Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Brain ; 10(1): 23, 2017 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-28683812

RESUMO

Mechano growth factor (MGF) is a splice variant of IGF-1 first described in skeletal muscle. MGF induces muscle cell proliferation in response to muscle stress and injury. In control mice we found endogenous expression of MGF in neurogenic areas of the brain and these levels declined with age. To better understand the role of MGF in the brain, we used transgenic mice that constitutively overexpressed MGF from birth. MGF overexpression significantly increased the number of BrdU+ proliferative cells in the dentate gyrus (DG) of the hippocampus and subventricular zone (SVG). Although MGF overexpression increased the overall rate of adult hippocampal neurogenesis at the proliferation stage it did not alter the distribution of neurons at post-mitotic maturation stages. We then used the lac-operon system to conditionally overexpress MGF in the mouse brain beginning at 1, 3 and 12 months with histological and behavioral observation at 24 months of age. With conditional overexpression there was an increase of BrdU+ proliferating cells and BrdU+ differentiated mature neurons in the olfactory bulbs at 24 months when overexpression was induced from 1 and 3 months of age but not when started at 12 months. This was associated with preserved olfactory function. In vitro, MGF increased the size and number of neurospheres harvested from SVZ-derived neural stem cells (NSCs). These findings indicate that MGF overexpression increases the number of neural progenitor cells and promotes neurogenesis but does not alter the distribution of adult newborn neurons at post-mitotic stages. Maintaining youthful levels of MGF may be important in reversing age-related neuronal loss and brain dysfunction.


Assuntos
Envelhecimento/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Neurogênese , Splicing de RNA/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Camundongos Transgênicos , Mitógenos/farmacologia , Mitose/efeitos dos fármacos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Bulbo Olfatório/efeitos dos fármacos , Bulbo Olfatório/metabolismo
2.
Aging (Albany NY) ; 7(12): 1212-23, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26796640

RESUMO

Bone marrow transplantation is used to examine survival, hematopoietic stem cell function and pathology in recipients of young and old wild type bone marrow derived stem cells (BMDSCs) as well as cells from p53-based models of premature aging. There is no difference in the long term survival of recipients of 8 week-old p53+/m donor cells compared to recipients of 8 week-old wild-type (WT) donor cells (70 weeks) or of recipients of 16-18 weeks-old donor cells from either p53+/m or WT mice. There is shorter survival in recipients of older versus younger WT donor bone marrow, but the difference is only significant when comparing 8 and 18 week-old donors. In the p44-based model, short term survival/engraftment is significantly reduced in recipients of 11 month-old p44 donor cells compared to 4 week-old p44 or wild type donor cells of either age; mid-life survival at 40 weeks is also significantly less in recipients of p44 cells. BMDSCs are readily detectable within recipient bone marrow, lymph node, intestinal villi and liver sinusoids, but not in epithelial derived cells. These results indicate that recipients of young BMDSCs may survive longer than recipients of old bone marrow, but the difference is marginal at best.


Assuntos
Envelhecimento/fisiologia , Transplante de Medula Óssea , Medula Óssea/efeitos da radiação , Animais , Células da Medula Óssea/fisiologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fatores Sexuais , Fatores de Transcrição , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
J Invest Dermatol ; 134(3): 791-800, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24037342

RESUMO

The TP53 gene encodes 12 distinct isoforms, some of which can alter p53 activity in the absence of genomic alteration. Endogenous p53 isoforms have been identified in cancers; however, the function of these isoforms remains unclear. In melanoma, the frequency of TP53 mutations is relatively low compared with other cancers, suggesting that these isoforms may have a larger role in regulating TP53 activity. We hypothesized that p53 function and therefore cell fate might be altered by the presence of Δ40p53, an embryonic isoform missing the first 40 N-terminal amino acids of the full-length protein including the transactivation and Mdm2-binding domains. To test this hypothesis, we transduced tumor and normal cells with a lentivirus encoding Δ40p53. We found that exogenous Δ40p53 caused apoptosis and increased the levels of endogenous, activated p53 in both cancerous and non-cancerous cells, which led to significant levels of cell death, particularly in cancer cells. Activated p53 molecules formed nuclear heterotetramers with Δ40p53 and altered downstream p53 transcription target levels including p53-induced protein with death domain and cyclin-dependent kinase inhibitor, p21. Δ40p53 altered the promoter occupancy of these downstream p53 target genes in such a way that it shifted cell fate toward apoptosis and away from cell cycle arrest. We show that tumor suppression by p53 can occur via an alternate route that relies on its interaction with Δ40p53.


Assuntos
Apoptose/fisiologia , Melanoma/genética , Neoplasias Cutâneas/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Neoplasias Encefálicas , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Fibroblastos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Glioblastoma , Humanos , Melanócitos/citologia , Melanoma/patologia , Camundongos , Regiões Promotoras Genéticas/fisiologia , Estrutura Terciária de Proteína , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/química
4.
Aging Cell ; 13(3): 449-56, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24341977

RESUMO

p44 is a short isoform of p53 with 'longevity-assurance' activity. Overexpression of p44 in the mouse (p44(+/+) transgenic mice) causes a progeroid phenotype that mimics an accelerated form of aging. The phenotype includes abnormal phosphorylation of the microtubule-binding protein tau, synaptic deficits, and cognitive decline. Genetic engineering demonstrated that the phosphorylation status of tau acts upstream of the synaptic deficits. Here, we provide evidence that p44 promotes the phosphorylation of tau in the mouse. Specifically, we show that p44 binds to the promoter of tau kinases Dyrk1A, GSK3ß, Cdk5, p35, and p39 and activates their transcription. The upregulation of the above kinases is followed by increased phosphorylation of tau. Finally, we show that p44 is preferentially found in the nucleus and that its levels increase with age in the mouse brain. Taken together, these results suggest that an imbalance in the p53:p44 ratio might be involved with the altered tau metabolism that characterizes aging.


Assuntos
Doença de Alzheimer/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas tau/metabolismo , Fatores Etários , Animais , Camundongos , Camundongos Transgênicos , Fosforilação , Isoformas de Proteínas , Transdução de Sinais , Fatores de Transcrição
5.
Mol Ther Nucleic Acids ; 2: e133, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24253258

RESUMO

Development of RNAi-based therapeutics has the potential to revolutionize treatment options for a range of human diseases. However, as with gene therapy, a major barrier to progress is the lack of methods to achieve and measure efficient delivery for systemic administration. We have developed a positive-readout pharmacodynamic transgenic reporter mouse model allowing noninvasive real-time assessment of siRNA activity. The model combines a luciferase reporter gene under the control of regulatory elements from the lac operon of Escherichia coli. Introduction of siRNA targeting lac repressor results in increased luciferase expression in cells where siRNA is biologically active. Five founder luciferase-expressing and three founder Lac-expressing lines were generated and characterized. Mating of ubiquitously expressing luciferase and lac lines generated progeny in which luciferase expression was significantly reduced compared with the parental line. Administration of isopropyl ß-D-1-thiogalactopyranoside either in drinking water or given intraperitoneally increased luciferase expression in eight of the mice examined, which fell rapidly when withdrawn. Intraperitoneal administration of siRNA targeting lac in combination with Lipofectamine 2000 resulted in increased luciferase expression in the liver while control nontargeting siRNA had no effect. We believe a sensitive positive readout pharmacodynamics reporter model will be of use to the research community in RNAi-based vector development.Molecular Therapy-Nucleic Acids (2013) 2, e133; doi:10.1038/mtna.2013.63; published online 19 November 2013.

6.
Aging Cell ; 12(3): 435-45, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23448364

RESUMO

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.


Assuntos
Senescência Celular/fisiologia , Estresse Oxidativo/fisiologia , Fragmentos de Peptídeos/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Células Cultivadas , Senescência Celular/genética , Hepatócitos/metabolismo , Longevidade , Camundongos , Camundongos Knockout , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Adaptadoras da Sinalização Shc/genética , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Timo/metabolismo , Fatores de Transcrição , Transcrição Gênica , Proteína Supressora de Tumor p53/genética
7.
Endocr Res ; 38(3): 139-150, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23102272

RESUMO

AIM: To test the transactivation domain-mediated control of glucose homeostasis by the tumor suppressor p53. BACKGROUND: The tumor suppressor p53 has a critical role in maintenance of glucose homeostasis. Phosphorylation of Ser18 in the transaction domain of p53 controls the expression of Zpf385a, a zinc finger protein that regulates adipogenesis and adipose function. This results suggest that the transactivation domain of p53 is essential to the control of glucose homeostasis. MATERIALS AND METHODS: Mice with mutations in the p53 transactivation domain were examined for glucose homeostasis as well as various metabolic parameters. Glucose tolerance and insulin tolerance tests were performed on age matched wild type and mutant animals. In addition, mice expressing increased dosage of p53 were also examined. RESULTS: Mice with a mutation in p53Ser18 exhibit reduced Zpf385a expression in adipose tissue, adipose tissue-specific insulin resistance, and glucose intolerance. Mice with relative deficits in the transactivation domain of p53 exhibit similar defects in glucose homeostasis, while "Super p53" mice with an increased dosage of p53 exhibit improved glucose tolerance. CONCLUSION: These data support the role of an ATM-p53 cellular stress axis that helps combat glucose intolerance and insulin resistance and regulates glucose homeostasis.

8.
Sci Transl Med ; 4(124): 124ps6, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22399263

RESUMO

Research reported in this issue of Science Translational Medicine illustrates the benefits of short-term food withdrawal (fasting) in the treatment of cancer. Fasting exploited fundamental differences in the way tumor cells and normal cells respond to stress, simultaneously strengthening normal cell function and weakening tumor cell survival in the presence of toxic doses of chemotherapeutic drugs.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Jejum/fisiologia , Neoplasias/tratamento farmacológico , Animais , Feminino , Humanos
9.
Diabetes ; 60(4): 1210-22, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21357466

RESUMO

OBJECTIVE: Investigating the dynamics of pancreatic ß-cell mass is critical for developing strategies to treat both type 1 and type 2 diabetes. p53, a key regulator of the cell cycle and apoptosis, has mostly been a focus of investigation as a tumor suppressor. Although p53 alternative transcripts can modulate p53 activity, their functions are not fully understood. We hypothesized that ß-cell proliferation and glucose homeostasis were controlled by Δ40p53, a p53 isoform lacking the transactivation domain of the full-length protein that modulates total p53 activity and regulates organ size and life span in mice. RESEARCH DESIGN AND METHODS: We phenotyped metabolic parameters in Δ40p53 transgenic (p44tg) mice and used quantitative RT-PCR, Western blotting, and immunohistochemistry to examine ß-cell proliferation. RESULTS: Transgenic mice with an ectopic p53 gene encoding Δ40p53 developed hypoinsulinemia and glucose intolerance by 3 months of age, which worsened in older mice and led to overt diabetes and premature death from ∼14 months of age. Consistent with a dramatic decrease in ß-cell mass and reduced ß-cell proliferation, lower expression of cyclin D2 and pancreatic duodenal homeobox-1, two key regulators of proliferation, was observed, whereas expression of the cell cycle inhibitor p21, a p53 target gene, was increased. CONCLUSIONS: These data indicate a significant and novel role for Δ40p53 in ß-cell proliferation with implications for the development of age-dependent diabetes.


Assuntos
Glucose/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Isoformas de Proteínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Western Blotting , Proliferação de Células , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Mutantes , Isoformas de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética
10.
Genes Dev ; 24(21): 2408-19, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21041409

RESUMO

Δ40p53 is a transactivation-deficient isoform of the tumor suppressor p53. We discovered that Δ40p53, in addition to being highly expressed in embryonic stem cells (ESCs), is the major p53 isoform during early stages of embryogenesis in the mouse. By altering the dose of Δ40p53 in ESCs, we identified a critical role for this isoform in maintaining the ESC state. Haploinsufficiency for Δ40p53 causes a loss of pluripotency in ESCs and acquisition of a somatic cell cycle, while increased dosage of Δ40p53 prolongs pluripotency and inhibits progression to a more differentiated state. Δ40p53 controls the switch from pluripotent ESCs to differentiated somatic cells by controlling the activity of full-length p53 at critical targets such as Nanog and the IGF-1 receptor (IGF-1R). The IGF axis plays a central role in the switch between pluripotency and differentiation in ESCs-and Δ40p53, by controlling the level of the IGF-1R, acts as a master regulator of this switch. We propose that this is the primary function of Δ40p53 in cells of the early embryo and stem cells, which are the only normal cells in which this isoform is expressed.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Receptor IGF Tipo 1/fisiologia , Transdução de Sinais , Proteína Supressora de Tumor p53/fisiologia , Sequência de Aminoácidos , Animais , Western Blotting , Ciclo Celular , Linhagem Celular , Núcleo Celular/metabolismo , Sobrevivência Celular , Citoplasma/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Proteína Homeobox Nanog , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Receptor IGF Tipo 1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
11.
Aging Cell ; 9(5): 667-84, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20701600

RESUMO

Fat tissue, frequently the largest organ in humans, is at the nexus of mechanisms involved in longevity and age-related metabolic dysfunction. Fat distribution and function change dramatically throughout life. Obesity is associated with accelerated onset of diseases common in old age, while fat ablation and certain mutations affecting fat increase life span. Fat cells turn over throughout the life span. Fat cell progenitors, preadipocytes, are abundant, closely related to macrophages, and dysdifferentiate in old age, switching into a pro-inflammatory, tissue-remodeling, senescent-like state. Other mesenchymal progenitors also can acquire a pro-inflammatory, adipocyte-like phenotype with aging. We propose a hypothetical model in which cellular stress and preadipocyte overutilization with aging induce cellular senescence, leading to impaired adipogenesis, failure to sequester lipotoxic fatty acids, inflammatory cytokine and chemokine generation, and innate and adaptive immune response activation. These pro-inflammatory processes may amplify each other and have systemic consequences. This model is consistent with recent concepts about cellular senescence as a stress-responsive, adaptive phenotype that develops through multiple stages, including major metabolic and secretory readjustments, which can spread from cell to cell and can occur at any point during life. Senescence could be an alternative cell fate that develops in response to injury or metabolic dysfunction and might occur in nondividing as well as dividing cells. Consistent with this, a senescent-like state can develop in preadipocytes and fat cells from young obese individuals. Senescent, pro-inflammatory cells in fat could have profound clinical consequences because of the large size of the fat organ and its central metabolic role.


Assuntos
Tecido Adiposo , Envelhecimento , Senescência Celular , Tecido Adiposo/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Senescência Celular/genética , Humanos
12.
Aging Cell ; 9(2): 174-90, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20409077

RESUMO

The longevity-assurance activity of the tumor suppressor p53 depends on the levels of Delta40p53 (p44), a short and naturally occurring isoform of the p53 gene. As such, increased dosage of p44 in the mouse leads to accelerated aging and short lifespan. Here we show that mice homozygous for a transgene encoding p44 (p44(+/+)) display cognitive decline and synaptic impairment early in life. The synaptic deficits are attributed to hyperactivation of insulin-like growth factor 1 receptor (IGF-1R) signaling and altered metabolism of the microtubule-binding protein tau. In fact, they were rescued by either Igf1r or Mapt haploinsufficiency. When expressing a human or a 'humanized' form of the amyloid precursor protein (APP), p44(+/+) animals developed a selective degeneration of memory-forming and -retrieving areas of the brain, and died prematurely. Mechanistically, the neurodegeneration was caused by both paraptosis- and autophagy-like cell deaths. These results indicate that altered longevity-assurance activity of p53:p44 causes memory loss and neurodegeneration by affecting IGF-1R signaling. Importantly, Igf1r haploinsufficiency was also able to correct the synaptic deficits of APP(695/swe) mice, a model of Alzheimer's disease.


Assuntos
Longevidade , Transtornos da Memória/metabolismo , Doenças Neurodegenerativas/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Autofagia , Transtornos da Memória/genética , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Fragmentos de Peptídeos/genética , Transdução de Sinais , Fatores de Transcrição , Proteína Supressora de Tumor p53/genética , Proteínas tau/deficiência , Proteínas tau/metabolismo
13.
Biochim Biophys Acta ; 1790(12): 1587-91, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19800395

RESUMO

We propose a model in which cell loss in the aging brain is seen as a root cause of behavioral changes that compromise quality of life, including the onset of generalized anxiety disorder, in elderly individuals. According to this model, as stem cells in neurogenic regions of the adult brain lose regenerative capacity, worn-out, dead, or damaged neurons fail to be replaced, leaving gaps in function. As most replacement involves inhibitory interneurons, either directly or indirectly, the net result is the acquisition over time of a hyper-excitable state. The stress axis is subserved by all three neurogenic regions in the adult brain, making it particularly susceptible to these age-dependent changes. We outline a molecular mechanism by which hyper-excitation of the stress axis in turn activates the tumor suppressor p53. This reinforces the loss of stem cell proliferative capacity and interferes with the feedback mechanism by which the glucocorticoid receptor turns off neuroendocrine pathways and resets the axis.


Assuntos
Envelhecimento/fisiologia , Ansiedade/etiologia , Encéfalo/fisiopatologia , Proteína Supressora de Tumor p53/fisiologia , Adulto , Animais , Humanos , Sistema Hipotálamo-Hipofisário/fisiopatologia , Modelos Biológicos , Neurogênese/genética , Neurogênese/fisiologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiologia , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Proteína Supressora de Tumor p53/genética
14.
Exp Gerontol ; 44(1-2): 93-100, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18598747

RESUMO

In higher organisms dependent on the regenerative ability of tissue stem cells to maintain tissue integrity throughout adulthood, the failure of stem cells to replace worn out, dead, or damaged cells is seen as one mechanism that limits life span. In these organisms, tumor suppressors such as p53 are central participants in the control of longevity because they regulate stem cell proliferation. Several recent reports have identified p53 as a longevity gene in organisms such as Caenorhabditis elegans and Drosophila melanogaster, which lack proliferative stem cells in all but the germline and have relatively short life spans. This has forced us to reevaluate the role of p53 in the control of life span. We discuss how p53 might regulate longevity in both long- and short-lived species by controlling the activity of insulin-like molecules that operate in proliferating and non-proliferating compartments of adult somatic tissues. We also discuss the hierarchical structure of life span regulation where loss of p53 has life span extending effects. Finally, we suggest a molecular mechanism by which p53 might facilitate the response to severe nutrient deprivation that allows metabolically active cells to survive periods of starvation. Paradoxically, loss of p53 function in these cells would compromise life span.


Assuntos
Insulina/metabolismo , Transdução de Sinais/fisiologia , Somatomedinas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Caenorhabditis elegans , Proliferação de Células , Sobrevivência Celular , Drosophila melanogaster , Humanos , Expectativa de Vida , Camundongos , Estresse Oxidativo , Inanição/metabolismo , Células-Tronco/fisiologia
15.
Neurobiol Aging ; 30(3): 483-97, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17850928

RESUMO

The question of whether or not stem cell loss drives aging in the brain has not been fully resolved. Here, we used mice over-expressing the short isoform of p53 (DeltaNp53 or p44) as a model of aging to gain insight into the cellular mechanisms underlying age-related functional deficits in the brain. By BrdU labeling, we observed an accelerated decline in the number of subventricular zone proliferating cells with age in p44Tg mice compared to mice with normal p53 expression. A 2-3-fold reduction in the number of slowly dividing stem cells was evident in the subventricular zone of 9-12-month-old p44Tg mice, but not in younger p44Tg mice or in normal mice. Consequently, the supply of new olfactory bulb neurons was also reduced. The number and size of neurospheres generated from subventricular zone cells from p44Tg mice was significantly reduced, and cells derived from these neurospheres had limited self-renewal and amplification capacities. At the cellular level, p44 lengthened the cell cycle and affected cell cycle reentry properties, evident by an increased proportion of cells in G0. At the functional level, p44 expression resulted in impaired olfactory discrimination in 15-16-month-old mice. This phenotype is driven by constitutive activation of p53 and constitutive expression of p21(Cip1/waf1) in neural stem cells. Our results demonstrate that p53 plays a crucial role in the maintenance of the regenerative capacity of the brain by regulating the proliferation of stem and progenitor cells.


Assuntos
Encéfalo/fisiologia , Diferenciação Celular/fisiologia , Neurônios/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Proteína Supressora de Tumor p53/genética , Animais , Encéfalo/citologia , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Regeneração/genética , Células-Tronco/citologia , Proteína Supressora de Tumor p53/fisiologia
16.
Neurobiol Aging ; 30(12): 2010-20, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18378044

RESUMO

The aging program mediated by IGF1-R is responsible for a naturally occurring TrkA to p75(NTR) switch that leads to activation of the second messenger ceramide and increased production of the Alzheimer's disease amyloid beta-peptide. Biochemical and genetic approaches that target IGF1-R signaling, p75(NTR), or ceramide are able to block the above events. Here, we show that the transcription factors Egr-1 and Hipk2 are required elements for the TrkA to p75(NTR) switch downstream of IGF1-R signaling. Specifically, Egr-1 is required for the upregulation of p75(NTR), whereas Hipk2 is required for the downregulation of TrkA. In fact, gene silencing of Egr-1 abolished the ability of IGF1 to upregulate p75(NTR), whereas similar approaches directed against Hipk2 blocked the downregulation of TrkA. In addition, IGF1 treatment favored binding of Egr-1 and Hipk2 to the promoter of p75(NTR) and TrkA, respectively. Finally, the expression levels of both Egr-1 and Hipk2 are upregulated in an age-dependent fashion. Such an event is opposed by caloric restriction, a model of delayed aging, and favored by the p44 transgene in p44(+/+) animals, a model of accelerated aging.


Assuntos
Proteínas de Transporte/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor trkA/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Envelhecimento/metabolismo , Animais , Encéfalo/metabolismo , Restrição Calórica , Linhagem Celular Tumoral , Proteína 1 de Resposta de Crescimento Precoce/genética , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/agonistas , Fator de Transcrição Brn-3A/metabolismo
17.
Mech Ageing Dev ; 130(1-2): 10-7, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18639575

RESUMO

George Williams' antagonistic pleiotropy theory of aging proposes that cellular damage and organismal aging are caused by pleiotrophic genes, or genes with multiple phenotypic effects [Williams, G.C., 1957. Pleiotropy, natural selection, and the evolution of senescence. Evolution 11, 398-411]. According to this theory, genes that exhibit antagonistic pleiotropy increase the odds of successful reproduction early in life, but have deleterious effects later in life. The tumor suppressor p53 confers protection against cancer (and death) by interrupting the abnormal proliferation of cells. When control of proliferation is applied to normal stem cells, however, it can impair tissue homeostasis and accelerate aging. We use data from recently developed models of accelerated aging in mice to determine if the deleterious effects of p53 on aging reflect antagonistic pleiotropy of the p53 gene or are attributable to genes that can modify p53 activity but are evolving independently.


Assuntos
Envelhecimento/fisiologia , Células-Tronco/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Idoso , Animais , Divisão Celular/fisiologia , Senescência Celular/fisiologia , Evolução Molecular , Humanos , Neoplasias/fisiopatologia , Células-Tronco/citologia
18.
PLoS One ; 3(12): e4020, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19107194

RESUMO

BACKGROUND: SIR2 is an NAD(+)-dependent deacetylase [1]-[3] implicated in the regulation of lifespan in species as diverse as yeast [4], worms [5], and flies [6]. We previously reported that the level of SIRT1, the mammalian homologue of SIR2 [7], [8], is coupled to the level of mitotic activity in cells both in vitro and in vivo[9]. Cells from long-lived mice maintained SIRT1 levels of young mice in tissues that undergo continuous cell replacement by proliferating stem cells. Changes in SIRT1 protein level were not associated with changes in mRNA level, suggesting that SIRT1 could be regulated post-transcriptionally. However, other than a recent report on sumoylation [10] and identification of SIRT1 as a nuclear phospho-protein by mass spectrometry [11], post-translational modifications of this important protein have not been reported. METHODOLOGY/PRINCIPAL FINDINGS: We identified 13 residues in SIRT1 that are phosphorylated in vivo using mass spectrometry. Dephosphorylation by phosphatases in vitro resulted in decreased NAD(+)-dependent deacetylase activity. We identified cyclinB/Cdk1 as a cell cycle-dependent kinase that forms a complex with and phosphorylates SIRT1. Mutation of two residues phosphorylated by Cyclin B/Cdk1 (threonine 530 and serine 540) disturbs normal cell cycle progression and fails to rescue proliferation defects in SIRT1-deficient cells [12], [13]. CONCLUSIONS/SIGNIFICANCE: Pharmacological manipulation of SIRT1 activity is currently being tested as a means of extending lifespan in mammals. Treatment of obese mice with resveratrol, a pharmacological activator of SIRT1, modestly but significantly improved longevity and, perhaps more importantly, offered some protection against the development of type 2 diabetes mellitus and metabolic syndrome [14]-[16]. Understanding the endogenous mechanisms that regulate the level and activity of SIRT1, therefore, has obvious relevance to human health and disease. Our results identify phosphorylation by cell cycle dependent kinases as a major mechanism controlling the level and function of this sirtuin and complement recent reports of factors that inhibit [17], [18] and activate [19] SIRT1 by protein-protein interactions.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Sirtuínas/metabolismo , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Proteína Quinase CDC2/metabolismo , Domínio Catalítico , Células Cultivadas , Ciclina B/metabolismo , Ciclinas/metabolismo , Ativação Enzimática , Camundongos , Modelos Moleculares , Fosforilação/fisiologia , Ligação Proteica , Serina/metabolismo , Sirtuína 1 , Sirtuínas/química , Sirtuínas/genética , Treonina/metabolismo
19.
Trends Immunol ; 29(10): 455-63, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18789764

RESUMO

Until recently, dogma dictated that the immune system and the central nervous system (CNS) live mostly separate, parallel lives, and any interactions between the two were assumed to be limited to extreme cases of pathological insult. It was only a decade ago that T cells in the injured brain were shown to play a protective rather than a destructive role. In this article, we explore the role of the immune system in the healthy brain, focusing on the key function that T lymphocytes have in the regulation of cognition. We discuss candidate mechanisms underlying T cell-mediated control of cognitive function in human cognitive diseases associated with immune decline, such as age- and HIV-related dementias, 'chemo-brain' and others.


Assuntos
Complexo AIDS Demência/imunologia , Sistema Nervoso Central/imunologia , Transtornos Cognitivos/imunologia , Demência/imunologia , Complexo AIDS Demência/patologia , Envelhecimento/imunologia , Animais , Sistema Nervoso Central/patologia , Transtornos Cognitivos/patologia , Citocinas/imunologia , Citocinas/metabolismo , Demência/patologia , Dopamina/imunologia , Dopamina/metabolismo , Humanos , Imunidade , Camundongos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Serotonina/imunologia , Serotonina/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
20.
Mech Ageing Dev ; 129(4): 238-41, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18242663

RESUMO

Loss of function mutations in the p53 tumor suppressor gene predispose mice and humans to cancer, resulting in abbreviated life spans. A dominant mutation in the murine HD gene, similar to mutations that cause Huntington's disease in humans, reverses some of the effects of p53 mutations on longevity. We attribute this to the enhanced apoptotic effect of the expanded polyglutamine region in the HD protein on proliferating cells lacking p53.


Assuntos
Envelhecimento/genética , Alelos , Longevidade/genética , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína Supressora de Tumor p53/deficiência , Animais , Apoptose , Genótipo , Proteína Huntingtina , Camundongos , Camundongos Knockout , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...