Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Biochemistry (Mosc) ; 89(5): 839-852, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38880645

RESUMEN

Tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment (TME) and the most abundant population of immune cells infiltrating a tumor. TAMs can largely determine direction of anti-tumor immune response by promoting it or, conversely, contribute to formation of an immunosuppressive TME that allows tumors to evade immune control. Through interactions with tumor cells or other cells in the microenvironment and, as a result of action of anti-cancer therapy, macrophages can enter senescence. In this review, we have attempted to summarize information available in the literature on the role of senescent macrophages in tumors. With the recent development of senolytic therapeutic strategies aimed at removing senescent cells from an organism, it seems important to discuss functions of the senescent macrophages and potential role of the senolytic drugs in reprogramming TAMs to enhance anti-tumor immune response and improve efficacy of cancer treatment.


Asunto(s)
Senescencia Celular , Neoplasias , Microambiente Tumoral , Macrófagos Asociados a Tumores , Microambiente Tumoral/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Biomarcadores de Tumor/metabolismo
2.
Int J Mol Sci ; 25(7)2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38612842

RESUMEN

The tumor microenvironment (TME) plays an essential role in tumor progression and in modulating tumor response to anticancer therapy. Cellular senescence leads to a switch in the cell secretome, characterized by the senescence-associated secretory phenotype (SASP), which may regulate tumorigenesis. Senolytic therapy is considered a novel anticancer strategy that eliminates the deleterious effects of senescent cells in the TME. Here, we show that two different types of senolytic drugs, despite efficiently depleting senescent cells, have opposite effects on cancer-associated fibroblasts (CAFs) and their ability to regulate epithelial-mesenchymal transition (EMT). We found that senolytic drugs, navitoclax and the combination of dasatinib/quercetin, reduced the number of spontaneously senescent and TNF-induced senescent CAFs. Despite the depletion of senescent cells, the combination of dasatinib/quercetin versus navitoclax increased the secretion of the SASP pro-inflammatory cytokine IL-6. This differential effect correlated with the promotion of enhanced migration and EMT in MC38 colorectal cancer cells. Our results demonstrate that some senolytics may have side effects unrelated to their senolytic activity and may promote tumorigenesis. We argue for more careful and extensive studies of the effects of senolytics on various aspects of tumor progression and tumor resistance to therapy before the senolytic strategy is implemented in the clinic.


Asunto(s)
Compuestos de Anilina , Fibroblastos Asociados al Cáncer , Senoterapéuticos , Sulfonamidas , Humanos , Dasatinib/farmacología , Quercetina/farmacología , Carcinogénesis , Transformación Celular Neoplásica , Transición Epitelial-Mesenquimal , Citocinas , Microambiente Tumoral
3.
PLoS Biol ; 18(7): e3000564, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32701952

RESUMEN

Amyloids are protein aggregates with a highly ordered spatial structure giving them unique physicochemical properties. Different amyloids not only participate in the development of numerous incurable diseases but control vital functions in archaea, bacteria and eukarya. Plants are a poorly studied systematic group in the field of amyloid biology. Amyloid properties have not yet been demonstrated for plant proteins under native conditions in vivo. Here we show that seeds of garden pea Pisum sativum L. contain amyloid-like aggregates of storage proteins, the most abundant one, 7S globulin Vicilin, forms bona fide amyloids in vivo and in vitro. Full-length Vicilin contains 2 evolutionary conserved ß-barrel domains, Cupin-1.1 and Cupin-1.2, that self-assemble in vitro into amyloid fibrils with similar physicochemical properties. However, Cupin-1.2 fibrils unlike Cupin-1.1 can seed Vicilin fibrillation. In vivo, Vicilin forms amyloids in the cotyledon cells that bind amyloid-specific dyes and possess resistance to detergents and proteases. The Vicilin amyloid accumulation increases during seed maturation and wanes at germination. Amyloids of Vicilin resist digestion by gastrointestinal enzymes, persist in canned peas, and exhibit toxicity for yeast and mammalian cells. Our finding for the first time reveals involvement of amyloid formation in the accumulation of storage proteins in plant seeds.


Asunto(s)
Amiloide/metabolismo , Pisum sativum/metabolismo , Proteínas de Almacenamiento de Semillas/metabolismo , Semillas/metabolismo , Amiloide/ultraestructura , Detergentes/farmacología , Escherichia coli/metabolismo , Iones , Pancreatina/metabolismo , Pisum sativum/efectos de los fármacos , Pepsina A/metabolismo , Agregado de Proteínas , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Saccharomyces cerevisiae/metabolismo , Proteínas de Almacenamiento de Semillas/química , Proteínas de Almacenamiento de Semillas/farmacología , Proteínas de Almacenamiento de Semillas/ultraestructura
4.
Int J Mol Sci ; 23(19)2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-36232714

RESUMEN

Acute myeloid leukemia (AML) is a rapidly progressing heterogeneous disease with a high mortality rate, which is characterized by hyperproliferation of atypical immature myeloid cells. The number of AML patients is expected to increase in the near future, due to the old-age-associated nature of AML and increased longevity in the human population. RUNX1 and CEBPA, key transcription factors (TFs) of hematopoiesis, are frequently and independently mutated in AML. RUNX1 and CEBPA can bind TET2 demethylase and attract it to their binding sites (TFBS) in cell lines, leading to DNA demethylation of the regions nearby. Since TET2 does not have a DNA-binding domain, TFs are crucial for its guidance to target genomic locations. In this paper, we show that RUNX1 and CEBPA mutations in AML patients affect the methylation of important regulatory sites that resulted in the silencing of several RUNX1 and CEBPA target genes, most likely in a TET2-dependent manner. We demonstrated that hypermethylation of TFBS in AML cells with RUNX1 mutations was associated with resistance to anticancer chemotherapy. Demethylation therapy restored expression of the RUNX1 target gene, BIK, and increased sensitivity of AML cells to chemotherapy. If our results are confirmed, mutations in RUNX1 could be an indication for prescribing the combination of cytotoxic and demethylation therapies.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Leucemia Mieloide Aguda , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , ADN/genética , ADN/metabolismo , Metilación de ADN/genética , Desmetilación/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutación
5.
Proc Natl Acad Sci U S A ; 109(2): E68-75, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22065775

RESUMEN

The inactivation of the p53 tumor suppressor pathway in many cancers often increases their resistance to anticancer therapy. Here we show that a previously proposed strategy directed to Wip1 inhibition could be ineffective in tumors lacking p53. On the contrary, Wip1 overexpression sensitized these tumors to chemotherapeutic agents. This effect was mediated through interaction between Wip1 and RUNX2 that resulted, in response to anticancer treatment, in RUNX2-dependent transcriptional induction of the proapoptotic Bax protein. The potentiating effects of Wip1 overexpression on chemotherapeutic agents were directed only to tumor cells lacking p53. The overexpression of Wip1 in normal tissues provided protection from cisplatin-induced apoptosis through decreased strength of upstream signaling to p53. Thus, Wip1 phosphatase promotes apoptosis in p53-negative tumors and protects normal tissues during treatment with anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias/tratamiento farmacológico , Fosfoproteínas Fosfatasas/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Línea Celular , Cartilla de ADN/genética , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Neoplasias/metabolismo , Plásmidos/genética , Proteína Fosfatasa 2C , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
6.
Nat Genet ; 31(2): 210-5, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12021785

RESUMEN

Expression of oncogenic Ras in primary human cells activates p53, thereby protecting cells from transformation. We show that in Ras-expressing IMR-90 cells, p53 is phosphorylated at Ser33 and Ser46 by the p38 mitogen-activated protein kinase (MAPK). Activity of p38 MAPK is regulated by the p53-inducible phosphatase PPM1D, creating a potential feedback loop. Expression of oncogenic Ras suppresses PPM1D mRNA induction, leaving p53 phosphorylated at Ser33 and Ser46 and in an active state. Retrovirus-mediated overexpression of PPM1D reduced p53 phosphorylation at these sites, abrogated Ras-induced apoptosis and partially rescued cells from cell-cycle arrest. Inactivation of p38 MAPK (the product of Mapk14) in vivo by gene targeting or by PPM1D overexpression expedited tumor formation after injection of mouse embryo fibroblasts (MEFs) expressing E1A+Ras into nude mice. The gene encoding PPM1D (PPM1D, at 17q22/q23) is amplified in human breast-tumor cell lines and in approximately 11% of primary breast tumors, most of which harbor wildtype p53. These findings suggest that inactivation of the p38 MAPK through PPM1D overexpression resulting from PPM1D amplification contributes to the development of human cancers by suppressing p53 activation.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 17 , Amplificación de Genes , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas de Neoplasias , Fosfoproteínas Fosfatasas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Neoplasias de la Mama/etiología , Femenino , Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/fisiología , Fosfoproteínas Fosfatasas/fisiología , Fosforilación , Proteína Fosfatasa 2C , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
7.
Cell Death Discov ; 9(1): 345, 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37726289

RESUMEN

Progress in the development of new sequencing techniques with wider accessibility and higher sensitivity of the protocol of deciphering genome particularities led to the discovery of a new phenomenon - clonal haematopoiesis. It is characterized by the presence in the bloodstream of elderly people a minor clonal population of cells with mutations in certain genes, but without any sign of disease related to the hematopoietic system. Here we will review this recent advancement in the field of clonal haematopoiesis and how it may affect the disease's development in old age.

8.
Nat Cell Biol ; 25(9): 1265-1278, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37652981

RESUMEN

Despite advances in four-factor (4F)-induced reprogramming (4FR) in vitro and in vivo, how 4FR interconnects with senescence remains largely under investigated. Here, using genetic and chemical approaches to manipulate senescent cells, we show that removal of p16High cells resulted in the 4FR of somatic cells into totipotent-like stem cells. These cells expressed markers of both pluripotency and the two-cell embryonic state, readily formed implantation-competent blastoids and, following morula aggregation, contributed to embryonic and extraembryonic lineages. We identified senescence-dependent regulation of nicotinamide N-methyltransferase as a key mechanism controlling the S-adenosyl-L-methionine levels during 4FR that was required for expression of the two-cell genes and acquisition of an extraembryonic potential. Importantly, a partial 4F epigenetic reprogramming in old mice was able to reverse several markers of liver aging only in conjunction with the depletion of p16High cells. Our results show that the presence of p16High senescent cells limits cell plasticity, whereas their depletion can promote a totipotent-like state and histopathological tissue rejuvenation during 4F reprogramming.


Asunto(s)
Plasticidad de la Célula , Reprogramación Celular , Animales , Ratones , Reprogramación Celular/genética , Envejecimiento/genética , Implantación del Embrión , Epigenómica
9.
J Exp Med ; 203(13): 2793-9, 2006 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-17158963

RESUMEN

The ataxia telangiectasia mutated (ATM) kinase is a key tumor suppressor that regulates numerous cell cycle checkpoints as well as apoptosis. Here, we report that ATM is a critical player in the regulation of apoptosis and lymphomagenesis in the presence of c-myc. In turn, deletion of the inhibitory ATM phosphatase, Wip1, results in ATM up-regulation and suppression of Emicro-myc-induced B cell lymphomas. Using mouse genetic crosses, we show that the onset of myc-induced lymphomas is dramatically delayed in Wip1-null mice in an ATM- and p53-, but not p38 MAPK- or Arf-, dependent manner. We propose that Wip1 phosphatase is critical for regulating the ATM-mediated tumor surveillance network.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Linfoma/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/metabolismo , Western Blotting , Inhibidores de Caspasas , Caspasas/metabolismo , Proteínas de Ciclo Celular/genética , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Expresión Génica , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Linfoma/genética , Linfoma/patología , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Proteínas de Neoplasias/genética , Fosfoproteínas Fosfatasas/genética , Fosforilación , Unión Proteica , Proteína Fosfatasa 2C , Proteínas Serina-Treonina Quinasas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Nat Commun ; 12(1): 3622, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131120

RESUMEN

PPM1D/Wip1 is a negative regulator of the tumor suppressor p53 and is overexpressed in several human solid tumors. Recent reports associate gain-of-function mutations of PPM1D in immune cells with worse outcomes for several human cancers. Here we show that mice with genetic knockout of Ppm1d or with conditional knockout of Ppm1d in the hematopoietic system, in myeloid cells, or in neutrophils all display significantly reduced growth of syngeneic melanoma or lung carcinoma tumors. Ppm1d knockout neutrophils infiltrate tumors extensively. Chemical inhibition of Wip1 in human or mouse neutrophils increases anti-tumor phenotypes, p53-dependent expression of co-stimulatory ligands, and proliferation of co-cultured cytotoxic T cells. These results suggest that inhibition of Wip1 in neutrophils enhances immune anti-tumor responses.


Asunto(s)
Daño del ADN , Inmunidad , Neutrófilos/metabolismo , Proteína Fosfatasa 2C/genética , Proteína Fosfatasa 2C/metabolismo , Animales , Antineoplásicos , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Pulmón , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Linfocitos T , Microambiente Tumoral , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
J Gastroenterol ; 56(5): 442-455, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33782752

RESUMEN

BACKGROUND: We previously showed that supernatants of Lactobacillus biofilms induced an anti-inflammatory response by affecting the secretion of macrophage-derived cytokines, which was abrogated upon immunodepletion of the stress protein GroEL. METHODS: We purified GroEL from L. reuteri and analysed its anti-inflammatory properties in vitro in human macrophages isolated from buffy coats, ex vivo in explants from human biopsies and in vivo in a mouse model of DSS induced intestinal inflammation. As a control, we used GroEL purified (LPS-free) from E. coli. RESULTS: We found that L. reuteri GroEL (but not E. coli GroEL) inhibited pro-inflammatory M1-like macrophages markers, and favored M2-like markers. Consequently, L. reuteri GroEL inhibited pro-inflammatory cytokines (TNFα, IL-1ß, IFNγ) while favouring an anti-inflammatory secretome. In colon tissues from human biopsies, L. reuteri GroEL was also able to decrease markers of inflammation and apoptosis (caspase 3) induced by LPS. In mice, we found that rectal administration of L. reuteri GroEL (but not E. coli GroEL) inhibited all signs of haemorrhagic colitis induced by DSS including intestinal mucosa degradation, rectal bleeding and weight loss. It also decreased intestinal production of inflammatory cytokines (such as IFNγ) while increasing anti-inflammatory IL-10 and IL-13. These effects were suppressed when animals were immunodepleted in macrophages. From a mechanistic point of view, the effect of L. reuteri GroEL seemed to involve TLR4, since it was lost in TRL4-/- mice, and the activation of a non-canonical TLR4 pathway. CONCLUSIONS: L. reuteri GroEL, by affecting macrophage inflammatory features, deserves to be explored as an alternative to probiotics.


Asunto(s)
Chaperonina 60/farmacología , Colon/efectos de los fármacos , Inflamación/prevención & control , Lactobacillus/metabolismo , Animales , Chaperonina 60/uso terapéutico , Colon/fisiopatología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Limosilactobacillus reuteri/efectos de los fármacos , Limosilactobacillus reuteri/metabolismo , Ratones Endogámicos BALB C , Estadísticas no Paramétricas
12.
Stem Cells ; 27(6): 1433-42, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19489034

RESUMEN

Continual generation of new neural cells from adult neural stem/progenitor cells (NPCs) is an important component of life-long brain plasticity. However, the intrinsic regulation of this process remains poorly defined. Here we report that Wip1 phosphatase, previously studied in oncogenesis, functions as a crucial physiological regulator in adult neural cell generation. Wip1 deficiency resulted in a 90% decrease in new cell formation in adult olfactory bulb, accompanied by aberrantly decreased NPC amplification, stem cell frequency, and self-renewal. At a cellular level, Wip1 knockout NPCs exhibit a prolonged cell cycle, an accumulation at G(2) to M phase transition, and enhanced p53 activity. Interestingly, the impaired M-phase entry and NPC amplification of Wip1-null mice can be reversed in Wip1/p53 double-null mice. Importantly, there is no difference in NPC amplification between p53-null and Wip1/p53 double-null mice. Our data demonstrate that Wip1 regulates the generation of new neural cells in adult olfactory bulb specifically through p53-dependent M-phase entry of the NPC cell cycle.


Asunto(s)
Células Madre Adultas/citología , Neurogénesis/fisiología , Neuronas/citología , Bulbo Olfatorio/citología , Fosfoproteínas Fosfatasas/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Células Madre Adultas/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Ciclo Celular , Diferenciación Celular/genética , Citometría de Flujo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 2C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Nucleic Acids Res ; 36(22): 7168-80, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19015127

RESUMEN

PPM1D (Wip1), a type PP2C phosphatase, is expressed at low levels in most normal tissues but is overexpressed in several types of cancers. In cells containing wild-type p53, the levels of PPM1D mRNA and protein increase following exposure to genotoxic stress, but the mechanism of regulation by p53 was unknown. PPM1D also has been identified as a CREB-regulated gene due to the presence of a cyclic AMP response element (CRE) in the promoter. Transient transfection and chromatin immunoprecipitation experiments in HCT116 cells were used to characterize a conserved p53 response element located in the 5' untranslated region (UTR) of the PPM1D gene that is required for the p53-dependent induction of transcription from the human PPM1D promoter. CREB binding to the CRE contributes to the regulation of basal expression of PPM1D and directs transcription initiation at upstream sites. Following exposure to ultraviolet (UV) or ionizing radiation, the abundance of transcripts with short 5' UTRs increased in cells containing wild-type p53, indicating increased utilization of downstream transcription initiation sites. In cells containing wild-type p53, exposure to UV resulted in increased PPM1D protein levels even when PPM1D mRNA levels remained constant, indicating post-transcriptional regulation of PPM1D protein levels.


Asunto(s)
Daño del ADN , Fosfoproteínas Fosfatasas/genética , Elementos de Respuesta , Sitio de Iniciación de la Transcripción , Proteína p53 Supresora de Tumor/metabolismo , Regiones no Traducidas 5' , Animales , Secuencia de Bases , Línea Celular Tumoral , Cromatina/metabolismo , Secuencia Conservada , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Fosfoproteínas Fosfatasas/biosíntesis , Filogenia , Regiones Promotoras Genéticas , Proteína Fosfatasa 2C , ARN Mensajero/biosíntesis , Radiación Ionizante , Rayos Ultravioleta
14.
Cell Death Differ ; 27(1): 117-129, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31068676

RESUMEN

Pro-survival stress-inducible chaperone HSP110 is the only HSP for which a mutation has been found in a cancer. Multicenter clinical studies demonstrated a direct association between HSP110 inactivating mutation presence and excellent prognosis in colorectal cancer patients. Here, we have combined crystallographic studies on human HSP110 and in silico modeling to identify HSP110 inhibitors that could be used in colorectal cancer therapy. Two molecules (foldamers 33 and 52), binding to the same cleft of HSP110 nucleotide-binding domain, were selected from a chemical library (by co-immunoprecipitation, AlphaScreening, Interference-Biolayer, Duo-link). These molecules block HSP110 chaperone anti-aggregation activity and HSP110 association to its client protein STAT3, thereby inhibiting STAT3 phosphorylation and colorectal cancer cell growth. These effects were strongly decreased in HSP110 knockdown cells. Foldamer's 33 ability to inhibit tumor growth was confirmed in two colorectal cancer animal models. Although tumor cell death (apoptosis) was noted after treatment of the animals with foldamer 33, no apparent toxicity was observed, notably in epithelial cells from intestinal crypts. Taken together, we identified the first HSP110 inhibitor, a possible drug-candidate for colorectal cancer patients whose unfavorable outcome is associated to HSP110.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas del Choque Térmico HSP110/antagonistas & inhibidores , Animales , Antineoplásicos/toxicidad , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Cristalografía por Rayos X , Proteínas del Choque Térmico HSP110/química , Proteínas del Choque Térmico HSP110/metabolismo , Humanos , Ratones , Modelos Moleculares , Factor de Transcripción STAT3/metabolismo
15.
Oncogene ; 38(15): 2767-2777, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30542121

RESUMEN

A multicenter clinical study demonstrated the presence of a loss-of-function HSP110 mutation in about 15% of colorectal cancers, which resulted from an alternative splicing and was produced at the detriment of wild-type HSP110. Patients expressing low levels of wild-type HSP110 had excellent outcomes (i.e. response to an oxaliplatin-based chemotherapy). Here, we show in vitro, in vivo, and in patients' biopsies that HSP110 co-localizes with DNA damage (γ-H2AX). In colorectal cancer cells, HSP110 translocates into the nucleus upon treatment with genotoxic chemotherapy such as oxaliplatin. Furthermore, we show that HSP110 interacts with the Ku70/Ku80 heterodimer, an essential element of the non-homologous end joining (NHEJ) repair machinery. We also demonstrate by evaluating the resolved 53BP1 foci that depletion in HSP110 impairs repair steps of the NHEJ pathway, which is associated with an increase in DNA double-strand breaks and in the cells' sensitivity to oxaliplatin. HSP110-depleted cells sensitization to oxaliplatin-induced DNA damage is abolished upon re-expression of HSP110. Confirming a role for HSP110 in DNA non-homologous repair, SCR7 and NU7026, two inhibitors of the NHEJ pathway, circumvents HSP110-induced resistance to chemotherapy. In conclusion, HSP110 through its interaction with the Ku70/80 heterodimer may participate in DNA repair, thereby inducing a protection against genotoxic therapy.


Asunto(s)
Núcleo Celular/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Reparación del ADN por Unión de Extremidades/genética , Proteínas del Choque Térmico HSP110/genética , Mutágenos/farmacología , Translocación Genética/genética , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Proteínas de Unión al ADN/genética , Células HCT116 , Humanos , Autoantígeno Ku/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Oxaliplatino/farmacología , Translocación Genética/efectos de los fármacos
16.
Mol Cell Biol ; 25(15): 6639-48, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16024799

RESUMEN

ING2 is a candidate tumor suppressor gene that can activate p53 by enhancing its acetylation. Here, we demonstrate that ING2 is also involved in p53-mediated replicative senescence. ING2 protein expression increased in late-passage human primary cells, and it colocalizes with serine 15-phosphorylated p53. ING2 and p53 also complexed with the histone acetyltransferase p300. ING2 enhanced the interaction between p53 and p300 and acted as a cofactor for p300-mediated p53 acetylation. The level of ING2 expression directly modulated the onset of replicative senescence. While overexpression of ING2 induced senescence in young fibroblasts in a p53-dependent manner, expression of ING2 small interfering RNA delayed the onset of senescence. Hence, ING2 can act as a cofactor of p300 for p53 acetylation and thereby plays a positive regulatory role during p53-mediated replicative senescence.


Asunto(s)
Senescencia Celular/fisiología , Proteínas de Homeodominio/fisiología , Proteínas Nucleares/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Transactivadores/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/fisiología , Acetilación , División Celular/fisiología , Línea Celular , Proliferación Celular , Proteínas de Homeodominio/biosíntesis , Humanos , Proteínas Nucleares/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores Citoplasmáticos y Nucleares/biosíntesis , Serina/metabolismo , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/biosíntesis
17.
Cancer Res ; 66(6): 2928-36, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540640

RESUMEN

Activation of apoptosis is believed to be critical for the role of p53 as a tumor suppressor. Here, we report a new mouse strain carrying a human p53 transgene in the mouse p53-null background. Expression of human p53 in these mice was comparable with wild-type murine p53; however, transactivation, induction of apoptosis, and G(1)-S checkpoint, but not transrepression or regulation of a centrosomal checkpoint, were deregulated. Although multiple functions of p53 were abrogated, mice carrying the human p53 transgene did not show early onset of tumors as typically seen for p53-null mice. In contrast, human p53 in the p53-null background did not prevent accelerated tumor development after genotoxic or oncogenic stress. Such behavior of human p53 expressed at physiologic levels in transgenic cells could be explained by unexpectedly high binding with Mdm2. By using Nutlin-3a, an inhibitor of the interaction between Mdm2 and p53, we were able to partially reconstitute p53 transactivation and apoptosis in transgenic cells. Our findings indicate that the interaction between p53 and Mdm2 controls p53 transcriptional activity in homeostatic tissues and regulates DNA damage- and oncogene-induced, but not spontaneous, tumorigenesis.


Asunto(s)
Apoptosis/genética , Genes p53 , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Transgenes , Proteína p53 Supresora de Tumor/biosíntesis , Animales , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteína p53 Supresora de Tumor/genética
18.
Mol Cell Biol ; 22(4): 1094-105, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11809801

RESUMEN

The Wip1 gene is a serine/threonine phosphatase that is induced in a p53-dependent manner by DNA-damaging agents. We show here that Wip1 message is expressed in moderate levels in all organs, but is present at very high levels in the testes, particularly in the postmeiotic round spermatid compartment of the seminiferous tubules. We have confirmed that Wip1 mRNA is induced by ionizing radiation in mouse tissues in a p53-dependent manner. To further determine the normal biological function of Wip1 in mammalian organisms, we have generated Wip1-deficient mice. Wip1 null mice are viable but show a variety of postnatal abnormalities, including variable male runting, male reproductive organ atrophy, reduced male fertility, and reduced male longevity. Mice lacking Wip1 show increased susceptibility to pathogens and diminished T- and B-cell function. Fibroblasts derived from Wip1 null embryos have decreased proliferation rates and appear to be compromised in entering mitosis. The data are consistent with an important role for Wip1 in spermatogenesis, lymphoid cell function, and cell cycle regulation.


Asunto(s)
Ciclo Celular/fisiología , Sistema Inmunológico/fisiología , Proteínas de Neoplasias , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/fisiología , Reproducción , Animales , Linfocitos B/inmunología , Constitución Corporal , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Desarrollo Embrionario y Fetal , Femenino , Regulación de la Expresión Génica , Marcación de Gen , Humanos , Hibridación in Situ , Riñón/fisiología , Riñón/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Fosfatasa 2C , ARN Mensajero/metabolismo , Radiación Ionizante , Espermatogénesis , Bazo/fisiología , Bazo/efectos de la radiación , Células Madre/fisiología , Linfocitos T/inmunología , Testículo/fisiología , Testículo/efectos de la radiación
19.
Oncotarget ; 7(21): 31563-71, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-26883196

RESUMEN

Cells undergoing oncogenic transformation frequently inactivate tumor suppressor pathways that could prevent their uncontrolled growth. Among those pathways p53 and p38MAPK pathways play a critical role in regulation of cell cycle, senescence and cell death in response to activation of oncogenes, stress and DNA damage. Consequently, these two pathways are important in determining the sensitivity of tumor cells to anti-cancer treatment. Wild type p53-induced phosphatase, Wip1, is involved in governance of both pathways. Recently, strategies directed to manipulation with Wip1 activity proposed to advance current day anticancer treatment and novel chemical compounds synthesized to improve specificity of manipulation with Wip1 activity. Here we reviewed the history of Wip1 studies in vitro and in vivo, in genetically modified animal models that support Wip1 role in tumorigenesis through regulation of p53 and p38MAPK pathways. Based on our knowledge we propose several recommendations for future more accurate studies of Wip1 interactions with other pathways involved in tumorigenesis using recently developed tools and for adoption of Wip1 manipulation strategies in anti-cancer therapy.


Asunto(s)
Proteína Fosfatasa 2C/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Daño del ADN , Humanos , Mutación , Proteína Fosfatasa 2C/genética
20.
Cancer Biol Ther ; 4(10): 1154-8, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16258255

RESUMEN

Wip1 is an amplified oncogene whose deletion causes a tumor resistant phenotype in mice. These observations provide justification for a search for Wip1 chemical inhibitors as potential anticancer drugs. Here we report a group of Wip1 inhibitors with anticancer properties both in vitro and in vivo. In vitro, inactivation of Wip1 reduces the proliferation rate of breast cancer cell lines and enhances growth inhibition caused by doxorubicin. In vivo, administration of Wip1 inhibitors decreases proliferation of xenograph tumors and tumors developed in MMTV-c-Neu transgenic mice. We propose that these agents may serve as lead compounds for the development of anticancer drugs targeting Wip1 phosphatase.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Adenoviridae/genética , Animales , Neoplasias de la Mama/prevención & control , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Viral , Embrión de Mamíferos/citología , Inhibidores Enzimáticos/química , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Genes ras , Humanos , Ratones , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Conformación Molecular , Trasplante de Neoplasias , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/aislamiento & purificación , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2C , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda