Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Eur Respir J ; 58(2)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33509955

RESUMEN

BACKGROUND: Cell senescence is a key process in age-associated dysfunction and diseases, notably chronic obstructive pulmonary disease (COPD). We previously identified phospholipase A2 receptor 1 (PLA2R1) as a positive regulator of cell senescence acting via Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling. Its role in pathology, however, remains unknown. Here, we assessed PLA2R1-induced senescence in COPD and lung emphysema pathogenesis. METHODS: We assessed cell senescence in lungs and cultured lung cells from patients with COPD and controls subjected to PLA2R1 knockdown, PLA2R1 gene transduction and treatment with the JAK1/2 inhibitor ruxolitinib. To assess whether PLA2R1 upregulation caused lung lesions, we developed transgenic mice overexpressing PLA2R1 (PLA2R1-TG) and intratracheally injected wild-type mice with a lentiviral vector carrying the Pla2r1 gene (LV-PLA2R1 mice). RESULTS: We found that PLA2R1 was overexpressed in various cell types exhibiting senescence characteristics in COPD lungs. PLA2R1 knockdown extended the population doubling capacity of these cells and inhibited their pro-inflammatory senescence-associated secretory phenotype (SASP). PLA2R1-mediated cell senescence in COPD was largely reversed by treatment with the potent JAK1/2 inhibitor ruxolitinib. Five-month-old PLA2R1-TG mice exhibited lung cell senescence, and developed lung emphysema and lung fibrosis together with pulmonary hypertension. Treatment with ruxolitinib induced reversal of lung emphysema and fibrosis. LV-PLA2R1-treated mice developed lung emphysema within 4 weeks and this was markedly attenuated by concomitant ruxolitinib treatment. CONCLUSIONS: Our data support a major role for PLA2R1 activation in driving lung cell senescence and lung alterations in COPD. Targeting JAK1/2 may represent a promising therapeutic approach for COPD.


Asunto(s)
Enfisema , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Animales , Senescencia Celular , Humanos , Pulmón , Ratones , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Receptores de Fosfolipasa A2
2.
Gut ; 68(4): 693-707, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30415234

RESUMEN

OBJECTIVE: Pancreatic cancer is associated with an abundant stromal reaction leading to immune escape and tumour growth. This massive stroma drives the immune escape in the tumour. We aimed to study the impact of ßig-h3 stromal protein in the modulation of the antitumoural immune response in pancreatic cancer. DESIGN: We performed studies with p48-Cre;KrasG12D, pdx1-Cre;KrasG12D;Ink4a/Arffl/fl, pdx1-Cre;KrasG12D; p53R172H mice and tumour tissues from patients with pancreatic ductal adenocarcinoma (PDA). Some transgenic mice were given injections of anti-ßig-h3, anti-CD8, anti-PD1 depleting antibodies. Tumour growth as well as modifications in the activation of local immune cells were analysed by flow cytometry, immunohistochemistry and immunofluorescence. Tissue stiffness was measured by atomic force microscopy. RESULTS: We identified ßig-h3 stromal-derived protein as a key actor of the immune paracrine interaction mechanism that drives pancreatic cancer. We found that ßig-h3 is highly produced by cancer-associated fibroblasts in the stroma of human and mouse. This protein acts directly on tumour-specific CD8+ T cells and F4/80 macrophages. Depleting ßig-h3 in vivo reduced tumour growth by enhancing the number of activated CD8+ T cell within the tumour and subsequent apoptotic tumour cells. Furthermore, we found that targeting ßig-h3 in established lesions released the tissue tension and functionally reprogrammed F4/80 macrophages in the tumour microenvironment. CONCLUSIONS: Our data indicate that targeting stromal extracellular matrix protein ßig-h3 improves the antitumoural response and consequently reduces tumour weight. Our findings present ßig-h3 as a novel immunological target in pancreatic cancer.


Asunto(s)
Adenocarcinoma/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma Ductal Pancreático/inmunología , Proteínas de la Matriz Extracelular/inmunología , Neoplasias Pancreáticas/inmunología , Factor de Crecimiento Transformador beta/inmunología , Microambiente Tumoral/inmunología , Animales , Fibroblastos/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Microscopía de Fuerza Atómica , Comunicación Paracrina/inmunología
3.
Int J Cancer ; 135(6): 1319-29, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24615579

RESUMEN

The clinical efficacy of anti-angiogenic monotherapies in metastatic breast cancer is less than originally anticipated, and it is not clear what the response of bone metastasis to anti-angiogenic therapies is. Here, we examined the impact of neutralizing tumor-derived vascular endothelial growth factor (VEGF) in animal models of subcutaneous tumor growth and bone metastasis formation. Silencing of VEGF expression (Sh-VEGF) in osteotropic human MDA-MB-231/B02 breast cancer cells led to a substantial growth inhibition of subcutaneous Sh-VEGF B02 tumor xenografts, as a result of reduced angiogenesis, when compared to that observed with animals bearing mock-transfected (Sc-VEGF) B02 tumors. However, there was scant evidence that either the silencing of tumor-derived VEGF or the use of a VEGF-neutralizing antibody (bevacizumab) affected B02 breast cancer bone metastasis progression in animals. We also examined the effect of vatalanib (a VEGF receptor tyrosine kinase inhibitor) in this mouse model of bone metastasis. However, vatalanib failed to inhibit bone metastasis caused by B02 breast cancer cells. In sharp contrast, vatalanib in combination with bevacizumab reduced not only bone destruction but also skeletal tumor growth in animals bearing breast cancer bone metastases, when compared with either agent alone. Thus, our study highlights the importance of targeting both the tumor compartment and the host tissue (i.e., skeleton) to efficiently block the development of bone metastasis. We believe this is a crucially important observation as the clinical benefit of anti-angiogenic monotherapies in metastatic breast cancer is relatively modest.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Óseas/secundario , Neoplasias Óseas/terapia , Neoplasias de la Mama/terapia , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Bevacizumab , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/genética , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Huésped Inmunocomprometido , Ratones , Ratones Endogámicos C3H , Osteólisis/tratamiento farmacológico , Osteólisis/patología , Ftalazinas/administración & dosificación , Embarazo , Piridinas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Transfección , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Redox Biol ; 73: 103204, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38810421

RESUMEN

The ELN gene encodes tropoelastin which is used to generate elastic fibers that insure proper tissue elasticity. Decreased amounts of elastic fibers and/or accumulation of bioactive products of their cleavage, named elastokines, are thought to contribute to aging. Cellular senescence, characterized by a stable proliferation arrest and by the senescence-associated secretory phenotype (SASP), increases with aging, fostering the onset and progression of age-related diseases and overall aging, and has so far never been linked with elastin. Here, we identified that decrease in ELN either by siRNA in normal human fibroblasts or by knockout in mouse embryonic fibroblasts results in premature senescence. Surprisingly this effect is independent of elastic fiber degradation or elastokines production, but it relies on the rapid increase in HMOX1 after ELN downregulation. Moreover, the induction of HMOX1 depends on p53 and NRF2 transcription factors, and leads to an increase in iron, further mediating ELN downregulation-induced senescence. Screening of iron-dependent DNA and histones demethylases revealed a role for histone PHF8 demethylase in mediating ELN downregulation-induced senescence. Collectively, these results unveil a role for ELN in protecting cells from cellular senescence through a non-canonical mechanism involving a ROS/HMOX1/iron accumulation/PHF8 histone demethylase pathway reprogramming gene expression towards a senescence program.


Asunto(s)
Senescencia Celular , Fibroblastos , Regulación de la Expresión Génica , Hemo-Oxigenasa 1 , Hierro , Tropoelastina , Animales , Humanos , Ratones , Fibroblastos/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Tropoelastina/metabolismo , Tropoelastina/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética
5.
Mol Biomed ; 4(1): 4, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36739330

RESUMEN

Pancreatic cancer is one of the deadliest cancers owing to its late diagnosis and of the strong resistance to available treatments. Despite a better understanding of the disease in the last two decades, no significant improvement in patient care has been made. Senescent cells are characterized by a stable proliferation arrest and some resistance to cell death. Increasing evidence suggests that multiple lines of antitumor therapy can induce a senescent-like phenotype in cancer cells, which may participate in treatment resistance. In this study, we describe that gemcitabine, a clinically-used drug against pancreatic cancer, induces a senescent-like phenotype in highly chemoresistant pancreatic cancer cells in vitro and in xenografted tumors in vivo. The use of ABT-263, a well-described senolytic compound targeting Bcl2 anti-apoptotic proteins, killed pancreatic gemcitabine-treated senescent-like cancer cells in vitro. In vivo, the combination of gemcitabine and ABT-263 decreased tumor growth, whereas their individual administration had no effect. Together these data highlight the possibility of improving the efficacy of conventional chemotherapies against pancreatic cancer by eliminating senescent-like cancer cells through senolytic intervention. Further studies testing different senolytics or their combination with available treatments will be necessary to optimize preclinical data in mouse models before transferring these findings to clinical trials.

6.
Aging Cell ; 22(11): e13971, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37667516

RESUMEN

Cellular senescence is induced by many stresses including telomere shortening, DNA damage, oxidative, or metabolic stresses. Senescent cells are stably cell cycle arrested and they secrete many factors including cytokines and chemokines. Accumulation of senescent cells promotes many age-related alterations and diseases. In this study, we investigated the role of the pro-senescent phospholipase A2 receptor 1 (PLA2R1) in regulating some age-related alterations in old mice and in mice subjected to a Western diet, whereas aged wild-type mice displayed a decreased ability to regulate their glycemia during glucose and insulin tolerance tests, aged Pla2r1 knockout (KO) mice efficiently regulated their glycemia and displayed fewer signs of aging. Loss of Pla2r1 was also found protective against the deleterious effects of a Western diet. Moreover, these Pla2r1 KO mice were partially protected from diet-induced senescent cell accumulation, steatosis, and fibrosis. Together these results support that Pla2r1 drives several age-related alterations, especially in the liver, arising during aging or through a Western diet.


Asunto(s)
Envejecimiento , Dieta Occidental , Animales , Ratones , Envejecimiento/genética , Senescencia Celular/genética , Ratones Noqueados , Acortamiento del Telómero
7.
Cancer Lett ; 546: 215850, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-35926818

RESUMEN

Oncogenic stress-induced senescence initially inhibits tumor initiation by blocking proliferation and by attracting immune cells to clear potentially harmful cells. If these cells are not eliminated they may resume proliferation upon loss-of-tumor suppressors, and be at risk of transformation. During tumor formation, depending on the sequence of events of gain-of-oncogenes and/or loss-of-tumor suppressors, cancer cells may emerge from senescent cells. Here, we show that these transformed cells after senescence (TS) display more aggressive tumorigenic features, with a greater capacity to migrate and a higher resistance to anti-tumoral drugs than cells having undergone transformation without senescence. Bulk transcriptomic analysis and single cell RNA sequencing revealed a signature unique to TS cells. A score of this signature was then generated and a high score was correlated with decreased survival of patients with lung adenocarcinoma, head-neck squamous cell carcinoma, adrenocortical carcinoma, liver hepatocellular carcinoma, skin cutaneous melanoma and low-grade glioma. Together, these findings strongly support that cancer cells arising from senescent cells are more dangerous, and that a molecular signature of these cells may be of prognostic value for some human cancers. It also raises questions about modeling human tumors, using cells or mice, without regards to the sequence of events leading to transformation.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Neoplasias Pulmonares , Melanoma , Neoplasias Cutáneas , Animales , Senescencia Celular , Humanos , Ratones , Fenotipo , Proteína p53 Supresora de Tumor , Melanoma Cutáneo Maligno
8.
Aging Cell ; 21(7): e13632, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35653631

RESUMEN

Cellular senescence is characterized by a stable proliferation arrest in response to stresses and the acquisition of a senescence-associated secretory phenotype, called SASP, composed of numerous factors including pro-inflammatory molecules, proteases, and growth factors. The SASP affects the environment of senescent cells, especially during aging, by inducing and modulating various phenotypes such as paracrine senescence, immune cell activity, and extracellular matrix deposition and organization, which critically impact various pathophysiological situations, including fibrosis and cancer. Here, we uncover a novel paracrine effect of the SASP: the neuroendocrine transdifferentiation (NED) of some epithelial cancer cells, evidenced both in the breast and prostate. Mechanistically, this effect is mediated by NF-κB-dependent SASP factors, and leads to an increase in intracellular Ca2+ levels. Consistently, buffering Ca2+ by overexpressing the CALB1 buffering protein partly reverts SASP-induced NED, suggesting that the SASP promotes NED through a SASP-induced Ca2+ signaling. Human breast cancer dataset analyses support that NED occurs mainly in p53 WT tumors and in older patients, in line with a role of senescent cells and its secretome, as they are increasing during aging. In conclusion, our work, uncovering SASP-induced NED in some cancer cells, paves the way for future studies aiming at better understanding the functional link between senescent cell accumulation during aging, NED and clinical patient outcome.


Asunto(s)
Neoplasias de la Mama , Transdiferenciación Celular , FN-kappa B , Anciano , Neoplasias de la Mama/metabolismo , Transdiferenciación Celular/fisiología , Senescencia Celular/genética , Senescencia Celular/fisiología , Humanos , Masculino , FN-kappa B/metabolismo , Células Neuroendocrinas/citología , Células Neuroendocrinas/metabolismo , Secretoma
9.
Cell Death Dis ; 12(2): 190, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594040

RESUMEN

Although aging is a major risk factor for most types of cancers, it is barely studied in this context. The transmembrane protein PLA2R1 (phospholipase A2 receptor) promotes cellular senescence, which can inhibit oncogene-induced tumor initiation. Functions and mechanisms of action of PLA2R1 during aging are largely unknown. In this study, we observed that old Pla2r1 knockout mice were more prone to spontaneously develop a wide spectrum of tumors compared to control littermates. Consistently, these knockout mice displayed increased Parp1, a master regulator of DNA damage repair, and decreased DNA damage, correlating with large human dataset analysis. Forced PLA2R1 expression in normal human cells decreased PARP1 expression, induced DNA damage and subsequent senescence, while the constitutive expression of PARP1 rescued cells from these PLA2R1-induced effects. Mechanistically, PARP1 expression is repressed by a ROS (reactive oxygen species)-Rb-dependent mechanism upon PLA2R1 expression. In conclusion, our results suggest that PLA2R1 suppresses aging-induced tumors by repressing PARP1, via a ROS-Rb signaling axis, and inducing DNA damage and its tumor suppressive responses.


Asunto(s)
Envejecimiento/metabolismo , Daño del ADN , Neoplasias/metabolismo , Neoplasias/prevención & control , Receptores de Fosfolipasa A2/metabolismo , Factores de Edad , Envejecimiento/genética , Envejecimiento/patología , Animales , Línea Celular , Proliferación Celular , Senescencia Celular , Bases de Datos Genéticas , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/genética , Neoplasias/patología , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Fosfolipasa A2/genética , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo
10.
Nat Commun ; 12(1): 720, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33526781

RESUMEN

Cellular senescence is induced by stresses and results in a stable proliferation arrest accompanied by a pro-inflammatory secretome. Senescent cells accumulate during aging, promoting various age-related pathologies and limiting lifespan. The endoplasmic reticulum (ER) inositol 1,4,5-trisphosphate receptor, type 2 (ITPR2) calcium-release channel and calcium fluxes from the ER to the mitochondria are drivers of senescence in human cells. Here we show that Itpr2 knockout (KO) mice display improved aging such as increased lifespan, a better response to metabolic stress, less immunosenescence, as well as less liver steatosis and fibrosis. Cellular senescence, which is known to promote these alterations, is decreased in Itpr2 KO mice and Itpr2 KO embryo-derived cells. Interestingly, ablation of ITPR2 in vivo and in vitro decreases the number of contacts between the mitochondria and the ER and their forced contacts induce premature senescence. These findings shed light on the role of contacts and facilitated exchanges between the ER and the mitochondria through ITPR2 in regulating senescence and aging.


Asunto(s)
Senescencia Celular/fisiología , Retículo Endoplásmico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Longevidad/fisiología , Mitocondrias/metabolismo , Animales , Calcio/metabolismo , Retículo Endoplásmico/ultraestructura , Femenino , Fibroblastos , Células HEK293 , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Mitocondrias/ultraestructura , ARN Interferente Pequeño , Periodo Refractario Electrofisiológico , Análisis de la Célula Individual
11.
Sci Rep ; 10(1): 8190, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424163

RESUMEN

The Phospholipase A2 Receptor 1 (PLA2R1) was first identified for its ability to bind some secreted PLA2s (sPLA2s). It belongs to the C-type lectin superfamily and it binds different types of proteins. It is likely a multifunctional protein that plays a role i) in inflammation and inflammatory diseases, ii) in cellular senescence, a mechanism participating in aging and age-related diseases including cancer, and iii) in membranous nephropathy (MN), a rare autoimmune kidney disease where PLA2R1 is the major autoantigen. To help study the role of PLA2R1 in these pathophysiological conditions, we have generated a versatile NeoR-hPLA2R1 conditional transgenic mice which will allow the specific expression of human PLA2R1 (hPLA2R1) in relevant organs and cells following Cre recombinase-driven excision of the NeoR-stop cassette flanked by LoxP sites. Proof-of-concept breeding of NeoR-hPLA2R1 mice with the ubiquitous adenoviral EIIa promoter-driven Cre mouse line resulted in the expected excision of the NeoR-stop cassette and the expression of hPLA2R1 in all tested tissues. These Tg-hPLA2R1 animals breed normally, with no reproduction or apparent growth defect. These models, especially the NeoR-hPLA2R1 conditional transgenic mouse line, will facilitate the future investigation of PLA2R1 functions in relevant pathophysiological contexts, including inflammatory diseases, age-related diseases and MN.


Asunto(s)
Modelos Animales de Enfermedad , Receptores de Fosfolipasa A2/genética , Animales , Expresión Génica , Técnicas de Genotipaje , Humanos , Ratones , Ratones Transgénicos , Especificidad de Órganos
12.
Cancer Res ; 80(16): 3359-3371, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32554750

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a deadly and aggressive cancer. Understanding mechanisms that drive preneoplastic pancreatic lesions is necessary to improve early diagnostic and therapeutic strategies. Mutations and inactivation of activin-like kinase (ALK4) have been demonstrated to favor PDAC onset. Surprisingly, little is known regarding the ligands that drive ALK4 signaling in pancreatic cancer or how this signaling pathway limits the initiation of neoplastic lesions. In this study, data mining and histologic analyses performed on human and mouse tumor tissues revealed that activin A is the major ALK4 ligand that drives PDAC initiation. Activin A, which is absent in normal acinar cells, was strongly induced during acinar-to-ductal metaplasia (ADM), which was promoted by pancreatitis or the activation of KrasG12D in mice. Activin A expression during ADM was associated with the cellular senescence program that is induced in precursor lesions. Blocking activin A signaling through the use of a soluble form of activin receptor IIB (sActRIIB-Fc) and ALK4 knockout in mice expressing KrasG12D resulted in reduced senescence associated with decreased expression of p21, reduced phosphorylation of H2A histone family member X (H2AX), and increased proliferation. Thus, this study indicates that activin A acts as a protective senescence-associated secretory phenotype factor produced by Kras-induced senescent cells during ADM, which limits the expansion and proliferation of pancreatic neoplastic lesions. SIGNIFICANCE: This study identifies activin A to be a beneficial, senescence-secreted factor induced in pancreatic preneoplastic lesions, which limits their proliferation and ultimately slows progression into pancreatic cancers.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Activinas/biosíntesis , Carcinoma Ductal Pancreático/etiología , Senescencia Celular/fisiología , Neoplasias Pancreáticas/etiología , Lesiones Precancerosas/etiología , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II/metabolismo , Activinas/antagonistas & inhibidores , Animales , Carcinoma Ductal Pancreático/metabolismo , Progresión de la Enfermedad , Genes ras , Humanos , Ratones , Neoplasias Pancreáticas/metabolismo , Fosforilación , Lesiones Precancerosas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Activación Transcripcional
13.
Biochim Biophys Acta Gene Regul Mech ; 1860(4): 427-437, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28215965

RESUMEN

Although Men1 is a well-known tumour suppressor gene, little is known about the functions of Menin, the protein it encodes for. Since few years, numerous publications support a major role of Menin in the control of epigenetics gene regulation. While Menin interaction with MLL complex favours transcriptional activation of target genes through H3K4me3 marks, Menin also represses gene expression via mechanisms involving the Polycomb repressing complex (PRC). Interestingly, Ezh2, the PRC-methyltransferase that catalyses H3K27me3 repressive marks and Menin have been shown to co-occupy a large number of promoters. However, lack of binding between Menin and Ezh2 suggests that another member of the PRC complex is mediating this indirect interaction. Having found that ActivinB - a TGFß superfamily member encoded by the Inhbb gene - is upregulated in insulinoma tumours caused by Men1 invalidation, we hypothesize that Menin could directly participate in the epigenetic-repression of Inhbb gene expression. Using Animal model and cell lines, we report that loss of Menin is directly associated with ActivinB-induced expression both in vivo and in vitro. Our work further reveals that ActivinB expression is mediated through a direct modulation of H3K27me3 marks on the Inhbb locus in Menin-KO cell lines. More importantly, we show that Menin binds on the promoter of Inhbb gene where it favours the recruitment of Ezh2 via an indirect mechanism involving Akt-phosphorylation. Our data suggests therefore that Menin could take an important part to the Ezh2-epigenetic repressive landscape in many cells and tissues through its capacity to modulate Akt phosphorylation.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Subunidades beta de Inhibinas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Línea Celular Tumoral , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Sitios Genéticos , Subunidades beta de Inhibinas/metabolismo , Lisina , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Transducción de Señal
14.
Mol Cell Biol ; 36(5): 756-64, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26711255

RESUMEN

Loss of pancreatic ß-cell maturity occurs in diabetes and insulinomas. Although both physiological and pathological stresses are known to promote ß-cell dedifferentiation, little is known about the molecules involved in this process. Here we demonstrate that activinB, a transforming growth factor ß (TGF-ß)-related ligand, is upregulated during tumorigenesis and drives the loss of insulin expression and ß-cell maturity in a mouse insulinoma model. Our data further identify Pax4 as a previously unknown activinB target and potent contributor to the observed ß-cell dedifferentiation. More importantly, using compound mutant mice, we found that deleting activinB expression abolishes tumor ß-cell dedifferentiation and, surprisingly, increases survival without significantly affecting tumor growth. Hence, this work reveals an unexpected role for activinB in the loss of ß-cell maturity, islet plasticity, and progression of insulinoma through its participation in ß-cell dedifferentiation.


Asunto(s)
Activinas/metabolismo , Desdiferenciación Celular , Células Secretoras de Insulina/patología , Insulinoma/patología , Páncreas/patología , Neoplasias Pancreáticas/patología , Activinas/genética , Animales , Regulación Neoplásica de la Expresión Génica , Insulina/genética , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Insulinoma/genética , Insulinoma/metabolismo , Ratones Endogámicos C57BL , Páncreas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
15.
Diabetes ; 64(12): 4212-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26470788

RESUMEN

ßig-h3/TGF-ßi is a secreted protein capable of binding to both extracellular matrix and cells. Human genetic studies recently revealed that in the tgfbi gene encoding for ßig-h3, three single nucleotide polymorphisms were significantly associated with type 1 diabetes (T1D) risk. Pancreatic islets express ßig-h3 in physiological conditions, but this expression is reduced in ß-cell insult in T1D. Since the integrity of islets is destroyed by autoimmune T lymphocytes, we thought to investigate the impact of ßig-h3 on T-cell activation. We show here that ßig-h3 inhibits T-cell activation markers as well as cytotoxic molecule production as granzyme B and IFN-γ. Furthermore, ßig-h3 inhibits early T-cell receptor signaling by repressing the activation of the early kinase protein Lck. Moreover, ßig-h3-treated T cells are unable to induce T1D upon transfer in Rag2 knockout mice. Our study demonstrates for the first time that T-cell activation is modulated by ßig-h3, an islet extracellular protein, in order to efficiently avoid autoimmune response.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Proteínas de la Matriz Extracelular/farmacología , Hipoglucemiantes/farmacología , Activación de Linfocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Biomarcadores/metabolismo , Cadáver , Células Cultivadas , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Humanos , Hipoglucemiantes/metabolismo , Ganglios Linfáticos/patología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/antagonistas & inhibidores , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Organismos Libres de Patógenos Específicos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
17.
J Bone Miner Res ; 29(8): 1886-99, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24619707

RESUMEN

The transcription factor TWIST1 induces epithelial-mesenchymal transition and/or escape to the oncogenic-induced failsafe program, facilitating the intravasation of breast cancer cells in the systemic circulation and their dissemination to the lungs. Its involvement in breast cancer bone metastasis is unknown. To address this question, human osteotropic MDA-MB-231/B02 breast cancer cells were stably transfected with a Tet-inducible vector encoding for TWIST1, whose expression was specifically repressed in the presence of doxycycline (dox). The intra-arterial inoculation of transfectants expressing TWIST1 in immunodeficient mice substantially increased the extent of osteolytic lesions in these animals, being 50% larger than that of animals bearing mock-transfected tumors, as determined by radiography. This difference was accompanied by a sharp reduction of the bone volume (indicating a higher bone destruction) and a twofold increase in the tumor volume compared with mice bearing mock-transfected tumors, as determined by histomorphometry. Importantly, the suppression of TWIST1 expression in MDA-MB-231/B02 cells in the presence of dox abolished the stimulatory effect of TWIST1 on bone metastasis formation in vivo. Additionally, examination of the bone marrow from untreated and dox-treated animals on day 7 after tumor cell inoculation, at which time there was no evidence of radiographic osteolytic lesions, revealed that the number of tumor cell colonies that were recovered from the bone marrow of untreated mice was dramatically increased compared with that of dox-fed animals. In vitro, TWIST1 expression promoted tumor cell invasion and enhanced microRNA 10b (miR-10b) expression, a proinvasive factor, but was dispensable for growth of tumor cells. In vivo, the repression of miR-10b substantially decreased the presence of TWIST1-expressing breast cancer cells in the bone marrow. Overall, these results establish that TWIST1 facilitates breast cancer bone metastasis formation through a mechanism dependent of miR-10b, which leads to increase tumor burden and bone destruction.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética , Animales , Western Blotting , Diferenciación Celular , Línea Celular Tumoral , Doxiciclina/farmacología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Osteoclastos/citología , Proteína 1 Relacionada con Twist/metabolismo
18.
Cancer Res ; 71(17): 5728-38, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21734015

RESUMEN

Bone metastasis is a complication occurring in up to 70% of advanced breast cancer patients. The estrogen receptor-related receptor alpha (ERRα) has been implicated in breast cancer and bone development, prompting us to examine whether ERRα may function in promoting the osteolytic growth of breast cancer cells in bone. In a mouse xenograft model of metastatic human breast cancer, overexpression of wild-type ERRα reduced metastasis, whereas overexpression of a dominant negative mutant promoted metastasis. Osteoclasts were directly affected and ERRα upregulated the osteoclastogenesis inhibitor, osteoprotegerin (OPG), providing a direct mechanistic basis for understanding how ERRα reduced breast cancer cell growth in bone. In contrast, ERRα overexpression increased breast cancer cell growth in the mammary gland. ERRα-overexpressing primary tumors were highly vascularized, consistent with an observed upregulation of angiogenic growth factor, the VEGF. In support of these findings, we documented that elevated expression of ERRα mRNA in breast carcinomas was associated with high expression of OPG and VEGF and with disease progression. In conclusion, our results show that ERRα plays a dual role in breast cancer progression in promoting the local growth of tumor cells, but decreasing metastatic growth of osteolytic lesions in bone.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Carcinoma/secundario , Osteoprotegerina/metabolismo , Receptores de Estrógenos/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/mortalidad , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Carcinoma/irrigación sanguínea , Carcinoma/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Receptores de Estrógenos/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Receptor Relacionado con Estrógeno ERRalfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA