Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Prev Res (Phila) ; 17(2): 59-75, 2024 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-37956420

RESUMEN

Risk and outcome of acute promyelocytic leukemia (APL) are particularly worsened in obese-overweight individuals, but the underlying molecular mechanism is unknown. In established mouse APL models (Ctsg-PML::RARA), we confirmed that obesity induced by high-fat diet (HFD) enhances leukemogenesis by increasing penetrance and shortening latency, providing an ideal model to investigate obesity-induced molecular events in the preleukemic phase. Surprisingly, despite increasing DNA damage in hematopoietic stem cells (HSC), HFD only minimally increased mutational load, with no relevant impact on known cancer-driving genes. HFD expanded and enhanced self-renewal of hematopoietic progenitor cells (HPC), with concomitant reduction in long-term HSCs. Importantly, linoleic acid, abundant in HFD, fully recapitulates the effect of HFD on the self-renewal of PML::RARA HPCs through activation of peroxisome proliferator-activated receptor delta, a central regulator of fatty acid metabolism. Our findings inform dietary/pharmacologic interventions to counteract obesity-associated cancers and suggest that nongenetic factors play a key role. PREVENTION RELEVANCE: Our work informs interventions aimed at counteracting the cancer-promoting effect of obesity. On the basis of our study, individuals with a history of chronic obesity may still significantly reduce their risk by switching to a healthier lifestyle, a concept supported by evidence in solid tumors but not yet in hematologic malignancies. See related Spotlight, p. 47.


Asunto(s)
Leucemia Promielocítica Aguda , PPAR delta , Animales , Ratones , Catepsina G , Dieta Alta en Grasa/efectos adversos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Obesidad/complicaciones , Proteínas de Fusión Oncogénica/genética , PPAR delta/uso terapéutico
2.
Cancer Res ; 83(13): 2155-2170, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37133448

RESUMEN

Metastatic breast cancer has a poor prognosis and is largely considered incurable. A better understanding of the molecular determinants of breast cancer metastasis could facilitate development of improved prevention and treatment strategies. We used lentiviral barcoding coupled to single-cell RNA sequencing to trace clonal and transcriptional evolution during breast cancer metastasis and showed that metastases derive from rare prometastatic clones that are underrepresented in primary tumors. Both low clonal fitness and high metastatic potential were independent of clonal origin. Differential expression and classification analyses revealed that the prometastatic phenotype was acquired by rare cells characterized by the concomitant hyperactivation of extracellular matrix remodeling and dsRNA-IFN signaling pathways. Notably, genetic silencing of key genes in these pathways (KCNQ1OT1 or IFI6, respectively) significantly impaired migration in vitro and metastasis in vivo, with marginal effects on cell proliferation and tumor growth. Gene expression signatures derived from the identified prometastatic genes predict metastatic progression in patients with breast cancer, independently of known prognostic factors. This study elucidates previously unknown mechanisms of breast cancer metastasis and provides prognostic predictors and therapeutic targets for metastasis prevention. SIGNIFICANCE: Transcriptional lineage tracing coupled with single-cell transcriptomics defined the transcriptional programs underlying metastatic progression in breast cancer, identifying prognostic signatures and prevention strategies.


Asunto(s)
Perfilación de la Expresión Génica , Transducción de Señal , Humanos , Línea Celular Tumoral , Transducción de Señal/genética , Pronóstico , Matriz Extracelular/genética , Metástasis de la Neoplasia , Regulación Neoplásica de la Expresión Génica
3.
Int J Mol Sci ; 23(11)2022 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-35682953

RESUMEN

Metastatic disease represents the primary cause of breast cancer (BC) mortality, yet it is still one of the most enigmatic processes in the biology of this tumor. Metastatic progression includes distinct phases: invasion, intravasation, hematogenous dissemination, extravasation and seeding at distant sites, micro-metastasis formation and metastatic outgrowth. Whole-genome sequencing analyses of primary BC and metastases revealed that BC metastatization is a non-genetically selected trait, rather the result of transcriptional and metabolic adaptation to the unfavorable microenvironmental conditions which cancer cells are exposed to (e.g., hypoxia, low nutrients, endoplasmic reticulum stress and chemotherapy administration). In this regard, the latest multi-omics analyses unveiled intra-tumor phenotypic heterogeneity, which determines the polyclonal nature of breast tumors and constitutes a challenge for clinicians, correlating with patient poor prognosis. The present work reviews BC classification and epidemiology, focusing on the impact of metastatic disease on patient prognosis and survival, while describing general principles and current in vitro/in vivo models of the BC metastatic cascade. The authors address here both genetic and phenotypic intrinsic heterogeneity of breast tumors, reporting the latest studies that support the role of the latter in metastatic spreading. Finally, the review illustrates the mechanisms underlying adaptive stress responses during BC metastatic progression.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Metástasis de la Neoplasia
4.
Cell Death Differ ; 29(12): 2429-2444, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35739253

RESUMEN

Aging is accompanied by the progressive decline in tissue regenerative capacity and functions of resident stem cells (SCs). Underlying mechanisms, however, remain unclear. Here we show that, during chronological aging, self-renewing mitoses of mammary SCs (MaSCs) are preferentially asymmetric and that their progeny divides less frequently, leading to decreased number of MaSCs and reduced regenerative potential. Underlying mechanisms are investigated in the p66Shc-/- mouse, which exhibits several features of delayed aging, including reduced involution of the mammary gland (MG). p66Shc is a mitochondrial redox sensor that activates a specific p53 transcriptional program, in which the aging-associated p44 isoform of p53 plays a pivotal role. We report here that aged p66Shc-/- MaSCs show increased symmetric divisions, increased proliferation and increased regenerative potential, to an extent reminiscent of young wild-type (WT) MaSCs. Mechanistically, we demonstrate that p66Shc, together with p53: (i) accumulates in the aged MG, (ii) sustains expression of the cell polarity determinant mInscuteable and, concomitantly, (iii) down-regulates critical cell cycle genes (e.g.,: Cdk1 and Cyclin A). Accordingly, overexpression of p53/p44 increases asymmetric divisions and decreases proliferation of young WT MaSCs in a p66Shc-dependent manner and overexpression of mInsc restores WT-like levels of asymmetric divisions in aged p66Shc-/- MaSCs. Notably, deletion of p66Shc has negligible effects in young MaSCs and MG development. These results demonstrate that MG aging is due to aberrant activation of p66Shc, which induces p53/p44 signaling, leading to failure of symmetric divisions, decreased proliferation and reduced regenerative potential of MaSCs.


Asunto(s)
Glándulas Mamarias Animales , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Células Madre , Proteína p53 Supresora de Tumor , Animales , Ratones , Proliferación Celular , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Glándulas Mamarias Animales/citología
6.
Aging Cell ; 15(3): 446-54, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26968134

RESUMEN

Oxidative stress and telomere attrition are considered the driving factors of aging. As oxidative damage to telomeric DNA favors the erosion of chromosome ends and, in turn, telomere shortening increases the sensitivity to pro-oxidants, these two factors may trigger a detrimental vicious cycle. To check whether limiting oxidative stress slows down telomere shortening and related progeria, we have investigated the effect of p66SHC deletion, which has been shown to reduce oxidative stress and mitochondrial apoptosis, on late-generation TERC (telomerase RNA component)-deficient mice having short telomeres and reduced lifespan. Double mutant (TERC(-/-) p66SHC(-/-) ) mice were generated, and their telomere length, fertility, and lifespan investigated in different generations. Results revealed that p66SHC deletion partially rescues sterility and weight loss, as well as organ atrophy, of TERC-deficient mice, but not their short lifespan and telomere erosion. Therefore, our data suggest that p66SHC-mediated oxidative stress and telomere shortening synergize in some tissues (including testes) to accelerate aging; however, early mortality of late-generation mice seems to be independent of any link between p66SHC-mediated oxidative stress and telomere attrition.


Asunto(s)
Fertilidad/genética , Eliminación de Gen , Longevidad , Progeria/genética , Progeria/patología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Telomerasa/deficiencia , Animales , Daño del ADN , Genotipo , Ratones Endogámicos C57BL , Estrés Oxidativo , Fenotipo , ARN/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Análisis de Supervivencia , Telomerasa/metabolismo , Acortamiento del Telómero , Pérdida de Peso
7.
Aging Dis ; 6(2): 95-108, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25821638

RESUMEN

Oxidative stress induced by reactive oxygen species (ROS) increases during lifespan and is involved in aging processes. The p66Shc adaptor protein is a master regulator of oxidative stress response in mammals. Ablation of p66Shc enhances oxidative stress resistance both in vitro and in vivo. Most importantly, it has been demonstrated that its deletion retards aging in mice. Recently, new insights in the molecular mechanisms involving p66Shc and the p53 tumor suppressor genes were given: a specific p66Shc/p53 transcriptional regulation pathway was uncovered as determinant in oxidative stress response and, likely, in aging. p53, in a p66Shc-dependent manner, negatively downregulates the expression of 200 genes which are involved in the G2/M transition of mitotic cell cycle and are downregulated during physiological aging. p66Shc modulates the response of p53 by activating a p53 isoform (p44/p53, also named Delta40p53). Based on these latest results, several developments are expected in the future, as the generation of animal models to study aging and the evaluation of the use of the p53/p66Shc target genes as biomarkers in aging related diseases. The aim of this review is to investigate the conservation of the p66Shc and p53 role in oxidative stress between fish and mammals. We propose to approach this study trough a new model organism, the annual fish Nothobranchius furzeri, that has been demonstrated to develop typical signs of aging, like in mammals, including senescence, neurodegeneration, metabolic disorders and cancer.

8.
J Gerontol A Biol Sci Med Sci ; 69(10): 1177-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24336818

RESUMEN

The signaling molecule p66Shc is often described as a longevity protein. This conclusion is based on a single life span study that used a small number of mice. The purpose of the present studies was to measure life span in a sufficient number of mice to determine if longevity is altered in mice with decreased Shc levels (ShcKO). Studies were completed at UC Davis and the European Institute of Oncology (EIO). At UC Davis, male C57BL/6J WT and ShcKO mice were fed 5% or 40% calorie-restricted (CR) diets. In the 5% CR group, there was no difference in survival curves between genotypes. There was also no difference between genotypes in prevalence of neoplasms or other measures of end-of-life pathology. At 40% calorie restriction group, 70th percentile survival was increased in ShcKO, while there were no differences between genotypes in median or subsequent life span measures. At EIO, there was no increase in life span in ShcKO male or female mice on C57BL/6J, 129Sv, or hybrid C57BL/6J-129Sv backgrounds. These studies indicate that p66Shc is not a longevity protein. However, additional studies are needed to determine the extent to which Shc proteins may influence the onset and severity of specific age-related diseases.


Asunto(s)
Longevidad , Proteínas Adaptadoras de la Señalización Shc/fisiología , Crianza de Animales Domésticos , Animales , Restricción Calórica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de la Especie , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
10.
Methods Enzymol ; 528: 99-110, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23849861

RESUMEN

Reactive oxygen species (ROS), mainly originated from mitochondrial respiration, are critical inducers of oxidative damage and involved in tissue dysfunction. It is not clear, however, whether oxidative stress is the result of an active gene program or it is the by-product of physiological processes. Recent findings demonstrate that ROS are produced by mitochondria in a controlled way through specialized enzymes, including p66Shc, and take part in cellular process aimed to ensure adaptation and fitness. Therefore, genes generating specifically ROS are selected determinants of life span in response to different environmental conditions.


Asunto(s)
Mitocondrias Hepáticas/efectos de los fármacos , Especies Reactivas de Oxígeno/agonistas , Proteínas Adaptadoras de la Señalización Shc/farmacología , Animales , Apoptosis/efectos de los fármacos , Regulación de la Temperatura Corporal/fisiología , Ciclosporina/farmacología , Técnicas Electroquímicas , Escherichia coli/genética , Escherichia coli/metabolismo , Etilmaleimida/farmacología , Humanos , Ratones , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Oxidación-Reducción , Estrés Oxidativo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Adaptadoras de la Señalización Shc/biosíntesis , Proteínas Adaptadoras de la Señalización Shc/genética , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
11.
Aging Cell ; 12(3): 435-45, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23448364

RESUMEN

Oxidative stress is a determining factor of cellular senescence and aging and a potent inducer of the tumour-suppressor p53. Resistance to oxidative stress correlates with delayed aging in mammals, in the absence of accelerated tumorigenesis, suggesting inactivation of selected p53-downstream pathways. We investigated p53 regulation in mice carrying deletion of p66, a mutation that retards aging and confers cellular resistance and systemic resistance to oxidative stress. We identified a transcriptional network of ~200 genes that are repressed by p53 and encode for determinants of progression through mitosis or suppression of senescence. They are selectively down-regulated in cultured fibroblasts after oxidative stress, and, in vivo, in proliferating tissues and during physiological aging. Selectivity is imposed by p66 expression and activation of p44/p53 (also named Delta40p53), a p53 isoform that accelerates aging and prevents mitosis after protein damage. p66 deletion retards aging and increases longevity of p44/p53 transgenic mice. Thus, oxidative stress activates a specific p53 transcriptional response, mediated by p44/p53 and p66, which regulates cellular senescence and aging.


Asunto(s)
Senescencia Celular/fisiología , Estrés Oxidativo/fisiología , Fragmentos de Péptidos/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Células Cultivadas , Senescencia Celular/genética , Hepatocitos/metabolismo , Longevidad , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Adaptadoras de la Señalización Shc/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Timo/metabolismo , Factores de Transcripción , Transcripción Genética , Proteína p53 Supresora de Tumor/genética
12.
Aging Cell ; 12(2): 177-83, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23237310

RESUMEN

Upon oxidative challenge the genome accumulates adducts and breaks that activate the DNA damage response to repair, arrest, or eliminate the damaged cell. Thus, reactive oxygen species (ROS) generated by endogenous oxygen metabolism are thought to affect mutation frequency. However, few studies determined the mutation frequency when oxidative stress is reduced. To test whether in vivo spontaneous mutation frequency is altered in mice with reduced oxidative stress and cell death rate, we crossed p66Shc knockout (p66KO) mice, characterized by reduced intracellular concentration of ROS and by impaired apoptosis, with a transgenic line harboring multiple copies of the lacZ mutation reporter gene as part of a plasmid that can be recovered from organs into Escherichia coli to measure mutation rate. Liver and small intestine from 2- to 24-month-old, lacZ (p66Shc+/+) and lacZp66KO mice, were investigated revealing no difference in overall mutation frequency but a significant increase in the frequency of size-change mutations in the intestine of lacZp66KO mice. This difference was further increased upon irradiation of mice with X-ray. In addition, we found that knocking down cyclophilin D, a gene that facilitates mitochondrial apoptosis acting downstream of p66Shc, increased the size-change mutation frequency in small intestine. Size-change mutations also accumulated in death-resistant embryonic fibroblasts from lacZp66KO mice treated with H2 O2 . These results indicate that p66Shc plays a role in the accumulation of DNA rearrangements and suggest that p66Shc functions to clear damaged cells rather than affect DNA metabolism.


Asunto(s)
Intestino Delgado/metabolismo , Operón Lac , Hígado/metabolismo , Tasa de Mutación , Proteínas Adaptadoras de la Señalización Shc/genética , Animales , Peptidil-Prolil Isomerasa F , Ciclofilinas/genética , Ciclofilinas/metabolismo , Escherichia coli/genética , Rayos gamma , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Peróxido de Hidrógeno/farmacología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/efectos de la radiación , Hígado/efectos de los fármacos , Hígado/efectos de la radiación , Ratones , Ratones Noqueados , Mutación , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Adaptadoras de la Señalización Shc/deficiencia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Transgenes
13.
Blood ; 120(22): 4391-9, 2012 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-23033271

RESUMEN

Although intrinsic apoptosis defects are causal to the extended survival of chronic lymphocytic leukemia (CLL) B cells, several lines of evidence support a contribution of the peripheral lymphoid organs and BM microenvironment to the extended lifespan of leukemic B cells. Lymphocyte trafficking is controlled by homing signals provided by stromal cell-derived chemokines and egress signals provided by sphingosine-1-phosphate (S1P). In the present study, we show that expression of S1P1, the S1P receptor responsible for lymphocyte egress, is selectively reduced in CLL B cells with unmutated IGHV. Expression of S1P2, which controls B-cell homeostasis, is also impaired in CLL B cells but independently of the IGHV mutational status. We provide evidence herein that p66Shc, a Shc adaptor family member the deficiency of which is implicated in the apoptosis defects of CLL B cells, controls S1P1 expression through its pro-oxidant activity. p66Shc also controls the expression of the homing receptor CCR7, which opposes S1P1 by promoting lymphocyte retention in peripheral lymphoid organs. The results of the present study provide insights into the regulation of S1P1 expression in B cells and suggest that defective egress caused by impaired S1P1 expression contributes to the extended survival of CLL B cells by prolonging their residency in the prosurvival niche of peripheral lymphoid organs.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/genética , Receptores de Lisoesfingolípidos/genética , Proteínas Adaptadoras de la Señalización Shc/fisiología , Adulto , Animales , Femenino , Regulación de la Expresión Génica/genética , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/mortalidad , Masculino , Ratones , Ratones Noqueados , Oxidantes/metabolismo , Pronóstico , Receptores de Lisoesfingolípidos/fisiología , Proteínas Adaptadoras de la Señalización Shc/genética , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Células Tumorales Cultivadas
14.
Aging Cell ; 11(1): 162-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22081964

RESUMEN

Deletion of the p66(Shc) gene results in lean and healthy mice, retards aging, and protects from aging-associated diseases, raising the question of why p66(Shc) has been selected, and what is its physiological role. We have investigated survival and reproduction of p66(Shc)-/- mice in a population living in a large outdoor enclosure for a year, subjected to food competition and exposed to winter temperatures. Under these conditions, deletion of p66(Shc) was strongly counterselected. Laboratory studies revealed that p66(Shc)-/- mice have defects in fat accumulation, thermoregulation, and reproduction, suggesting that p66(Shc) has been evolutionarily selected because of its role in energy metabolism. These findings imply that the health impact of targeting aging genes might depend on the specific energetic niche and caution should be exercised against premature conclusions regarding gene functions that have only been observed in protected laboratory conditions.


Asunto(s)
Envejecimiento/genética , Longevidad/genética , Proteínas Adaptadoras de la Señalización Shc/genética , Envejecimiento/metabolismo , Animales , Evolución Biológica , Regulación de la Temperatura Corporal/genética , Metabolismo Energético/genética , Femenino , Aptitud Genética/genética , Heterocigoto , Homocigoto , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Noqueados , Estaciones del Año , Proteínas Adaptadoras de la Señalización Shc/deficiencia , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
15.
Aging Cell ; 10(1): 55-65, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21040401

RESUMEN

Longevity of a p66Shc knockout strain (ShcP) was previously attributed to increased stress resistance and altered mitochondria. Microarrays of ShcP tissues indicated alterations in insulin signaling. Consistent with this observation, ShcP mice were more insulin sensitive and glucose tolerant at organismal and tissue levels, as was a novel p66Shc knockout (ShcL). Increasing and decreasing Shc expression in cell lines decreased and increased insulin sensitivity, respectively - consistent with p66Shc's function as a repressor of insulin signaling. However, differences between the two p66Shc knockout strains were also observed. ShcL mice were fatter and susceptible to fatty diets, and their fat was more insulin sensitive than controls. On the other hand, ShcP mice were leaner and resisted fatty diets, and their adipose was less insulin sensitive than controls. ShcL and ShcP strains are both highly inbred on the C57Bl/6 background, so we investigated gene expression at the Shc locus, which encodes three isoforms, p66, p52, and p46. Isoform p66 is absent in both strains; thus, the remaining difference to which to attribute the 'lean' phenotype is expression of the other two isoforms. ShcL mice have a precise deletion of p66Shc and normal expression of p52 and p46Shc isoforms in all tissues; thus, a simple deletion of p66Shc results in a 'fat' phenotype. However, ShcP mice in addition to p66Shc deletion have a fourfold increase in p46Shc expression in white fat. Thus, p46Shc overexpression in fat, rather than p66Shc deletion, is the likely cause of decreased adiposity and reduced insulin sensitivity in the fat of ShcP mice, which has implications for the longevity of the strain.


Asunto(s)
Adiposidad/genética , Insulina/metabolismo , Isoformas de Proteínas/genética , Proteínas Adaptadoras de la Señalización Shc/genética , Transducción de Señal/fisiología , Animales , Células Cultivadas , Grasas de la Dieta/metabolismo , Femenino , Sitios Genéticos/fisiología , Glucosa/metabolismo , Resistencia a la Insulina/genética , Longevidad/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/genética , Isoformas de Proteínas/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Delgadez/genética , Delgadez/metabolismo
16.
J Biol Chem ; 285(2): 1153-65, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19892704

RESUMEN

A decrease in reactive oxygen species (ROS) production has been associated with extended life span in animal models of longevity. Mice deficient in the p66Shc gene are long-lived, and their cells are both resistant to oxidative stress and produce less ROS. Our microarray analysis of p66Shc(-/-) mouse tissues showed alterations in transcripts involved in heme and superoxide production and insulin signaling. Thus, we carried out analysis of ROS production by NADPH oxidase (PHOX) in macrophages of control and p66Shc knock-out mice. p66Shc(-/-) mice had a 40% reduction in PHOX-dependent superoxide production. To confirm whether the defect in superoxide production was a direct consequence of p66Shc deficiency, p66Shc was knocked down with siRNA in the macrophage cell line RAW264, and a 30% defect in superoxide generation was observed. The pathway of PHOX-dependent superoxide generation was investigated. PHOX protein levels were not decreased in mutant macrophages; however, the rate and extent of phosphorylation of p47phox was decreased in mutants, as was membrane translocation of the complex. Consistently, phosphorylation of protein kinase Cdelta, Akt, and ERK (the kinases responsible for phosphorylation of p47phox) was decreased. Thus, p66Shc deficiency causes a defect in activation of the PHOX complex that results in decreased superoxide production. p66Shc-deficient mice have recently been observed to be resistant to atherosclerosis and to oxidant injury in kidney and brain. Because phagocyte-derived superoxide is often a component of oxidant injury and inflammation, we suggest that the decreased superoxide production by PHOX in p66Shc-deficient mice could contribute significantly to their relative protection from oxidant injury and consequent longevity.


Asunto(s)
Longevidad , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Proteínas Adaptadoras de la Señalización Shc , Superóxidos/metabolismo , Animales , Línea Celular , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , NADPH Oxidasas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
17.
Aging (Albany NY) ; 1(6): 503-10, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-20157533

RESUMEN

Oxygen metabolism is thought to impact on aging through the formation of reactive oxygen species (ROS) that are supposed to damage biological molecules. The study of p66(Shc), a crucial regulator of ROS level involved in aging dysfunction, suggests that the incidence of degenerative disease and longevity are determined by a specific signaling function of ROS other than their unspecific damaging property.


Asunto(s)
Envejecimiento/metabolismo , Regulación de la Expresión Génica/fisiología , Longevidad/genética , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Envejecimiento/genética , Animales , Humanos , Longevidad/fisiología , Mutación , Oxígeno/metabolismo , Isoformas de Proteínas , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
18.
Endothelium ; 15(5-6): 276-87, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19065319

RESUMEN

Previous studies showed that p66(Shc-/-) mice on a very-high-fat diet (HFD) had reduced oxidative stress, foam cell, and early atherosclerotic lesion formation. Here, the authors have used hypercholesterolemic apolipoprotein E (ApoE(-/-)) mice to investigate the role of p66Shc deletion in advanced atheroma. The authors generated mice deficient of both ApoE and p66Shc genes (ApoE(-/-) /p66(Shc-/-)). They used microsatellite polymerase chain reaction (PCR) analysis to analyze the genetic background and considered only animals with a constant percentages of C57B6L and 129SV background strands (it was obtained the 50.3% +/- 6.4% of C57B6L background). Computer-assisted analysis revealed that advanced atherosclerotic lesions in ApoE(-/-)/p66(Shc+/+) were significantly larger than those observed in ApoE(-/-)/p66(Shc-/-). Accordingly, the lipid-laden macrophage foam cells and oxidation-specific epitopes in ApoE(-/-)/p66(shc+/+) HFD-treated groups were higher than those observed in normal diet (ND)-treated groups. Thus, p66(Shc-/-) plays an important protective role also against advanced atherosclerotic lesion formation. Finally, the authors have used microarray to investigate major changes in gene expression in aortas of mice with ApoE(-/-)/p66(Shc-/-) background treated with a very HFD in comparison to ApoE(-/-)/p66(Shc+/+) (these data have been confirmed by by real-time PCR and immunohistochemistry). DAVID (Database for Annotation, Visualization and Integrated Discovery) analysis revealed that CD36 antigen (CD36), tissue inhibitor of metalloproteinase 2 (TIMP2), apolipoprotein E (ApoE), acetyl-coenzyme A acetyltransferase 1 (ACAT1), and thrombospondin 1 (THBS1) can be involved in p66 deletion-dependent vascular protection through the adipocytokine/lipid signaling pathway.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Proteínas Adaptadoras de la Señalización Shc/genética , Adipoquinas/genética , Adipoquinas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aorta/fisiopatología , Apolipoproteínas E/metabolismo , Aterosclerosis/fisiopatología , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Células Espumosas/metabolismo , Células Espumosas/patología , Alimentos Formulados/efectos adversos , Eliminación de Gen , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , Genotipo , Hipercolesterolemia/fisiopatología , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
19.
J Biol Chem ; 283(49): 34283-93, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18838380

RESUMEN

Reactive oxygen species (ROS) and insulin signaling in the adipose tissue are critical determinants of aging and age-associated diseases. It is not clear, however, if they represent independent factors or they are mechanistically linked. We investigated the effects of ROS on insulin signaling using as model system the p66(Shc)-null mice. p66(Shc) is a redox enzyme that generates mitochondrial ROS and promotes aging in mammals. We report that insulin activates the redox enzyme activity of p66(Shc) specifically in adipocytes and that p66(Shc)-generated ROS regulate insulin signaling through multiple mechanisms, including AKT phosphorylation, Foxo localization, and regulation of selected insulin target genes. Deletion of p66(Shc) resulted in increased mitochondrial uncoupling and reduced triglyceride accumulation in adipocytes and in vivo increased metabolic rate and decreased fat mass and resistance to diet-induced obesity. In addition, p66(Shc-/-) mice showed impaired thermo-insulation. These findings demonstrate that p66(Shc)-generated ROS regulate the effect of insulin on the energetic metabolism in mice and suggest that intracellular oxidative stress might accelerate aging by favoring fat deposition and fat-related disorders.


Asunto(s)
Estrés Oxidativo , Proteínas Adaptadoras de la Señalización Shc/fisiología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Oxidación-Reducción , Oxígeno/metabolismo , Fosforilación , Especies Reactivas de Oxígeno , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
20.
Nat Rev Mol Cell Biol ; 8(9): 722-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17700625

RESUMEN

The reactive oxygen species that are generated by mitochondrial respiration, including hydrogen peroxide (H2O2), are potent inducers of oxidative damage and mediators of ageing. It is not clear, however, whether oxidative stress is the result of a genetic programme or the by-product of physiological processes. Recent findings demonstrate that a fraction of mitochondrial H2O2, produced by a specialized enzyme as a signalling molecule in the pathway of apoptosis, induces intracellular oxidative stress and accelerates ageing. We propose that genes that control H2O2 production are selected determinants of lifespan.


Asunto(s)
Envejecimiento/metabolismo , Peróxido de Hidrógeno/metabolismo , Transducción de Señal/fisiología , Envejecimiento/genética , Animales , Humanos , Longevidad , Estrés Oxidativo/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...