Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Cell Biol ; 223(8)2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38865089

RESUMO

During aging and in some contexts, like embryonic development, wound healing, and diseases such as cancer, senescent cells accumulate and play a key role in different pathophysiological functions. A long-held belief was that cellular senescence decreased normal cell functions, given the loss of proliferation of senescent cells. This view radically changed following the discovery of the senescence-associated secretory phenotype (SASP), factors released by senescent cells into their microenvironment. There is now accumulating evidence that cellular senescence also promotes gain-of-function effects by establishing, reinforcing, or changing cell identity, which can have a beneficial or deleterious impact on pathophysiology. These effects may involve both proliferation arrest and autocrine SASP production, although they largely remain to be defined. Here, we provide a historical overview of the first studies on senescence and an insight into emerging trends regarding the effects of senescence on cell identity.


Assuntos
Senescência Celular , Fenótipo Secretor Associado à Senescência , Humanos , Fenótipo Secretor Associado à Senescência/genética , Animais , Proliferação de Células
2.
Commun Biol ; 7(1): 691, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839869

RESUMO

Cellular senescence is a stress-induced, permanent cell cycle arrest involved in tumor suppression and aging. Senescent cells secrete bioactive molecules such as pro-inflammatory cytokines and chemokines. This senescence-associated secretory phenotype (SASP) has been implicated in immune-mediated elimination of senescent cells and age-associated chronic inflammation. However, the mechanisms regulating the SASP are incompletely understood. Here, we show that the stress-responsive kinase apoptosis signal-regulating kinase 1 (ASK1) promotes inflammation in senescence and aging. ASK1 is activated during senescence and increases the expression of pro-inflammatory cytokines and chemokines by activating p38, a kinase critical for the SASP. ASK1-deficient mice show impaired elimination of oncogene-induced senescent cells and an increased rate of tumorigenesis. Furthermore, ASK1 deficiency prevents age-associated p38 activation and inflammation and attenuates glomerulosclerosis. Our results suggest that ASK1 is a driver of the SASP and age-associated chronic inflammation and represents a potential therapeutic target for age-related diseases.


Assuntos
Envelhecimento , Senescência Celular , Inflamação , MAP Quinase Quinase Quinase 5 , MAP Quinase Quinase Quinase 5/metabolismo , MAP Quinase Quinase Quinase 5/genética , Animais , Inflamação/metabolismo , Camundongos , Humanos , Camundongos Knockout , Camundongos Endogâmicos C57BL , Fenótipo Secretor Associado à Senescência/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Citocinas/metabolismo , Citocinas/genética
3.
Cancer Lett ; 593: 216930, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38705566

RESUMO

Radiotherapy (RT) in non-small cell lung cancer (NSCLC) triggers cellular senescence, complicating tumor microenvironments and affecting treatment outcomes. This study examines the role of lymphocyte immunoglobulin-like receptor B2 (LILRB2) in modulating RT-induced senescence and radiosensitivity in NSCLC. Through methodologies including irradiation, lentivirus transfection, and various molecular assays, we assessed LILRB2's expression and its impact on cellular senescence levels and tumor cell behaviors. Our findings reveal that RT upregulates LILRB2, facilitating senescence and a senescence-associated secretory phenotype (SASP), which in turn enhances tumor proliferation and resistance to radiation. Importantly, LILRB2 silencing attenuates these effects by inhibiting the JAK2/STAT3 pathway, significantly increasing radiosensitivity in NSCLC models. Clinical data correlate high LILRB2 expression with reduced RT response and poorer prognosis, suggesting LILRB2's pivotal role in RT-induced senescence and its potential as a therapeutic target to improve NSCLC radiosensitivity.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Senescência Celular , Neoplasias Pulmonares , Tolerância a Radiação , Receptores Imunológicos , Humanos , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Senescência Celular/efeitos da radiação , Tolerância a Radiação/genética , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Animais , Janus Quinase 2/metabolismo , Janus Quinase 2/genética , Camundongos , Transdução de Sinais , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Fenótipo Secretor Associado à Senescência/genética , Células A549 , Feminino
4.
Nucleic Acids Res ; 52(11): 6298-6316, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38682582

RESUMO

Senescent cells can influence the function of tissues in which they reside, and their propensity for disease. A portion of adult human pancreatic beta cells express the senescence marker p16, yet it is unclear whether they are in a senescent state, and how this affects insulin secretion. We analyzed single-cell transcriptome datasets of adult human beta cells, and found that p16-positive cells express senescence gene signatures, as well as elevated levels of beta-cell maturation genes, consistent with enhanced functionality. Senescent human beta-like cells in culture undergo chromatin reorganization that leads to activation of enhancers regulating functional maturation genes and acquisition of glucose-stimulated insulin secretion capacity. Strikingly, Interferon-stimulated genes are elevated in senescent human beta cells, but genes encoding senescence-associated secretory phenotype (SASP) cytokines are not. Senescent beta cells in culture and in human tissue show elevated levels of cytoplasmic DNA, contributing to their increased interferon responsiveness. Human beta-cell senescence thus involves chromatin-driven upregulation of a functional-maturation program, and increased responsiveness of interferon-stimulated genes, changes that could increase both insulin secretion and immune reactivity.


Assuntos
Senescência Celular , Montagem e Desmontagem da Cromatina , Células Secretoras de Insulina , Interferons , Humanos , Células Secretoras de Insulina/metabolismo , Senescência Celular/genética , Interferons/metabolismo , Interferons/genética , Secreção de Insulina , Insulina/metabolismo , Cromatina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Células Cultivadas , Fenótipo Secretor Associado à Senescência/genética , Transcriptoma , Análise de Célula Única
5.
Ageing Res Rev ; 97: 102287, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38570142

RESUMO

The components that comprise the senescence-associated secretory phenotype (SASP) include growth factors, proteases, chemokines, cytokines, and bioactive lipids. It drives secondary aging and disrupts tissue homeostasis, ultimately leading to tissue repair and regeneration loss. It has a two-way regulatory effect on tumor cells, resisting cancer occurrence and promoting its progression. A category of single-stranded circular non-coding RNA molecules known as circular RNAs (circRNAs) carries out a series of cellular activities, including sequestering miRNAs and modulating gene editing and expression. Research has demonstrated that a large number of circRNAs exhibit aberrant expression in pathological settings, and play a part in the onset and progress of cancer via modulating SASP factors. However, the research related to SASP and circRNAs in tumors is still in its infancy at this stage. This review centers on the bidirectional modulation of SASP and the role of circRNAs in regulating SASP factors across different types of tumors. The aim is to present novel perspectives for the diagnosis and therapeutic management of malignancies.


Assuntos
Neoplasias , RNA Circular , Fenótipo Secretor Associado à Senescência , Humanos , RNA Circular/genética , RNA Circular/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fenótipo Secretor Associado à Senescência/genética , Animais
6.
Cell Rep ; 38(2): 110211, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021076

RESUMO

Oncogene-induced senescence (OIS) is a form of stable cell-cycle arrest arising in response to oncogenic stimulation. OIS must be bypassed for transformation, but the mechanisms of OIS establishment and bypass remain poorly understood, especially at the post-transcriptional level. Here, we show that the RNA-binding protein UNR/CSDE1 enables OIS in primary mouse keratinocytes. Depletion of CSDE1 leads to senescence bypass, cell immortalization, and tumor formation, indicating that CSDE1 behaves as a tumor suppressor. Unbiased high-throughput analyses uncovered that CSDE1 promotes OIS by two independent molecular mechanisms: enhancement of the stability of senescence-associated secretory phenotype (SASP) factor mRNAs and repression of Ybx1 mRNA translation. Importantly, depletion of YBX1 from immortal keratinocytes rescues senescence and uncouples proliferation arrest from the SASP, revealing multilayered mechanisms exerted by CSDE1 to coordinate senescence. Our data highlight the relevance of post-transcriptional control in the regulation of senescence.


Assuntos
Senescência Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular , Proliferação de Células/fisiologia , Senescência Celular/genética , Proteínas de Ligação a DNA/fisiologia , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Oncogenes/genética , Cultura Primária de Células , Processamento Pós-Transcricional do RNA/fisiologia , Proteínas de Ligação a RNA/fisiologia , Fenótipo Secretor Associado à Senescência/genética , Fenótipo Secretor Associado à Senescência/fisiologia , Transdução de Sinais/fisiologia , Proteína 1 de Ligação a Y-Box/metabolismo
7.
Hepatology ; 75(3): 584-599, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34687050

RESUMO

BACKGROUND AND AIMS: The mechanisms involved in liver regeneration after partial hepatectomy (pHx) are complicated. Cellular senescence, once linked to aging, plays a pivotal role in wound repair. However, the regulatory effects of cellular senescence on liver regeneration have not been fully elucidated. APPROACH AND RESULTS: Mice subjected to pHx were analyzed 14 days after surgery. The incomplete remodeling of liver sinusoids affected shear stress-induced endothelial nitric oxide synthase (eNOS) signaling on day 14, resulting in the accumulation of senescent LSECs. Removing macrophages to augment LSEC senescence led to a malfunction of the regenerating liver. A dynamic fluctuation in Notch activity accompanied senescent LSEC accumulation during liver regeneration. Endothelial Notch activation by using Cdh5-CreERT NICeCA mice triggered LSEC senescence and senescence-associated secretory phenotype, which disrupted liver regeneration. Blocking the Notch by γ-secretase inhibitor (GSI) diminished senescence and promoted LSEC expansion. Mechanically, Notch-hairy and enhancer of split 1 signaling inhibited sirtuin 1 (Sirt1) transcription by binding to its promoter region. Activation of Sirt1 by SRT1720 neutralized the up-regulation of P53, P21, and P16 caused by Notch activation and eliminated Notch-driven LSEC senescence. Finally, Sirt1 activator promoted liver regeneration by abrogating LSEC senescence and improving sinusoid remodeling. CONCLUSIONS: Shear stress-induced LSEC senescence driven by Notch interferes with liver regeneration after pHx. Sirt1 inhibition accelerates liver regeneration by abrogating Notch-driven senescence, providing a potential opportunity to target senescent cells and facilitate liver repair after injury.


Assuntos
Senescência Celular , Regeneração Hepática , Receptores Notch , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/metabolismo , Animais , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Inibidores e Moduladores de Secretases gama/farmacologia , Hepatectomia/métodos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Regeneração Hepática/efeitos dos fármacos , Regeneração Hepática/fisiologia , Camundongos , Óxido Nítrico Sintase Tipo III/metabolismo , Receptores Notch/antagonistas & inibidores , Receptores Notch/metabolismo , Fenótipo Secretor Associado à Senescência/genética
8.
Exp Mol Med ; 53(12): 1834-1841, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34916607

RESUMO

Senescent cells in cancer tissue, including senescent fibroblasts and macrophages, have been reported to increase the malignant potency of cancer cells by secreting senescence-associated secretory phenotype (SASP). Otherwise, Senescence of tumor cells has been believed to inhibit tumor growth by halting the massive proliferation and increasing the chances of immune clearance. In particular, senescent tumor cells (STCs) have been thought that they rarely exist in carcinomas because oncogene-induced senescence needs to be overcome for protumorigenic cells to become malignant. However, recent studies have revealed that a considerable number of STCs are present in cancer tissue, even in metastatic sites. In fact, STCs are widely involved in cancer progression by leading to collective invasion and building a cytokine barrier to protect nonsenescent tumor cells from immune attack. Furthermore, therapy-induced STCs can induce tumor progression and recurrence by increasing stemness. However, obscure causative factors and their heterogeneity in various cancers make it difficult to establish the physiological role of STCs. Here, we summarize and review the current knowledge of the pathophysiology and role of STCs. We also outline the current status of therapeutic strategies for directly removing STCs or modulating the SASPs to maximize the positive functions of STCs while suppressing the negative functions.


Assuntos
Senescência Celular , Neoplasias/metabolismo , Senescência Celular/genética , Gerenciamento Clínico , Suscetibilidade a Doenças , Desenvolvimento de Medicamentos , Regulação da Expressão Gênica , Humanos , Neoplasias/etiologia , Neoplasias/patologia , Neoplasias/terapia , Especificidade de Órgãos/genética , Fenótipo Secretor Associado à Senescência/genética , Microambiente Tumoral
9.
Nat Metab ; 3(12): 1706-1726, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34873338

RESUMO

Ageing-associated functional decline of organs and increased risk for age-related chronic pathologies is driven in part by the accumulation of senescent cells, which develop the senescence-associated secretory phenotype (SASP). Here we show that procyanidin C1 (PCC1), a polyphenolic component of grape seed extract (GSE), increases the healthspan and lifespan of mice through its action on senescent cells. By screening a library of natural products, we find that GSE, and PCC1 as one of its active components, have specific effects on senescent cells. At low concentrations, PCC1 appears to inhibit SASP formation, whereas it selectively kills senescent cells at higher concentrations, possibly by promoting production of reactive oxygen species and mitochondrial dysfunction. In rodent models, PCC1 depletes senescent cells in a treatment-damaged tumour microenvironment and enhances therapeutic efficacy when co-administered with chemotherapy. Intermittent administration of PCC1 to either irradiated, senescent cell-implanted or naturally aged old mice alleviates physical dysfunction and prolongs survival. We identify PCC1 as a natural senotherapeutic agent with in vivo activity and high potential for further development as a clinical intervention to delay, alleviate or prevent age-related pathologies.


Assuntos
Flavonoides/farmacologia , Longevidade/efeitos dos fármacos , Senoterapia/farmacologia , Animais , Apoptose , Linhagem Celular , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Biologia Computacional/métodos , Desenvolvimento de Medicamentos , Metabolismo Energético/efeitos dos fármacos , Flavonoides/química , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Estresse Oxidativo , Fenótipo Secretor Associado à Senescência/genética , Senoterapia/química
10.
Signal Transduct Target Ther ; 6(1): 354, 2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34675187

RESUMO

Senescent endothelial cells (ECs) could impair the integrity of the blood vessel endothelium, leading to vascular aging and a series of diseases, such as atherosclerosis, diabetes. Preventing or mitigating EC senescence might serve as a promising therapeutic paradigm for these diseases. Recent studies showed that small extracellular vesicles (sEV) have the potential to transfer bioactive molecules into recipient cells and induce phenotypic changes. Since mesenchymal stem cells (MSCs) have long been postulated as an important source cell in regenerative medicine, herein we investigated the role and mechanism of MSC-derived sEV (MSC-sEV) on EC senescence. In vitro results showed that MSC-sEV reduced senescent biomarkers, decreased senescence-associated secretory phenotype (SASP), rescued angiogenesis, migration and other dysfunctions in senescent EC induced by oxidative stress. In the In vivo natural aging and type-2 diabetes mouse wound-healing models (both of which have senescent ECs), MSC-sEV promoted wound closure and new blood vessel formation. Mechanically, miRNA microarray showed that miR-146a was highly expressed in MSC-sEV and also upregulated in EC after MSC-sEV treatment. miR-146a inhibitors abolished the stimulatory effects of MSC-sEV on senescence. Moreover, we found miR-146a could suppress Src phosphorylation and downstream targets VE-cadherin and Caveolin-1. Collectively, our data indicate that MSC-sEV mitigated endothelial cell senescence and stimulate angiogenesis through miR-146a/Src.


Assuntos
Envelhecimento/genética , Senescência Celular/genética , Vesículas Extracelulares/genética , MicroRNAs/genética , Quinases da Família src/genética , Envelhecimento/patologia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Regulação da Expressão Gênica/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Estresse Oxidativo/genética , Fenótipo Secretor Associado à Senescência/genética , Cicatrização/genética
11.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34426493

RESUMO

Cellular senescence causes a dramatic alteration of chromatin organization and changes the gene expression profile of proinflammatory factors, thereby contributing to various age-related pathologies through the senescence-associated secretory phenotype (SASP). Chromatin organization and global gene expression are maintained by the CCCTC-binding factor (CTCF); however, the molecular mechanism underlying CTCF regulation and its association with SASP gene expression remains unclear. We discovered that noncoding RNA (ncRNA) derived from normally silenced pericentromeric repetitive sequences directly impairs the DNA binding of CTCF. This CTCF disturbance increases the accessibility of chromatin and activates the transcription of SASP-like inflammatory genes, promoting malignant transformation. Notably, pericentromeric ncRNA was transferred into surrounding cells via small extracellular vesicles acting as a tumorigenic SASP factor. Because CTCF blocks the expression of pericentromeric ncRNA in young cells, the down-regulation of CTCF during cellular senescence triggers the up-regulation of this ncRNA and SASP-related inflammatory gene expression. In this study, we show that pericentromeric ncRNA provokes chromosomal alteration by inhibiting CTCF, leading to a SASP-like inflammatory response in a cell-autonomous and non-cell-autonomous manner and thus may contribute to the risk of tumorigenesis during aging.


Assuntos
Envelhecimento/genética , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Inflamação/genética , RNA não Traduzido/fisiologia , Fenótipo Secretor Associado à Senescência/genética , Animais , Senescência Celular/genética , Centrômero , DNA de Neoplasias/metabolismo , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias , Ligação Proteica/genética
12.
Cancer Lett ; 520: 68-79, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34237406

RESUMO

Cellular senescence is a stress response that imposes a growth arrest on cancer and nonmalignant cells during cancer therapy. By secreting a plethora of proinflammatory factors collectively termed the senescence-associated secretory phenotype (SASP), therapy-induced senescent cells can promote tumorigenesis. Moreover, the SASP from senescent cells is also able to drive therapy resistance and mediate many adverse effects of cancer therapy. Because senescent cell production often occurs during cancer therapy, it is important to carefully consider these potential detrimental effects. Senotherapy, which refers to selective removal of senescent cells, has been proposed as a promising adjuvant approach to eliminate the adverse effects of senescent cells. Thus, in this review we summarize in detail the mechanisms by which senescent cells contribute to tumorigenesis and therapeutic resistance. Also, we thoroughly discuss the potential strategies regarding how to effectively circumvent the undesirable effects of therapy-induced senescent cells.


Assuntos
Carcinogênese/genética , Senescência Celular/genética , Neoplasias/genética , Humanos , Neoplasias/patologia , Fenótipo Secretor Associado à Senescência/genética
13.
Cell Rep ; 36(4): 109441, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320349

RESUMO

Cellular senescence is characterized as a stable proliferation arrest that can be triggered by multiple stresses. Most knowledge about senescent cells is obtained from studies in primary cells. However, senescence features may be different in cancer cells, since the pathways that are involved in senescence induction are often deregulated in cancer. We report here a comprehensive analysis of the transcriptome and senolytic responses in a panel of 13 cancer cell lines rendered senescent by two distinct compounds. We show that in cancer cells, the response to senolytic agents and the composition of the senescence-associated secretory phenotype are more influenced by the cell of origin than by the senescence trigger. Using machine learning, we establish the SENCAN gene expression classifier for the detection of senescence in cancer cell samples. The expression profiles and senescence classifier are available as an interactive online Cancer SENESCopedia.


Assuntos
Senescência Celular , Neoplasias/patologia , Compostos de Anilina/farmacologia , Azepinas/farmacologia , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Etoposídeo/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias/genética , Pirimidinas/farmacologia , Reprodutibilidade dos Testes , Fenótipo Secretor Associado à Senescência/efeitos dos fármacos , Fenótipo Secretor Associado à Senescência/genética , Senoterapia/farmacologia , Sulfonamidas/farmacologia
14.
Aging Cell ; 20(4): e13329, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33734555

RESUMO

Senescence phenotypes and mitochondrial dysfunction are implicated in aging and in premature aging diseases, including ataxia telangiectasia (A-T). Loss of mitochondrial function can drive age-related decline in the brain, but little is known about whether improving mitochondrial homeostasis alleviates senescence phenotypes. We demonstrate here that mitochondrial dysfunction and cellular senescence with a senescence-associated secretory phenotype (SASP) occur in A-T patient fibroblasts, and in ATM-deficient cells and mice. Senescence is mediated by stimulator of interferon genes (STING) and involves ectopic cytoplasmic DNA. We further show that boosting intracellular NAD+ levels with nicotinamide riboside (NR) prevents senescence and SASP by promoting mitophagy in a PINK1-dependent manner. NR treatment also prevents neurodegeneration, suppresses senescence and neuroinflammation, and improves motor function in Atm-/- mice. Our findings suggest a central role for mitochondrial dysfunction-induced senescence in A-T pathogenesis, and that enhancing mitophagy as a potential therapeutic intervention.


Assuntos
Ataxia Telangiectasia/dietoterapia , Ataxia Telangiectasia/metabolismo , Suplementos Nutricionais , Proteínas de Membrana/metabolismo , Mitofagia/efeitos dos fármacos , NAD/metabolismo , Niacinamida/análogos & derivados , Compostos de Piridínio/administração & dosagem , Fenótipo Secretor Associado à Senescência/genética , Transdução de Sinais/efeitos dos fármacos , Animais , Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Estudos de Casos e Controles , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Mitofagia/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Niacinamida/administração & dosagem , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Transfecção , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA