 (Cdk2) activity and has been reported to mediate an antiapoptotic function when localized to the cytoplasm. We found that cells with elevated AMPK signaling and cytoplasmic p27 localization exhibited elevated Cdk2 activity, which could be suppressed by inhibiting AMPK signaling. In addition, cells with elevated AMPK signaling and cytoplasmic p27 localization were resistant to apoptosis, which could be overcome by inhibition of AMPK signaling and relocalization of p27 to the nucleus. These data show that AMPK signaling determines the subcellular localization of p27, and identifies loss of integration of pathways controlling energy balance, the cell cycle, and apoptosis due to aberrant AMPK and p27 function as a feature of cells that have lost the Tsc2 tumor suppressor gene.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Citoplasma/metabolismo , Complejos Multienzimáticos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal , Proteínas Supresoras de Tumor/fisiología , Proteínas Quinasas Activadas por AMP , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Humanos , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación , Ratas , Fracciones Subcelulares , Proteína 2 del Complejo de la Esclerosis Tuberosa
8.
Toxicol Pathol ; 35(2): 233-41, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17366317

RESUMEN

Multiple renal tubular cell adenomas and atypical tubular hyperplasia were diagnosed in 2 high-dose and 1 mid-dose female Sprague-Dawley (Crl:CD (SD)IGS BR) rats from a 90-day toxicity study of an amino acid found in green tea. The tumors were bilateral multicentric adenomas accompanied by atypical foci of renal tubular hyperplasia in both kidneys of the 3 animals. Toxic tubular changes that typically accompany renal carcinogenesis were not seen in any of the other animals of the study, suggesting rather, an underlying germline mutation of a tumor suppressor gene in these three rats. The histological appearance of these tumors and short latency was reminiscent of the spontaneous lesions reported to arise in Sprague-Dawley rats in the Nihon rat model. Nihon rats develop kidney tumors as a result of a spontaneous mutation in the rat homologue of the Birt-Hogg-Dubé gene (Bhd). Frozen samples of liver from two tumor-bearing rats were assayed for germline alterations in the Bhd gene. The entire coding region (exons 3-13) of the Bhd gene was sequenced, and a guanine (nt106G) to adenine (nt106A) polymorphism was detected resulting in a glycine to arginine (G36R) substitution in both tumor-bearing animals. In the study animals, the frequency of the A-allele (adenine) was determined to be 27% (19/70). Interestingly, rats obtained from two other sources (n = 17) only carried the nt106G-allele, consistent with the published rat sequence for this gene. Genetic fingerprinting of microsatellite loci indicated that the rats had a shared genetic background. Laser capture microdissection (LCM) of the tumor cells demonstrated a loss of heterozygosity in the Bhd gene in neoplastic cells of one of the two animals. Taken together, these data suggest that the tumors observed in these animals arose spontaneously as a result of a shared genetic susceptibility leading to the development of renal tubular neoplasms.


Asunto(s)
Adenoma/inducido químicamente , Glutamatos/toxicidad , Neoplasias Renales/inducido químicamente , Túbulos Renales/patología , Adenoma/genética , Adenoma/patología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal/genética , Hiperplasia/inducido químicamente , Hiperplasia/genética , Hiperplasia/patología , Neoplasias Renales/genética , Neoplasias Renales/patología , Túbulos Renales/efectos de los fármacos , Pérdida de Heterocigocidad/genética , Masculino , Proteínas/genética , Ratas , Ratas Sprague-Dawley
9.
Proc Natl Acad Sci U S A ; 102(24): 8644-9, 2005 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-15937110

RESUMEN

Gene-environment interactions are important determinants of cancer risk. Traditionally, gene-environment interactions are thought to contribute to tumor-suppressor-gene penetrance by facilitating or inhibiting the acquisition of additional somatic mutations required for tumorigenesis. Here, we demonstrate that a distinctive type of gene-environment interaction can occur during development to enhance the penetrance of a tumor-suppressor-gene defect in the adult. Using rats carrying a germ-line defect in the tuberous sclerosis complex 2 (Tsc-2) tumor-suppressor gene predisposed to uterine leiomyomas, we show that an early-life exposure to diethylstilbestrol during development of the uterus increased tumor-suppressor-gene penetrance from 65% to >90% and tumor multiplicity and size in genetically predisposed animals, but it failed to induce tumors in wild-type rats. This exposure was shown to impart a hormonal imprint on the developing uterine myometrium, causing an increase in expression of estrogen-responsive genes before the onset of tumors. Loss of function of the normal Tsc-2 allele remained the rate-limiting event for tumorigenesis; however, tumors that developed in exposed animals displayed an enhanced proliferative response to steroid hormones relative to tumors that developed in unexposed animals. These data suggest that exposure to environmental factors during development can permanently reprogram normal physiological tissue responses and thus lead to increased tumor-suppressor-gene penetrance in genetically susceptible individuals.


Asunto(s)
Dietilestilbestrol/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Predisposición Genética a la Enfermedad/genética , Leiomioma/genética , Penetrancia , Efectos Tardíos de la Exposición Prenatal , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética , Animales , Western Blotting , Femenino , Hormonas Esteroides Gonadales/metabolismo , Inmunohistoquímica , Leiomioma/metabolismo , Miometrio/metabolismo , Miometrio/patología , Embarazo , Ratas , Ratas Mutantes , Proteínas Represoras/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA