Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(34): e2302738120, 2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-37579159

RESUMO

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is characterized by various disabling symptoms including exercise intolerance and is diagnosed in the absence of a specific cause, making its clinical management challenging. A better understanding of the molecular mechanism underlying this apparent bioenergetic deficiency state may reveal insights for developing targeted treatment strategies. We report that overexpression of Wiskott-Aldrich Syndrome Protein Family Member 3 (WASF3), here identified in a 38-y-old woman suffering from long-standing fatigue and exercise intolerance, can disrupt mitochondrial respiratory supercomplex formation and is associated with endoplasmic reticulum (ER) stress. Increased expression of WASF3 in transgenic mice markedly decreased their treadmill running capacity with concomitantly impaired respiratory supercomplex assembly and reduced complex IV levels in skeletal muscle mitochondria. WASF3 induction by ER stress using endotoxin, well known to be associated with fatigue in humans, also decreased skeletal muscle complex IV levels in mice, while decreasing WASF3 levels by pharmacologic inhibition of ER stress improved mitochondrial function in the cells of the patient with chronic fatigue. Expanding on our findings, skeletal muscle biopsy samples obtained from a cohort of patients with ME/CFS showed increased WASF3 protein levels and aberrant ER stress activation. In addition to revealing a potential mechanism for the bioenergetic deficiency in ME/CFS, our study may also provide insights into other disorders associated with fatigue such as rheumatic diseases and long COVID.


Assuntos
COVID-19 , Síndrome de Fadiga Crônica , Animais , Feminino , Humanos , Camundongos , COVID-19/metabolismo , Síndrome de Fadiga Crônica/diagnóstico , Mitocôndrias/metabolismo , Síndrome de COVID-19 Pós-Aguda , Respiração , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Camundongos Transgênicos
2.
FEBS J ; 289(22): 6959-6968, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34235856

RESUMO

Molecular oxygen possesses a dual nature due to its highly reactive free radical property: it is capable of oxidizing metabolic substrates to generate cellular energy, but can also serve as a substrate for genotoxic reactive oxygen species generation. As a labile substance upon which aerobic life depends, the mechanisms for handling cellular oxygen have been fine-tuned and orchestrated in evolution. Protection from atmospheric oxygen toxicity as originally posited by the Endosymbiotic Theory of the Mitochondrion is likely to be one basic principle underlying oxygen homeostasis. We briefly review the literature on oxygen homeostasis both in vitro and in vivo with a focus on the role of the mitochondrion where the majority of cellular oxygen is consumed. The insights gleaned from these basic mechanisms are likely to be important for understanding disease pathogenesis and developing strategies for maintaining health.


Assuntos
Mitocôndrias , Oxigênio , Mitocôndrias/metabolismo , Radicais Livres/metabolismo , Oxigênio/metabolismo , Homeostase , Espécies Reativas de Oxigênio/metabolismo , Estresse Oxidativo
3.
Arterioscler Thromb Vasc Biol ; 41(11): 2648-2660, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34587760

RESUMO

Significant progress has been made in developing new treatments and refining the use of preexisting ones against cancer. Their successful use and the longer survival of cancer patients have been associated with reports of new cardiotoxicities and the better characterization of the previously known cardiac complications. Immunotherapies with monoclonal antibodies against specific cancer-promoting genes, chimeric antigen receptor T cells, and immune checkpoint inhibitors have been developed to fight cancer cells, but they can also show off-target effects on the heart. Some of these cardiotoxicities are thought to be due to nonspecific immune activation and inflammatory damage. Unlike immunotherapy-associated cardiotoxicities which are relatively new entities, there is extensive literature on anthracycline-induced cardiomyopathy. Here, we provide a brief overview of the cardiotoxicities of immunotherapies for the purpose of distinguishing them from anthracycline cardiomyopathy. This is especially relevant as the expansion of oncological treatments presents greater diagnostic challenges in determining the cause of cardiac dysfunction in cancer survivors with a history of multiple cancer treatments including anthracyclines and immunotherapies administered concurrently or serially over time. We then provide a focused review of the mechanisms proposed to underlie the development of anthracycline cardiomyopathy based on experimental data mostly in mouse models. Insights into its pathogenesis may stimulate the development of new strategies to identify patients who are susceptible to anthracycline cardiomyopathy while permitting low cardiac risk patients to receive optimal treatment for their cancer.


Assuntos
Antraciclinas/efeitos adversos , Antibióticos Antineoplásicos/efeitos adversos , Cardiopatias/induzido quimicamente , Miócitos Cardíacos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Cardiotoxicidade , Dano ao DNA , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Cardiopatias/prevenção & controle , Humanos , Terapia de Alvo Molecular/efeitos adversos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Medição de Risco , Fatores de Risco , Transdução de Sinais
4.
Cancer Prev Res (Phila) ; 14(1): 31-40, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32958587

RESUMO

Germline mutations of TP53, which cause the cancer predisposition disorder Li-Fraumeni syndrome (LFS), can increase mitochondrial activity as well as fatty acid ß-oxidation (FAO) in mice. Increased fatty acid metabolism can promote cancer malignancy, but its specific contribution to tumorigenesis in LFS remains unclear. To investigate this, we crossed LFS mice carrying the p53 R172H knock-in mutation (p53172H/H , homolog of the human TP53 R175H LFS mutation) with myoglobin-knockout (MB-/- ) mice known to have decreased FAO. MB-/- p53172H/H double-mutant mice also showed mildly reduced FAO in thymus, a common site of T lymphoma development in LFS mice, in association with an approximately 40% improvement in cancer-free survival time. RNA sequencing profiling revealed that the p53 R172H mutation promotes mitochondrial metabolism and ribosome biogenesis, both of which are suppressed by the disruption of MB. The activation of ribosomal protein S6, involved in protein translation and implicated in cancer promotion, was also inhibited in the absence of MB. To further confirm the role of FAO in lymphomagenesis, mitochondrial FAO enzyme, carnitine palmitoyltransferase 2 (CPT2), was specifically disrupted in T cells of p53172H/H mice using a Cre-loxP-mediated strategy. The heterozygous knockout of CPT2 resulted in thymus FAO haploinsufficiency and an approximately 30% improvement in survival time, paralleling the antiproliferative signaling observed with MB disruption. Thus, this study demonstrates that moderating FAO in LFS can suppress tumorigenesis and improve cancer-free survival with potential implications for cancer prevention. PREVENTION RELEVANCE: Mildly inhibiting the increased fatty acid oxidation observed in a mouse model of Li-Fraumeni syndrome, a cancer predisposition disorder caused by inherited mutations of TP53, dampens aberrant pro-tumorigenic cell signaling and improves the survival time of these mice, thereby revealing a potential strategy for cancer prevention in patients.


Assuntos
Carcinogênese/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Ácidos Graxos/metabolismo , Síndrome de Li-Fraumeni/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Carcinogênese/genética , Carnitina O-Palmitoiltransferase/genética , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Intervalo Livre de Doença , Metabolismo Energético , Feminino , Técnicas de Introdução de Genes , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Heterozigoto , Humanos , Síndrome de Li-Fraumeni/complicações , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/mortalidade , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Mioblastos , Mioglobina/genética , Oxirredução , Cultura Primária de Células , Estudos Prospectivos , Proteína Supressora de Tumor p53/genética , Adulto Jovem
5.
Mol Cell Oncol ; 7(3): 1724598, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32391420

RESUMO

Doxorubicin is widely used against cancer but carries the risk of a progressive cardiomyopathy associated with mitochondrial loss. Using genetic models, our recent study demonstrates that mitochondrial genomic DNA regulation by tumor protein p53 (TP53, best known as p53) prevents the cardiotoxicity of low dose doxorubicin which does not activate the p53-dependent cell death pathway.

6.
Cell Rep ; 30(3): 783-792.e5, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31968253

RESUMO

The physiological effects of the many germline mutations of TP53, encoding the tumor suppressor protein p53, are poorly understood. Here we report generating a p53 R178C knockin mouse modeling the human TP53 R181C mutation, which is notable for its prevalence and prior molecular characterization. Consistent with its weak cancer penetrance in humans, homozygous p53178C/C mice show a modest increase in tumorigenesis but, surprisingly, are lean with decreased body fat content. They display evidence of increased lipolysis and upregulation of fatty acid metabolism in their inguinal white adipose tissue (iWAT). Gene expression and chromatin immunoprecipitation sequencing (ChIP-seq) analyses show that the mutant p53 bound and transactivated Beta-3-Adrenergic Receptor (ADRB3), a gene that is known to promote lipolysis and is associated with obesity. This study reveals that a germline mutation of p53 can affect fat metabolism, which has been implicated in cancer development.


Assuntos
Mutação em Linhagem Germinativa/genética , Lipólise/genética , Homologia de Sequência de Aminoácidos , Proteína Supressora de Tumor p53/genética , Células 3T3-L1 , Adipócitos/metabolismo , Tecido Adiposo Branco/patologia , Animais , Sequência de Bases , Ácidos Graxos/sangue , Regulação da Expressão Gênica , Homozigoto , Humanos , Síndrome de Li-Fraumeni/genética , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Análise de Componente Principal , Receptores Adrenérgicos beta 3/genética , Transdução de Sinais
7.
JNCI Cancer Spectr ; 4(6): pkaa063, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33490865

RESUMO

BACKGROUND: Li-Fraumeni syndrome (LFS) is a highly penetrant autosomal dominant cancer predisposition disorder caused by germline TP53 pathogenic variants. Patients with LFS have increased oxidative phosphorylation capacity in skeletal muscle and oxidative stress in blood. Metformin inhibits oxidative phosphorylation, reducing available energy for cancer cell proliferation and decreasing production of reactive oxygen species that cause DNA damage. Thus, metformin may provide pharmacologic risk reduction for cancer in patients with LFS, but its safety in nondiabetic patients with germline TP53 pathogenic variants has not been documented. METHODS: This study assessed safety and tolerability of metformin in nondiabetic LFS patients and measured changes in metabolic profiles. Adult patients with LFS and germline TP53 variant received 14 weeks of metformin. Blood samples were obtained for measurement of serum insulin-like growth factor-1, insulin, and insulin-like growth factor binding protein 3. Hepatic mitochondrial function was assessed with fasting exhaled CO2 after ingestion of 13C-labeled methionine. Changes in serum metabolome were measured. All statistical tests were 2-sided. RESULTS: We enrolled 26 participants: 20 females and 6 males. The most common adverse events were diarrhea (50.0%) and nausea (46.2%). Lactic acidosis did not occur, and there were no changes in fasting glucose. Cumulative mean 13C exhalation was statistically significantly suppressed by metformin (P = .001). Mean levels of insulin-like growth factor binding protein 3 and insulin-like growth factor-1 were statistically significantly lowered (P = .02). Lipid metabolites and branched-chain amino acids accumulated. CONCLUSIONS: Metformin was safe and tolerable in patients with LFS. It suppressed hepatic mitochondrial function as expected in these individuals. This study adds to the rationale for development of a pharmacologic risk-reduction clinical trial of metformin in LFS.

8.
Proc Natl Acad Sci U S A ; 116(39): 19626-19634, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31488712

RESUMO

Doxorubicin is a widely used chemotherapeutic agent that causes dose-dependent cardiotoxicity in a subset of treated patients, but the genetic determinants of this susceptibility are poorly understood. Here, we report that a noncanonical tumor suppressor activity of p53 prevents cardiac dysfunction in a mouse model induced by doxorubicin administered in divided low doses as in the clinics. While relatively preserved in wild-type (p53+/+ ) state, mice deficient in p53 (p53-/- ) developed left ventricular (LV) systolic dysfunction after doxorubicin treatment. This functional decline in p53-/- mice was associated with decreases in cardiac oxidative metabolism, mitochondrial mass, and mitochondrial genomic DNA (mtDNA) homeostasis. Notably, mice with homozygous knockin of the p53 R172H (p53172H/H ) mutation, which like p53-/- state lacks the prototypical tumor suppressor activities of p53 such as apoptosis but retains its mitochondrial biogenesis capacity, showed preservation of LV function and mitochondria after doxorubicin treatment. In contrast to p53-null state, wild-type and mutant p53 displayed distinct mechanisms of transactivating mitochondrial transcription factor A (TFAM) and p53-inducible ribonucleotide reductase 2 (p53R2), which are involved in mtDNA transcription and maintenance. Importantly, supplementing mice with a precursor of NAD+ prevented the mtDNA depletion and cardiac dysfunction. These findings suggest that loss of mtDNA contributes to cardiomyopathy pathogenesis induced by doxorubicin administered on a schedule simulating that in the clinics. Given a similar mtDNA protection role of p53 in doxorubicin-treated human induced pluripotent stem cell (iPSC)-derived cardiomyocytes, the mitochondrial markers associated with cardiomyopathy development observed in blood and skeletal muscle cells may have prognostic utility.


Assuntos
Cardiotoxicidade/metabolismo , Cardiotoxicidade/prevenção & controle , Doxorrubicina/toxicidade , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/fisiologia , Cardiomiopatias/metabolismo , DNA Mitocondrial/genética , Proteínas de Ligação a DNA , Cardiopatias/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais , Mutação , Miócitos Cardíacos/metabolismo , Biogênese de Organelas , Cultura Primária de Células , Fatores de Transcrição , Proteína Supressora de Tumor p53/genética
9.
Oncotarget ; 10(6): 631-632, 2019 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-30774760
10.
Cancer Res ; 78(18): 5375-5383, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30042151

RESUMO

Inheritance of germline mutations in the tumor suppressor gene TP53 causes Li-Fraumeni syndrome (LFS), a cancer predisposition disorder. The arginine to histidine substitution at amino acid position 337 of p53 (R337H) is a founder mutation highly prevalent in southern and southeastern Brazil and is considered an LFS mutation. Although this mutation is of significant clinical interest, its role in tumorigenesis using animal models has not been described. Here, we generate a knockin mouse model containing the homologous R337H mutation (mouse R334H). De novo tumorigenesis was not significantly increased in either heterozygous (p53334R/H ) or homozygous (p53334H/H ) p53 R334H knockin mice compared with wild-type mice. However, susceptibility to diethylnitrosamine (DEN)-induced liver carcinogenesis was increased in a mutant allele dose-dependent manner. In parallel, p53334H/H mice exposed to DEN exhibited increased DNA damage but decreased cell-cycle regulation in the liver. Oligomerization of p53, which is required for transactivation of target genes, was reduced in R334H liver, consistent with its decreased nuclear activity compared with wild-type. By modeling a TP53 mutation in mice that has relatively weak cancer penetrance, this study provides in vivo evidence that the human R337H mutation can compromise p53 activity and promote tumorigenesis.Significance: A germline mutation in the oligomerization domain of p53 decreases its transactivation potential and renders mice susceptible to carcinogen-induced liver tumorigenesis. Cancer Res; 78(18); 5375-83. ©2018 AACR.


Assuntos
Carcinogênese/genética , Neoplasias Hepáticas/patologia , Proteína Supressora de Tumor p53/genética , Alelos , Animais , Brasil , Transformação Celular Neoplásica/genética , Dano ao DNA , Fibroblastos/metabolismo , Técnicas de Introdução de Genes , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Homozigoto , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Ativação Transcricional
11.
J Clin Invest ; 127(1): 132-136, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-27869650

RESUMO

Li-Fraumeni syndrome (LFS) is a cancer predisposition disorder caused by germline mutations in TP53 that can lead to increased mitochondrial metabolism in patients. However, the implications of altered mitochondrial function for tumorigenesis in LFS are unclear. Here, we have reported that genetic or pharmacologic disruption of mitochondrial respiration improves cancer-free survival in a mouse model of LFS that expresses mutant p53. Mechanistically, inhibition of mitochondrial function increased autophagy and decreased the aberrant proliferation signaling caused by mutant p53. In a pilot study, LFS patients treated with metformin exhibited decreases in mitochondrial activity concomitant with activation of antiproliferation signaling, thus reproducing the effects of disrupting mitochondrial function observed in LFS mice. These observations indicate that a commonly prescribed diabetic medicine can restrain mitochondrial metabolism and tumorigenesis in an LFS model, supporting its further consideration for cancer prevention in LFS patients.


Assuntos
Síndrome de Li-Fraumeni/prevenção & controle , Metformina/farmacologia , Mitocôndrias/metabolismo , Neoplasias Experimentais/prevenção & controle , Consumo de Oxigênio/efeitos dos fármacos , Adulto , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Feminino , Humanos , Células Jurkat , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Pessoa de Meia-Idade , Mitocôndrias/genética , Mitocôndrias/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Consumo de Oxigênio/genética , Projetos Piloto , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
J Biol Chem ; 291(48): 24819-24827, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27687729

RESUMO

Although exercise is linked with improved health, the specific molecular mechanisms underlying its various benefits require further clarification. Here we report that exercise increases the nuclear localization and activity of p53 by acutely down-regulating coiled-coil-helix-coiled-coil-helix domain 4 (CHCHD4), a carrier protein that mediates p53 import into the mitochondria. This response to exercise is lost in transgenic mice with constitutive expression of CHCHD4. Mechanistically, exercise-induced nuclear transcription factor FOXO3 binds to the CHCHD4 promoter and represses its expression, preventing the translocation of p53 to the mitochondria and thereby increasing p53 nuclear localization. The synergistic increase in nuclear p53 and FOXO3 by exercise can facilitate their known interaction in transactivating Sirtuin 1 (SIRT1), a NAD+-dependent histone deacetylase that mediates adaptation to various stresses. Thus, our results reveal one mechanism by which exercise could be involved in preventing cancer and potentially other diseases associated with aging.


Assuntos
Núcleo Celular/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Condicionamento Físico Animal , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/genética , Proteína Forkhead Box O3/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas de Transporte da Membrana Mitocondrial/genética , Elementos de Resposta , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/genética
13.
J Med Chem ; 59(21): 9575-9598, 2016 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-27416328

RESUMO

Poly(ADP-ribose)polymerase-1 (PARP-1) is a critical DNA repair enzyme in the base excision repair pathway. Inhibitors of this enzyme comprise a new type of anticancer drug that selectively kills cancer cells by targeting homologous recombination repair defects. Since 2010, important advances have been achieved in PARP-1 inhibitors. Specifically, the approval of olaparib in 2014 for the treatment of ovarian cancer with BRCA mutations validated PARP-1 as an anticancer target and established its clinical importance in cancer therapy. Here, we provide an update on PARP-1 inhibitors, focusing on breakthroughs in their clinical applications and investigations into relevant mechanisms of action, biomarkers, and drug resistance. We also provide an update on the design strategies and the structural types of PARP-1 inhibitors. Opportunities and challenges in PARP-1 inhibitors for cancer therapy will be discussed based on the above advances.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Modelos Moleculares , Estrutura Molecular , Neoplasias/patologia , Inibidores de Poli(ADP-Ribose) Polimerases/síntese química , Inibidores de Poli(ADP-Ribose) Polimerases/química , Relação Estrutura-Atividade
14.
Curr Opin Genet Dev ; 38: 16-22, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27003724

RESUMO

Under normal conditions, basal levels of wild-type p53 promote mitochondrial function through multiple mechanisms. Remarkably, some missense mutations of p53, in contrast to the null state, can result in the retention of its metabolic activities. These effects are particularly prominent in the mitochondria and demonstrate a functional role for mutant p53 in cancer metabolism. This review summarizes accumulating data on the mechanisms by which p53 missense mutations can regulate mitochondrial metabolism and promote the viability and survival of both normal and cancer cells, thus acting as a double edged sword for the host. Greater understanding of these mechanisms may provide insights for developing new treatment or preventive strategies against cancer.


Assuntos
Mitocôndrias/genética , Neoplasias/genética , Proteína Supressora de Tumor p53/genética , Humanos , Mitocôndrias/patologia , Mutação de Sentido Incorreto , Neoplasias/patologia
15.
Prog Cardiovasc Dis ; 57(4): 337-46, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25559061

RESUMO

Regular exercise and a physically active lifestyle have favorable effects on health. Several issues related to this theme are addressed in this report. A comment on the requirements of personalized exercise medicine and in-depth biological profiling along with the opportunities that they offer is presented. This is followed by a brief overview of the evidence for the contributions of genetic differences to the ability to benefit from regular exercise. Subsequently, studies showing that mutations in TP53 influence exercise capacity in mice and humans are succinctly described. The evidence for effects of exercise on endothelial function in health and disease also is covered. Finally, changes in cardiac and skeletal muscle in response to exercise and their implications for patients with cardiac disease are summarized. Innovative research strategies are needed to define the molecular mechanisms involved in adaptation to exercise and to translate them into useful clinical and public health applications.


Assuntos
Doenças Cardiovasculares , Terapia por Exercício/métodos , Tolerância ao Exercício/genética , Músculo Esquelético/fisiologia , Medicina de Precisão/métodos , Proteína Supressora de Tumor p53/genética , Adaptação Fisiológica/genética , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Doenças Cardiovasculares/prevenção & controle , Exercício Físico , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Atividade Motora/fisiologia , Mutação
16.
Free Radic Biol Med ; 73: 270-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24887096

RESUMO

We previously reported that Polo-like kinase 2 (PLK2) is highly expressed in cells with defective mitochondrial respiration and is essential for their survival. Although PLK2 has been widely studied as a cell cycle regulator, we have uncovered an antioxidant function for this kinase that activates the GSK3-NRF2 signaling pathway. Here, we report that the expression of PLK2 is responsive to oxidative stress and that PLK2 mediates antioxidant signaling by phosphorylating GSK3, thereby promoting the nuclear translocation of NRF2. We further show that the antioxidant activity of PLK2 is essential for preventing p53-dependent necrotic cell death. Thus, the regulation of redox homeostasis by PLK2 promotes the survival of cells with dysfunctional mitochondria, which may have therapeutic implications for cancer and neurodegenerative diseases.


Assuntos
Antioxidantes/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Mitocôndrias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Células HCT116 , Humanos , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno , Transplante Heterólogo
17.
Methods Enzymol ; 542: 209-21, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24862268

RESUMO

There is growing evidence in the basic science field that aberrant metabolism plays an important role in tumorigenesis. Therefore, it is imperative to perform investigations in human subjects to determine (1) whether the metabolic observations made in model systems are applicable to humans; and (2) if indeed applicable, whether the metabolic alterations are clinically significant for cancer development. As an initial step, here we describe methods for measuring the mitochondrial metabolism of blood lymphocytes and skeletal muscle myoblasts that can be obtained from human subjects.


Assuntos
Bioquímica/métodos , Linfócitos/metabolismo , Mitocôndrias/metabolismo , Mioblastos Esqueléticos/metabolismo , Células Cultivadas , Humanos , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(43): 17356-61, 2013 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-24101517

RESUMO

p53, a critical tumor suppressor, regulates mitochondrial respiration, but how a nuclear protein can orchestrate the function of an organelle encoded by two separate genomes, both of which require p53 for their integrity, remains unclear. Here we report that the mammalian homolog of the yeast mitochondrial disulfide relay protein Mia40 (CHCHD4) is necessary for the respiratory-dependent translocation of p53 into the mitochondria. In the setting of oxidative stress, increased CHCHD4 expression partitions p53 into the mitochondria and protects its genomic integrity while decreasing p53 nuclear localization and transcriptional activity. Conversely, decreased CHCHD4 expression prevents the mitochondrial translocation of p53 while augmenting its nuclear localization and activity. Thus, the mitochondrial disulfide relay system allows p53 to regulate two spatially segregated genomes depending on oxidative metabolic activity.


Assuntos
Dissulfetos/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Western Blotting , Núcleo Celular/metabolismo , Reparo do DNA , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Mutação , Ligação Proteica , Transporte Proteico , Interferência de RNA , Proteína Supressora de Tumor p53/genética
19.
N Engl J Med ; 368(11): 1027-32, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23484829

RESUMO

There is growing evidence that alterations in metabolism may contribute to tumorigenesis. Here, we report on members of families with the Li-Fraumeni syndrome who carry germline mutations in TP53, the gene encoding the tumor-suppressor protein p53. As compared with family members who are not carriers and with healthy volunteers, family members with these mutations have increased oxidative phosphorylation of skeletal muscle. Basic experimental studies of tissue samples from patients with the Li-Fraumeni syndrome and a mouse model of the syndrome support this in vivo finding of increased mitochondrial function. These results suggest that p53 regulates bioenergetic homeostasis in humans. (Funded by the National Heart, Lung, and Blood Institute and the National Institutes of Health; ClinicalTrials.gov number, NCT00406445.).


Assuntos
Metabolismo Energético/genética , Exercício Físico/fisiologia , Genes p53 , Síndrome de Li-Fraumeni/metabolismo , Mitocôndrias Musculares/metabolismo , Fosfocreatina/metabolismo , Animais , Estudos de Casos e Controles , Modelos Animais de Doenças , Feminino , Mutação em Linhagem Germinativa , Heterozigoto , Humanos , Síndrome de Li-Fraumeni/genética , Masculino , Camundongos , Músculo Esquelético/metabolismo , Consumo de Oxigênio/genética , Consumo de Oxigênio/fisiologia , Projetos Piloto , Levantamento de Peso/fisiologia
20.
Curr Opin Oncol ; 24(1): 76-82, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22123233

RESUMO

PURPOSE OF REVIEW: There is an inverse relationship between cancer incidence and cardiorespiratory fitness in large population studies. Mechanistic insights into these observations may strengthen the rationale for encouraging exercise fitness in the clinics for cancer prevention and may promote the development of new preventive strategies. RECENT FINDINGS: Studying the multifaceted activities of p53, a critical tumor suppressor gene, has revealed various cellular pathways necessary for adapting to environmental stresses. Genetic connections are being made between p53 and an increasing number of metabolic activities such as oxidative phosphorylation, glycolysis and fatty acid oxidation. In-vivo mouse models show that p53 plays an important role in determining both basal aerobic exercise capacity and its improvement by training. SUMMARY: The genetic pathways by which p53 regulates metabolism and exercise may help explain significant epidemiologic observations connecting cardiorespiratory fitness and cancer. Further understanding of these molecular pathways through human translational studies may promote the development of new cancer preventive strategies.


Assuntos
Exercício Físico/fisiologia , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Aerobiose , Anaerobiose , Animais , Respiração Celular/fisiologia , Humanos , Camundongos , Mitocôndrias/fisiologia , Neoplasias/genética , Neoplasias/prevenção & controle , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA