Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell ; 187(16): 4150-4175, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39121846

RESUMO

Cellular senescence is a cell fate triggered in response to stress and is characterized by stable cell-cycle arrest and a hypersecretory state. It has diverse biological roles, ranging from tissue repair to chronic disease. The development of new tools to study senescence in vivo has paved the way for uncovering its physiological and pathological roles and testing senescent cells as a therapeutic target. However, the lack of specific and broadly applicable markers makes it difficult to identify and characterize senescent cells in tissues and living organisms. To address this, we provide practical guidelines called "minimum information for cellular senescence experimentation in vivo" (MICSE). It presents an overview of senescence markers in rodent tissues, transgenic models, non-mammalian systems, human tissues, and tumors and their use in the identification and specification of senescent cells. These guidelines provide a uniform, state-of-the-art, and accessible toolset to improve our understanding of cellular senescence in vivo.


Assuntos
Senescência Celular , Humanos , Animais , Biomarcadores/metabolismo , Guias como Assunto , Neoplasias/patologia
2.
Nat Cell Biol ; 26(8): 1336-1345, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39103548

RESUMO

The accumulation of senescent cells promotes ageing and age-related diseases, but molecular mechanisms that senescent cells use to evade immune clearance and accumulate in tissues remain to be elucidated. Here we report that p16-positive senescent cells upregulate the immune checkpoint protein programmed death-ligand 1 (PD-L1) to accumulate in ageing and chronic inflammation. We show that p16-mediated inhibition of cell cycle kinases CDK4/6 induces PD-L1 stability in senescent cells via downregulation of its ubiquitin-dependent degradation. p16-expressing senescent alveolar macrophages elevate PD-L1 to promote an immunosuppressive environment that can contribute to an increased burden of senescent cells. Treatment with activating anti-PD-L1 antibodies engaging Fcγ receptors on effector cells leads to the elimination of PD-L1 and p16-positive cells. Our study uncovers a molecular mechanism of p16-dependent regulation of PD-L1 protein stability in senescent cells and reveals the potential of targeting PD-L1 to improve immunosurveillance of senescent cells and ameliorate senescence-associated inflammation.


Assuntos
Antígeno B7-H1 , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina , Estabilidade Proteica , Senescência Celular/imunologia , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Animais , Humanos , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/genética , Vigilância Imunológica , Camundongos Endogâmicos C57BL , Quinase 6 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/genética , Camundongos , Proteólise , Receptores de IgG/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/genética
3.
Nat Cardiovasc Res ; 3(8): 915-932, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39196027

RESUMO

Senescence plays a key role in various physiological and pathological processes. We reported that injury-induced transient senescence correlates with heart regeneration, yet the multi-omics profile and molecular underpinnings of regenerative senescence remain obscure. Using proteomics and single-cell RNA sequencing, here we report the regenerative senescence multi-omic signature in the adult mouse heart and establish its role in neonatal heart regeneration and agrin-mediated cardiac repair in adult mice. We identified early growth response protein 1 (Egr1) as a regulator of regenerative senescence in both models. In the neonatal heart, Egr1 facilitates angiogenesis and cardiomyocyte proliferation. In adult hearts, agrin-induced senescence and repair require Egr1, activated by the integrin-FAK-ERK-Akt1 axis in cardiac fibroblasts. We also identified cathepsins as injury-induced senescence-associated secretory phenotype components that promote extracellular matrix degradation and potentially assist in reducing fibrosis. Altogether, we uncovered the molecular signature and functional benefits of regenerative senescence during heart regeneration, with Egr1 orchestrating the process.


Assuntos
Proliferação de Células , Senescência Celular , Proteína 1 de Resposta de Crescimento Precoce , Miócitos Cardíacos , Regeneração , Animais , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Regeneração/fisiologia , Senescência Celular/fisiologia , Miócitos Cardíacos/metabolismo , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/fisiologia , Transdução de Sinais , Fibroblastos/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Cultivadas , Animais Recém-Nascidos , Modelos Animais de Doenças , Fenótipo Secretor Associado à Senescência , Proteômica , Análise de Célula Única , Masculino , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos Knockout , Quinase 1 de Adesão Focal
4.
Nat Neurosci ; 27(6): 1116-1124, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38637622

RESUMO

Alzheimer's disease (AD) and dementia in general are age-related diseases with multiple contributing factors, including brain inflammation. Microglia, and specifically those expressing the AD risk gene TREM2, are considered important players in AD, but their exact contribution to pathology remains unclear. In this study, using high-throughput mass cytometry in the 5×FAD mouse model of amyloidosis, we identified senescent microglia that express high levels of TREM2 but also exhibit a distinct signature from TREM2-dependent disease-associated microglia (DAM). This senescent microglial protein signature was found in various mouse models that show cognitive decline, including aging, amyloidosis and tauopathy. TREM2-null mice had fewer microglia with a senescent signature. Treating 5×FAD mice with the senolytic BCL2 family inhibitor ABT-737 reduced senescent microglia, but not the DAM population, and this was accompanied by improved cognition and reduced brain inflammation. Our results suggest a dual and opposite involvement of TREM2 in microglial states, which must be considered when contemplating TREM2 as a therapeutic target in AD.


Assuntos
Envelhecimento , Doença de Alzheimer , Encéfalo , Modelos Animais de Doenças , Glicoproteínas de Membrana , Microglia , Receptores Imunológicos , Animais , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Microglia/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Camundongos , Envelhecimento/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Camundongos Transgênicos , Senescência Celular/fisiologia , Senescência Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Aging (Albany NY) ; 15(7): 2395-2417, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36996500

RESUMO

Cellular senescence is a stable state of cell cycle arrest that regulates tissue integrity and protects the organism from tumorigenesis. However, the accumulation of senescent cells during aging contributes to age-related pathologies. One such pathology is chronic lung inflammation. p21 (CDKN1A) regulates cellular senescence via inhibition of cyclin-dependent kinases (CDKs). However, its role in chronic lung inflammation and functional impact on chronic lung disease, where senescent cells accumulate, is less understood. To elucidate the role of p21 in chronic lung inflammation, we subjected p21 knockout (p21-/-) mice to repetitive inhalations of lipopolysaccharide (LPS), an exposure that leads to chronic bronchitis and accumulation of senescent cells. p21 knockout led to a reduced presence of senescent cells, alleviated the pathological manifestations of chronic lung inflammation, and improved the fitness of the mice. The expression profiling of the lung cells revealed that resident epithelial and endothelial cells, but not immune cells, play a significant role in mediating the p21-dependent inflammatory response following chronic LPS exposure. Our results implicate p21 as a critical regulator of chronic bronchitis and a driver of chronic airway inflammation and lung destruction.


Assuntos
Bronquite Crônica , Pneumonia , Camundongos , Animais , Células Endoteliais/metabolismo , Bronquite Crônica/genética , Lipopolissacarídeos/toxicidade , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Pneumonia/metabolismo , Ciclo Celular , Senescência Celular/fisiologia , Inflamação
6.
Gut ; 71(2): 345-355, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33649045

RESUMO

OBJECTIVE: Cellular senescence limits tumourigenesis by blocking the proliferation of premalignant cells. Additionally, however, senescent cells can exert paracrine effects influencing tumour growth. Senescent cells are present in premalignant pancreatic intraepithelial neoplasia (PanIN) lesions, yet their effects on the disease are poorly characterised. It is currently unknown whether senolytic drugs, aimed at eliminating senescent cells from lesions, could be beneficial in blocking tumour development. DESIGN: To uncover the functions of senescent cells and their potential contribution to early pancreatic tumourigenesis, we isolated and characterised senescent cells from PanINs formed in a Kras-driven mouse model, and tested the consequences of their targeted elimination through senolytic treatment. RESULTS: We found that senescent PanIN cells exert a tumour-promoting effect through expression of a proinflammatory signature that includes high Cox2 levels. Senolytic treatment with the Bcl2-family inhibitor ABT-737 eliminated Cox2-expressing senescent cells, and an intermittent short-duration treatment course dramatically reduced PanIN development and progression to pancreatic ductal adenocarcinoma. CONCLUSIONS: These findings reveal that senescent PanIN cells support tumour growth and progression, and provide a first indication that elimination of senescent cells may be effective as preventive therapy for the progression of precancerous lesions.


Assuntos
Adenocarcinoma/patologia , Senescência Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/patologia , Senoterapia/uso terapêutico , Adenocarcinoma/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Neoplasias Pancreáticas/metabolismo , Lesões Pré-Cancerosas/metabolismo
7.
Semin Cancer Biol ; 87: 214-219, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-33486077

RESUMO

Cellular senescence, a stable form of cell cycle arrest, accompanied by pronounced secretory activity, has functional roles in both physiological and pathological conditions. Although senescence has been linked for a long time with cancer and ageing, recent studies have revealed a functional role of senescence in development, regeneration and reprogramming. Notably, the transient presence of senescent cells may be beneficial, in contrast to the potential deleterious effects of persistent senescence in aged or chronically damaged tissues. We will discuss how senescence contributes to embryonic development, cell plasticity and tissue regeneration, as a highly coordinated and programmed cellular state.


Assuntos
Plasticidade Celular , Neoplasias , Humanos , Idoso , Senescência Celular/genética , Envelhecimento/genética , Pontos de Checagem do Ciclo Celular , Neoplasias/genética , Neoplasias/metabolismo
8.
Nat Rev Mol Cell Biol ; 22(2): 75-95, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33328614

RESUMO

Cellular senescence, first described in vitro in 1961, has become a focus for biotech companies that target it to ameliorate a variety of human conditions. Eminently characterized by a permanent proliferation arrest, cellular senescence occurs in response to endogenous and exogenous stresses, including telomere dysfunction, oncogene activation and persistent DNA damage. Cellular senescence can also be a controlled programme occurring in diverse biological processes, including embryonic development. Senescent cell extrinsic activities, broadly related to the activation of a senescence-associated secretory phenotype, amplify the impact of cell-intrinsic proliferative arrest and contribute to impaired tissue regeneration, chronic age-associated diseases and organismal ageing. This Review discusses the mechanisms and modulators of cellular senescence establishment and induction of a senescence-associated secretory phenotype, and provides an overview of cellular senescence as an emerging opportunity to intervene through senolytic and senomorphic therapies in ageing and ageing-associated diseases.


Assuntos
Envelhecimento , Senescência Celular , Telômero , Pesquisa Translacional Biomédica , Animais , Proliferação de Células , Dano ao DNA , Humanos , Fenótipo
9.
Antioxid Redox Signal ; 34(4): 324-334, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32212823

RESUMO

Significance: Senescence is an essential biological process that blocks tumorigenesis, limits tissue damage, and aids embryonic development. However, once senescent cells accumulate in tissues during aging, they promote the development of age-related diseases and limit health span. Thus, it is essential to expand the boundaries of our knowledge about the mechanisms responsible for controlling cellular senescence. Recent Advances: Cellular metabolism plays a significant role in the regulation of various signaling processes involved in cell senescence. In the past decade, our knowledge about the interplay between cell signaling, cell metabolism, and cellular senescence has significantly expanded. Critical Issues: In this study, we review metabolic pathways in senescent cells and the impact of these pathways on the response to DNA damage and the senescence-associated secretory phenotype. Future Directions: Future research should elucidate metabolic mechanisms that promote specific alterations in senescent cell phenotype, with a final goal of developing a new therapeutic strategy. Antioxid. Redox Signal. 34, 324-334.


Assuntos
Senescência Celular/fisiologia , Dano ao DNA , Metabolismo Energético , Envelhecimento/fisiologia , Animais , Biomarcadores , Humanos , Fenótipo , Transdução de Sinais
10.
Nat Genet ; 52(11): 1208-1218, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33128048

RESUMO

Cultured cell lines are the workhorse of cancer research, but the extent to which they recapitulate the heterogeneity observed among malignant cells in tumors is unclear. Here we used multiplexed single-cell RNA-seq to profile 198 cancer cell lines from 22 cancer types. We identified 12 expression programs that are recurrently heterogeneous within multiple cancer cell lines. These programs are associated with diverse biological processes, including cell cycle, senescence, stress and interferon responses, epithelial-mesenchymal transition and protein metabolism. Most of these programs recapitulate those recently identified as heterogeneous within human tumors. We prioritized specific cell lines as models of cellular heterogeneity and used them to study subpopulations of senescence-related cells, demonstrating their dynamics, regulation and unique drug sensitivities, which were predictive of clinical response. Our work describes the landscape of heterogeneity within diverse cancer cell lines and identifies recurrent patterns of heterogeneity that are shared between tumors and specific cell lines.


Assuntos
Linhagem Celular Tumoral , Heterogeneidade Genética , Neoplasias/genética , Lesões Pré-Cancerosas/genética , Linhagem Celular Tumoral/efeitos dos fármacos , Senescência Celular/genética , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , RNA-Seq , Estresse Fisiológico/genética , Microambiente Tumoral
11.
FEBS J ; 287(13): 2636-2646, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32145148

RESUMO

The extracellular matrix (ECM) is a key noncellular component in all organs and tissues. It is composed of a large number of proteins including collagens, glycoproteins (GP), and ECM-associated proteins, which show diversity of biochemical and biophysical functions. The ECM is dynamic both in normal physiology of tissues and under pathological conditions. One cellular phenomenon associated with changes in both ECM components expression and in ECM remodeling enzymes secretion is cellular senescence. It represents a stable state form of cell cycle arrest induced in proliferating cells by various forms of stress. Short-term induction of senescence is essential for tumor suppression and tissue repair. However, long-term presence of senescent cells in tissues may have a detrimental role in promoting tissue damage and aging. Up to date, there is insufficient knowledge about the interplay between the ECM and senescence cells. Since changes in the ECM occur in many physiological and pathological conditions in which senescent cells are present, a better understanding of ECM-senescence interactions is necessary. Here, we will review the functions of the different ECM components and will discuss the current knowledge about their regulation in senescent cells and their influence on the senescence state.


Assuntos
Envelhecimento/patologia , Transformação Celular Neoplásica/patologia , Senescência Celular , Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Neoplasias/patologia , Envelhecimento/metabolismo , Animais , Transformação Celular Neoplásica/metabolismo , Humanos , Neoplasias/metabolismo , Via Secretória , Cicatrização
12.
EMBO J ; 38(18): e100849, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31424120

RESUMO

The placenta is an autonomous organ that maintains fetal growth and development. Its multinucleated syncytiotrophoblast layer, providing fetal nourishment during gestation, exhibits characteristics of cellular senescence. We show that in human placentas from pregnancies with intrauterine growth restriction, these characteristics are decreased. To elucidate the functions of pathways regulating senescence in syncytiotrophoblast, we used dynamic contrast-enhanced MRI in mice with attenuated senescence programs. This approach revealed an altered dynamics in placentas of p53-/- , Cdkn2a-/- , and Cdkn2a-/- ;p53-/- mice, accompanied by histopathological changes in placental labyrinths. Human primary syncytiotrophoblast upregulated senescence markers and molecular pathways associated with cell-cycle inhibition and senescence-associated secretory phenotype. The pathways and components of the secretory phenotype were compromised in mouse placentas with attenuated senescence and in human placentas from pregnancies with intrauterine growth restriction. We propose that molecular mediators of senescence regulate placental structure and function, through both cell-autonomous and non-autonomous mechanisms.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Retardo do Crescimento Fetal/genética , Redes Reguladoras de Genes , Placenta/diagnóstico por imagem , Proteína Supressora de Tumor p53/genética , Animais , Senescência Celular , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Imageamento por Ressonância Magnética , Camundongos , Fenótipo , Placenta/metabolismo , Gravidez , Transdução de Sinais , Trofoblastos/metabolismo
13.
Methods Mol Biol ; 1884: 259-267, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30465209

RESUMO

Cellular senescence, a state of permanent growth arrest, is an important mechanism preventing the propagation of damaged cells. It suppresses cancer development in premalignant lesions in response to activated oncogenes and in tumors following therapy. The presence of senescent cells in premalignant lesions and tumors is controlled by the immune system. The ability to identify and quantify senescent cells more efficiently in vivo is necessary in order to evaluate the effect of these cells on tumorigenesis and cancer therapy. Through combining senescent-associated beta-galactosidase staining with ImageStream X analysis, we have developed an effective method to identify and quantify senescent cancer cells in vivo.


Assuntos
Senescência Celular/imunologia , Citometria de Fluxo/métodos , Neoplasias/patologia , Coloração e Rotulagem/métodos , Animais , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos , Citometria de Fluxo/instrumentação , Galactose/metabolismo , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Nus , Neoplasias/imunologia , Coloração e Rotulagem/instrumentação , Transfecção/instrumentação , Transfecção/métodos , beta-Galactosidase/metabolismo
14.
Nat Commun ; 9(1): 5435, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575733

RESUMO

Cellular senescence is a stress response that imposes stable cell-cycle arrest in damaged cells, preventing their propagation in tissues. However, senescent cells accumulate in tissues in advanced age, where they might promote tissue degeneration and malignant transformation. The extent of immune-system involvement in regulating age-related accumulation of senescent cells, and its consequences, are unknown. Here we show that Prf1-/- mice with impaired cell cytotoxicity exhibit both higher senescent-cell tissue burden and chronic inflammation. They suffer from multiple age-related disorders and lower survival. Strikingly, pharmacological elimination of senescent-cells by ABT-737 partially alleviates accelerated aging phenotype in these mice. In LMNA+/G609G progeroid mice, impaired cell cytotoxicity further promotes senescent-cell accumulation and shortens lifespan. ABT-737 administration during the second half of life of these progeroid mice abrogates senescence signature and increases median survival. Our findings shed new light on mechanisms governing senescent-cell presence in aging, and could motivate new strategies for regenerative medicine.


Assuntos
Senescência Celular , Imunossenescência , Perforina/fisiologia , Animais , Compostos de Bifenilo/farmacologia , Compostos de Bifenilo/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Feminino , Inflamação/etiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitrofenóis/farmacologia , Nitrofenóis/uso terapêutico , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Progéria/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico
15.
J Clin Invest ; 128(4): 1247-1254, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29608140

RESUMO

Cellular senescence is a physiological phenomenon that has both beneficial and detrimental consequences. Senescence limits tumorigenesis and tissue damage throughout the lifetime. However, at the late stages of life, senescent cells increasingly accumulate in tissues and might also contribute to the development of various age-related pathologies. Recent studies have revealed the molecular pathways that preserve the viability of senescent cells and the ones regulating their immune surveillance. These studies provide essential initial insights for the development of novel therapeutic strategies for targeting senescent cells. At the same time they stress the need to understand the limitations of the existing strategies, their efficacy and safety, and the possible deleterious consequences of senescent cell elimination. Here we discuss the existing strategies for targeting senescent cells and upcoming challenges in translating these strategies into safe and efficient therapies. Successful translation of these strategies could have implications for treating a variety of diseases at old age and could potentially reshape our view of health management during aging.


Assuntos
Envelhecimento , Transformação Celular Neoplásica , Senescência Celular/imunologia , Vigilância Imunológica , Neoplasias , Envelhecimento/imunologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia
16.
Cell Rep ; 22(13): 3468-3479, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590616

RESUMO

The tumor suppressor p53 limits tumorigenesis by inducing apoptosis, cell cycle arrest, and senescence. Although p53 is known to limit inflammation during tumor development, its role in regulating chronic lung inflammation is less well understood. To elucidate the function of airway epithelial p53 in such inflammation, we subjected genetically modified mice, whose bronchial epithelial club cells lack p53, to repetitive inhalations of lipopolysaccharide (LPS), an exposure that leads to severe chronic bronchitis and airway senescence in wild-type mice. Surprisingly, the club cell p53 knockout mice exhibited reduced airway senescence and bronchitis in response to chronic LPS exposure and were significantly protected from global lung destruction. Furthermore, pharmacological elimination of senescent cells also protected wild-type mice from chronic LPS-induced bronchitis. Our results implicate p53 in induction of club-cell senescence and correlate epithelial cell senescence of chronic airway inflammation and lung destruction.


Assuntos
Brônquios/metabolismo , Pneumonia/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Brônquios/patologia , Senescência Celular/fisiologia , Doença Crônica , Progressão da Doença , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/patologia
17.
EMBO Mol Med ; 10(2): 294-308, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29212784

RESUMO

Epidermal growth factor receptor (EGFR) mutations identify patients with lung cancer who derive benefit from kinase inhibitors. However, most patients eventually develop resistance, primarily due to the T790M second-site mutation. Irreversible inhibitors (e.g., osimertinib/AZD9291) inhibit T790M-EGFR, but several mechanisms, including a third-site mutation, C797S, confer renewed resistance. We previously reported that a triple mixture of monoclonal antibodies, 3×mAbs, simultaneously targeting EGFR, HER2, and HER3, inhibits T790M-expressing tumors. We now report that 3×mAbs, including a triplet containing cetuximab and trastuzumab, inhibits C797S-expressing tumors. Unlike osimertinib, which induces apoptosis, 3×mAbs promotes degradation of the three receptors and induces cellular senescence. Consistent with distinct mechanisms, treatments combining 3×mAbs plus sub-inhibitory doses of osimertinib synergistically and persistently eliminated tumors. Thus, oligoclonal antibodies, either alone or in combination with kinase inhibitors, might preempt repeated cycles of treatment and rapid emergence of resistance.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Cetuximab/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/terapia , Piperazinas/farmacologia , Trastuzumab/farmacologia , Acrilamidas , Compostos de Anilina , Apoptose , Carcinoma Pulmonar de Células não Pequenas/genética , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/genética , Humanos , Imunoterapia , Neoplasias Pulmonares/genética , Mutação , Piperazinas/administração & dosagem , Inibidores de Proteínas Quinases
18.
Reprod Sci ; 25(8): 1254-1260, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29108468

RESUMO

OBJECTIVE: Placenta percreta (PP) is an abnormal condition of trophoblast maturation and terminal differentiation through the uterine wall. We opted to study telomere homeostasis and senescence expression in trophoblasts from PP, the most severe subgroup of placenta accreta. STUDY DESIGN: Paraffin-embedded placental biopsies from pregnancies with percreta and normal placentation, matched by gestational age at delivery, were assessed for telomere length, aggregates, and senescence-associated heterochromatin foci using quantitative fluorescence in situ hybridization. Cyclin-dependent kinase inhibitors p21, p15, p16, and the tumor suppressor protein p53, known senescence-related markers, were assessed using immunohistochemical staining. RESULTS: Short telomeres were found more often in trophoblasts from the samples of PP (n = 9) compared to controls (n = 8; 54% ± 20% vs 2.3% ± 1.16%, respectively; P < .05). More cells with telomere aggregates (18.3% ± 6.9%) were observed in the PP than in the control group (4.8% ± 5.4%; P = .0005). The percentage of nucleic senescence-associated heterochromatin foci in the PP and control samples was similar (10.9% ± 10.4% vs 10.7% ± 15%, respectively; P = .97). Immunohistochemistry of senescence markers was expressed differently in PP compared to the controls: higher p15 expression (46.42% ± 15.2% vs 36.63% ± 12.2%, P = .004), higher p21 expression (59.8% ± 22.1% vs 47.5% ± 21.9%, P = .011), lower p16 expression (54.8% ± 26.3% vs 73.4% ± 18.9%, P = .000), and lower p53 expression (24.4% ± 33.8% vs 34% ± 14.4%, P = .000). CONCLUSION: Placenta percreta exhibits telomere alterations and changes in expression of several senescence markers. These might be related to altered trophoblast invasion maturation and placental detachment postpartum.


Assuntos
Senescência Celular , Placenta Acreta/fisiopatologia , Homeostase do Telômero , Adulto , Feminino , Humanos , Placenta Acreta/metabolismo , Gravidez , Trofoblastos/metabolismo , Trofoblastos/fisiologia
19.
EMBO J ; 36(15): 2280-2295, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28607003

RESUMO

Cellular senescence is a permanent state of cell cycle arrest that protects the organism from tumorigenesis and regulates tissue integrity upon damage and during tissue remodeling. However, accumulation of senescent cells in tissues during aging contributes to age-related pathologies. A deeper understanding of the mechanisms regulating the viability of senescent cells is therefore required. Here, we show that the CDK inhibitor p21 (CDKN1A) maintains the viability of DNA damage-induced senescent cells. Upon p21 knockdown, senescent cells acquired multiple DNA lesions that activated ataxia telangiectasia mutated (ATM) and nuclear factor (NF)-κB kinase, leading to decreased cell survival. NF-κB activation induced TNF-α secretion and JNK activation to mediate death of senescent cells in a caspase- and JNK-dependent manner. Notably, p21 knockout in mice eliminated liver senescent stellate cells and alleviated liver fibrosis and collagen production. These findings define a novel pathway that regulates senescent cell viability and fibrosis.


Assuntos
Caspases/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Regulação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Animais , Linhagem Celular , Sobrevivência Celular , Humanos , Camundongos
20.
Aging Cell ; 16(4): 661-671, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28455874

RESUMO

Senescent cells are present in premalignant lesions and sites of tissue damage and accumulate in tissues with age. In vivo identification, quantification and characterization of senescent cells are challenging tasks that limit our understanding of the role of senescent cells in diseases and aging. Here, we present a new way to precisely quantify and identify senescent cells in tissues on a single-cell basis. The method combines a senescence-associated beta-galactosidase assay with staining of molecular markers for cellular senescence and of cellular identity. By utilizing technology that combines flow cytometry with high-content image analysis, we were able to quantify senescent cells in tumors, fibrotic tissues, and tissues of aged mice. Our approach also yielded the finding that senescent cells in tissues of aged mice are larger than nonsenescent cells. Thus, this method provides a basis for quantitative assessment of senescent cells and it offers proof of principle for combination of different markers of senescence. It paves the way for screening of senescent cells for identification of new senescence biomarkers, genes that bypass senescence or senolytic compounds that eliminate senescent cells, thus enabling a deeper understanding of the senescent state in vivo.


Assuntos
Envelhecimento/genética , Senescência Celular/genética , Neoplasias/genética , Análise de Célula Única/métodos , Coloração e Rotulagem/métodos , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Biomarcadores/análise , Senescência Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Etoposídeo/farmacologia , Fibrose , Citometria de Fluxo , Expressão Gênica , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Linfócitos/metabolismo , Linfócitos/patologia , Camundongos , Imagem Molecular , Neoplasias/metabolismo , Neoplasias/patologia , Cultura Primária de Células , Células Estromais/metabolismo , Células Estromais/patologia , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA