Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Blood ; 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39226460

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of patients with relapsed or refractory hematologic malignancies, but it comes with unique toxicities, notably cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). As experience with CAR T-cell therapy grows, distinct and infrequent neurological complications are becoming increasingly evident. Recently, reports of acute myelopathy following the administration of CAR T-cell therapies have been accumulating. Despite the establishment of consensus guidelines for managing ICANS, there remains limited guidance on appropriate investigations and treatments for this rare complication. Here, we delve into the clinical features, pathophysiology, and strategies for optimally managing acute myelitis after CAR T-cell therapy, drawing insights from reported cases in the literature.

2.
Genes Dev ; 27(8): 900-15, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23599344

RESUMEN

Constitutive activation of growth factor signaling pathways paradoxically triggers a cell cycle arrest known as cellular senescence. In primary cells expressing oncogenic ras, this mechanism effectively prevents cell transformation. Surprisingly, attenuation of ERK/MAP kinase signaling by genetic inactivation of Erk2, RNAi-mediated knockdown of ERK1 or ERK2, or MEK inhibitors prevented the activation of the senescence mechanism, allowing oncogenic ras to transform primary cells. Mechanistically, ERK-mediated senescence involved the proteasome-dependent degradation of proteins required for cell cycle progression, mitochondrial functions, cell migration, RNA metabolism, and cell signaling. This senescence-associated protein degradation (SAPD) was observed not only in cells expressing ectopic ras, but also in cells that senesced due to short telomeres. Individual RNAi-mediated inactivation of SAPD targets was sufficient to restore senescence in cells transformed by oncogenic ras or trigger senescence in normal cells. Conversely, the anti-senescence viral oncoproteins E1A, E6, and E7 prevented SAPD. In human prostate neoplasms, high levels of phosphorylated ERK were found in benign lesions, correlating with other senescence markers and low levels of STAT3, one of the SAPD targets. We thus identified a mechanism that links aberrant activation of growth signaling pathways and short telomeres to protein degradation and cellular senescence.


Asunto(s)
Senescencia Celular/genética , Sistema de Señalización de MAP Quinasas/fisiología , Proteolisis , Animales , Línea Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/enzimología , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas ras/metabolismo
3.
Mol Cell ; 36(5): 754-67, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005840

RESUMEN

SOCS1 is lost in many human tumors, but its tumor suppression activities are not well understood. We report that SOCS1 is required for transcriptional activity, DNA binding, and serine 15 phosphorylation of p53 in the context of STAT5 signaling. In agreement, inactivation of SOCS1 disabled p53-dependent senescence in response to oncogenic STAT5A and radiation-induced apoptosis in T cells. In addition, SOCS1 was sufficient to induce p53-dependent senescence in fibroblasts. The mechanism of activation of p53 by SOCS1 involved a direct interaction between the SH2 domain of SOCS1 and the N-terminal transactivation domain of p53, while the C-terminal domain of SOCS1 containing the SOCS Box mediated interaction with the DNA damage-regulated kinases ATM/ATR. Also, SOCS1 colocalized with ATM at DNA damage foci induced by oncogenic STAT5A. Collectively, these results add another component to the p53 and DNA damage networks and reveal a mechanism by which SOCS1 functions as a tumor suppressor.


Asunto(s)
Senescencia Celular , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/análisis , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
4.
Leuk Lymphoma ; : 1-9, 2024 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-39129334

RESUMEN

This study reports characteristics and outcomes of adults who received Azacitidine-Venetoclax (AZA-VEN) compared to other salvage therapies (NO-AZA-VEN) as first salvage therapy for acute myeloid leukemia (AML). The clinical data of 81 patients with a diagnosis of relapsed or refractory AML were analyzed. The ORR was comparable for both groups (55% vs 57%, p = 0.852). Median OS (6.8 vs 11.2 months, p = 0.053) and median RFS (6.9 vs 11.2 months, p = 0.488) showed a trend in favor of the NO-AZA-VEN group. OS was significantly longer with NO-AZA-VEN for ELN 2022 risk category sub-group, patients under 60 years old, primary AML and for patients who underwent allo-hematopoietic stem cell transplant after salvage therapy. There was no statistical difference in complications of treatment such as febrile neutropenia, intensive care unit stay, septic shock and total parenteral nutrition. Those results do not support the preferential use of AZA-VEN over other regimens in R/R acute myeloid leukemia.

5.
Cancers (Basel) ; 16(1)2023 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-38201521

RESUMEN

This comprehensive review delves into the multifaceted aspects of ERK signaling and the intricate mechanisms underlying distinct cellular fates. ERK1 and ERK2 (ERK) govern proliferation, transformation, epithelial-mesenchymal transition, differentiation, senescence, or cell death, contingent upon activation strength, duration, and context. The biochemical mechanisms underlying these outcomes are inadequately understood, shaped by signaling feedback and the spatial localization of ERK activation. Generally, ERK activation aligns with the Goldilocks principle in cell fate determination. Inadequate or excessive ERK activity hinders cell proliferation, while balanced activation promotes both cell proliferation and survival. Unraveling the intricacies of how the degree of ERK activation dictates cell fate requires deciphering mechanisms encompassing protein stability, transcription factors downstream of ERK, and the chromatin landscape.

6.
Thromb Res ; 205: 29-39, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34246011

RESUMEN

OBJECTIVES: Venous thrombotic events (VTEs) are a frequent complication of non-small cell lung cancer (NSCLC) and are associated with increased morbidity. Immune checkpoint inhibitors (ICIs) are revolutionizing the management of NSCLC, but little is known about their impact on thrombosis. This study aims to define the incidence and clinical relevance of VTEs in NSCLC patients receiving these treatments. METHODS: A retrospective multicentric cohort study including 593 patients from three centers in Canada and France was performed. The cumulative incidence of VTEs after ICIs was estimated using competing risk analysis, and the association of these events with survival and response to treatment was determined. Finally, univariate and multivariate tests were performed to identify VTE risk factors. RESULTS: The cumulative incidence of VTEs in the cohort was 14.8% (95% CI = 7.4-22.2%) for an incidence rate of 76.5 (95% CI = 59.9-97.8) thrombosis per 1000 person-years, with most thromboses occurring rapidly after treatment initiation. VTEs were not correlated with overall survival, progression-free survival, or objective response to ICIs. Age ˂ 65 years old (HR = 2.00; 95% CI = 1.11-3.59) and tumors with PD-L1 1-49% (HR = 3.36; 95% CI = 1.19-9.50) or PD-L1 ≥ 50% (HR = 3.22; 95% CI = 1.21-8.57) were associated with more VTEs after 12 months of ICI initiation. Also, a delay of less than 12 months from diagnosis to the first ICI treatment (HR = 2.06; 95% CI = 1.09-3.89) and active smoking (HR = 2.00; 95% CI = 1.12-3.58) are probable risk factors of VTEs. CONCLUSION: This study suggests that the incidence of VTEs in NSCLC patients treated with ICIs is comparable to what is reported in other cohorts of patients treated with chemotherapy. In our cohort, VTEs were not associated with a decreased survival or response to therapy. Patient age < 65 and tumors with PD-L1 ≥ 1% were associated with a higher risk of VTEs under ICIs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Trombosis , Anciano , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Estudios de Cohortes , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias Pulmonares/tratamiento farmacológico , Estudios Retrospectivos , Trombosis/inducido químicamente , Trombosis/epidemiología
7.
Aging Cell ; 18(2): e12889, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30614183

RESUMEN

Most cancers arise in old individuals, which also accumulate senescent cells. Cellular senescence can be experimentally induced by expression of oncogenes or telomere shortening during serial passage in culture. In vivo, precursor lesions of several cancer types accumulate senescent cells, which are thought to represent a barrier to malignant progression and a response to the aberrant activation of growth signaling pathways by oncogenes (oncogene toxicity). Here, we sought to define gene expression changes associated with cells that bypass senescence induced by oncogenic RAS. In the context of pancreatic ductal adenocarcinoma (PDAC), oncogenic KRAS induces benign pancreatic intraepithelial neoplasias (PanINs), which exhibit features of oncogene-induced senescence. We found that the bypass of senescence in PanINs leads to malignant PDAC cells characterized by gene signatures of epithelial-mesenchymal transition, stem cells, and mitochondria. Stem cell properties were similarly acquired in PanIN cells treated with LPS, and in primary fibroblasts and mammary epithelial cells that bypassed Ras-induced senescence after reduction of ERK signaling. Intriguingly, maintenance of cells that circumvented senescence and acquired stem cell properties was blocked by metformin, an inhibitor of complex I of the electron transport chain or depletion of STAT3, a protein required for mitochondrial functions and stemness. Thus, our studies link bypass of senescence in premalignant lesions to loss of differentiation, acquisition of stemness features, and increased reliance on mitochondrial functions.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Metformina/farmacología , Células Madre/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Células Madre/citología , Relación Estructura-Actividad , Células Tumorales Cultivadas
8.
Nat Cell Biol ; 20(7): 789-799, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29941930

RESUMEN

Cellular senescence is a tumour suppressor programme characterized by a stable cell cycle arrest. Here we report that cellular senescence triggered by a variety of stimuli leads to diminished ribosome biogenesis and the accumulation of both rRNA precursors and ribosomal proteins. These defects were associated with reduced expression of several ribosome biogenesis factors, the knockdown of which was also sufficient to induce senescence. Genetic analysis revealed that Rb but not p53 was required for the senescence response to altered ribosome biogenesis. Mechanistically, the ribosomal protein S14 (RPS14 or uS11) accumulates in the soluble non-ribosomal fraction of senescent cells, where it binds and inhibits CDK4 (cyclin-dependent kinase 4). Overexpression of RPS14 is sufficient to inhibit Rb phosphorylation, inducing cell cycle arrest and senescence. Here we describe a mechanism for maintaining the senescent cell cycle arrest that may be relevant for cancer therapy, as well as biomarkers to identify senescent cells.


Asunto(s)
Puntos de Control del Ciclo Celular , Senescencia Celular , Neoplasias/metabolismo , Proteína de Retinoblastoma/metabolismo , Ribosomas/metabolismo , Factores de Coagulación Sanguínea/genética , Factores de Coagulación Sanguínea/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Células HEK293 , Humanos , Neoplasias/genética , Neoplasias/patología , Células PC-3 , Fosforilación , Unión Proteica , Precursores del ARN/biosíntesis , Precursores del ARN/genética , ARN Ribosómico/biosíntesis , ARN Ribosómico/genética , Proteínas de Unión al ARN , Proteína de Retinoblastoma/genética , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Ribosomas/genética , Transducción de Señal , Factores de Tiempo
10.
Cancer Res ; 74(2): 412-9, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24408923

RESUMEN

The extracellular signal-regulated kinase ERK1 and ERK2 (ERK1/2) cascade regulates a variety of cellular processes by phosphorylating multiple target proteins. The outcome of its activation ranges from stimulation of cell survival and proliferation to triggering tumor suppressor responses such as cell differentiation, cell senescence, and apoptosis. This pathway is intimately linked to cancer as several of its upstream activators are frequently mutated in human disease and are shown to accelerate tumorigenesis when engineered in the mouse genome. However, measurement of activated ERKs in human cancers or mouse models does not always support a role in tumorigenesis, and data consistent with a role in tumor suppression have been reported as well. The intensity of ERK signaling, negative feedback loops that regulate the pathway, and cross-talks with other signaling pathways, seem to be of primary importance in determining the final cellular outcome. Cell senescence, a putative tumor-suppression mechanism, depends on high-intensity ERK signals that trigger phosphorylation-dependent protein degradation of multiple proteins required for cell-cycle progression. This response may be circumvented during carcinogenesis by a variety of mechanisms, some of them yet to be discovered, which in essence turn ERK functions from tumor suppression to tumor promotion. The use of pharmacologic inhibitors targeting this pathway must be carefully evaluated so they are applied to cases in which ERKs are mainly oncogenic.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias/enzimología , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica , Senescencia Celular , Inhibidores Enzimáticos/uso terapéutico , Eliminación de Gen , Genes Supresores de Tumor , Humanos , Isoenzimas/metabolismo , Ratones , Mutación , Fenotipo , Fosforilación
11.
Cell Cycle ; 13(12): 1840-58, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24866342

RESUMEN

Autophagy and the ubiquitin-proteasome pathway (UPP) are the major protein degradation systems in eukaryotic cells. Whereas the former mediate a bulk nonspecific degradation, the UPP allows a rapid degradation of specific proteins. Both systems have been shown to play a role in tumorigenesis, and the interest in developing therapeutic agents inhibiting protein degradation is steadily growing. However, emerging data point to a critical role for autophagy in cellular senescence, an established tumor suppressor mechanism. Recently, a selective protein degradation process mediated by the UPP was also shown to contribute to the senescence phenotype. This process is tightly regulated by E3 ubiquitin ligases, deubiquitinases, and several post-translational modifications of target proteins. Illustrating the complexity of UPP, more than 600 human genes have been shown to encode E3 ubiquitin ligases, a number which exceeds that of the protein kinases. Nevertheless, our knowledge of proteasome-dependent protein degradation as a regulated process in cellular contexts such as cancer and senescence remains very limited. Here we discuss the implications of protein degradation in senescence and attempt to relate this function to the protein degradation pattern observed in cancer cells.


Asunto(s)
Senescencia Celular , Neoplasias/patología , Proteolisis , Autofagia , Puntos de Control del Ciclo Celular , Nucléolo Celular/metabolismo , Daño del ADN , Humanos , Neoplasias/metabolismo , Proteoma/metabolismo , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
12.
J Cell Biol ; 202(3): 403-5, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23918935

RESUMEN

Chronic inflammation drives initiation and progression of many malignancies, including pancreatic cancer. In this issue, Liou et al. (2013. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201301001) report that inflammatory macrophages are major players in the earliest stages of pancreatic cancer. They show that paracrine signals from the macrophages activate the nuclear factor κB transcriptional program in normal pancreatic acinar cells, resulting in acinar-ductal metaplasia, a dedifferentiated state that is poised for oncogenic transformation.


Asunto(s)
Células Acinares/metabolismo , Citocinas/metabolismo , Macrófagos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , FN-kappa B/metabolismo , Páncreas/metabolismo , Animales
13.
Aging Cell ; 12(3): 489-98, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23521863

RESUMEN

We show that the antidiabetic drug metformin inhibits the expression of genes coding for multiple inflammatory cytokines seen during cellular senescence. Conditioned medium (CM) from senescent cells stimulates the growth of prostate cancer cells but treatment of senescent cells with metformin inhibited this effect. Bioinformatic analysis of genes downregulated by metformin suggests that the drug blocks the activity of the transcription factor NF-κB. In agreement, metformin prevented the translocation of NF-κB to the nucleus and inhibited the phosphorylation of IκB and IKKα/ß, events required for activation of the NF-κB pathway. These effects were not dependent on AMPK activation or on the context of cellular senescence, as metformin inhibited the NF-κB pathway stimulated by lipopolysaccharide (LPS) in ampk null fibroblasts and in macrophages. Taken together, our results provide a novel mechanism for the antiaging and antineoplastic effects of metformin reported in animal models and in diabetic patients taking this drug.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Metformina/farmacología , FN-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Citocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Hipoglucemiantes/farmacología , Inflamación/genética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Cancer Prev Res (Phila) ; 5(4): 536-43, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22262811

RESUMEN

Pharmacoepidemiologic studies provide evidence that use of metformin, a drug commonly prescribed for type II diabetes, is associated with a substantial reduction in cancer risk. Experimental models show that metformin inhibits the growth of certain neoplasms by cell autonomous mechanisms such as activation of AMP kinase with secondary inhibition of protein synthesis or by an indirect mechanism involving reduction in gluconeogenesis leading to a decline in insulin levels and reduced proliferation of insulin-responsive cancers. Here, we show that metformin attenuates paraquat-induced elevations in reactive oxygen species (ROS), and related DNA damage and mutations, but has no effect on similar changes induced by H(2)0(2), indicating a reduction in endogenous ROS production. Importantly, metformin also inhibited Ras-induced ROS production and DNA damage. Our results reveal previously unrecognized inhibitory effects of metformin on ROS production and somatic cell mutation, providing a novel mechanism for the reduction in cancer risk reported to be associated with exposure to this drug.


Asunto(s)
Daño del ADN , Hipoglucemiantes/farmacología , Metformina/farmacología , Especies Reactivas de Oxígeno , Adenilato Quinasa/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 2/genética , Ensayo de Inmunoadsorción Enzimática/métodos , Factor de Crecimiento Epidérmico/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Peróxido de Hidrógeno/metabolismo , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos , Mutagénesis , Mutación , NADP/metabolismo
15.
Mol Cell Biol ; 29(16): 4495-507, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19528227

RESUMEN

The expression of oncogenic ras in normal human cells quickly induces an aberrant proliferation response that later is curtailed by a cell cycle arrest known as cellular senescence. Here, we show that cells expressing oncogenic ras display an increase in the mitochondrial mass, the mitochondrial DNA, and the mitochondrial production of reactive oxygen species (ROS) prior to the senescent cell cycle arrest. By the time the cells entered senescence, dysfunctional mitochondria accumulated around the nucleus. The mitochondrial dysfunction was accompanied by oxidative DNA damage, a drop in ATP levels, and the activation of AMPK. The increase in mitochondrial mass and ROS in response to oncogenic ras depended on intact p53 and Rb tumor suppression pathways. In addition, direct interference with mitochondrial functions by inhibiting the expression of the Rieske iron sulfur protein of complex III or the use of pharmacological inhibitors of the electron transport chain and oxidative phosphorylation was sufficient to trigger senescence. Taking these results together, this work suggests that mitochondrial dysfunction is an effector pathway of oncogene-induced senescence.


Asunto(s)
Senescencia Celular/genética , Genes ras , Mitocondrias , Proteínas ras , Adenilato Quinasa/metabolismo , Animales , Ciclo Celular/fisiología , Línea Celular , Senescencia Celular/fisiología , Daño del ADN , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Activación Enzimática , Perfilación de la Expresión Génica , Genes Mitocondriales , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA