Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37445973

RESUMEN

Cellular senescence is a complex cell state that can occur during physiological ageing or after exposure to stress signals, regardless of age. It is a dynamic process that continuously evolves in a context-dependent manner. Senescent cells interact with their microenvironment by producing a heterogenous and plastic secretome referred to as the senescence-associated secretory phenotype (SASP). Hence, understanding the cross-talk between SASP and the microenvironment can be challenging due to the complexity of signal exchanges. In this review, we first aim to update the definition of senescence and its associated biomarkers from its discovery to the present day. We detail the regulatory mechanisms involved in the expression of SASP at multiple levels and develop how SASP can orchestrate microenvironment modifications, by focusing on extracellular matrix modifications, neighboring cells' fate, and intercellular communications. We present hypotheses on how these microenvironmental events may affect dynamic changes in SASP composition in return. Finally, we discuss the various existing approaches to targeting SASP and clarify what is currently known about the biological effects of these modified SASPs on the cellular environment.


Asunto(s)
Comunicación Celular , Senescencia Celular , Senescencia Celular/genética , Biomarcadores , Fenotipo
2.
Elife ; 112022 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-35302491

RESUMEN

A rare but severe complication of curative-intent radiation therapy is the induction of second primary cancers. These cancers preferentially develop not inside the planning target volume (PTV) but around, over several centimeters, after a latency period of 1-40 years. We show here that normal human or mouse dermal fibroblasts submitted to the out-of-field dose scattering at the margin of a PTV receiving a mimicked patient's treatment do not die but enter in a long-lived senescent state resulting from the accumulation of unrepaired DNA single-strand breaks, in the almost absence of double-strand breaks. Importantly, a few of these senescent cells systematically and spontaneously escape from the cell cycle arrest after a while to generate daughter cells harboring mutations and invasive capacities. These findings highlight single-strand break-induced senescence as the mechanism of second primary cancer initiation, with clinically relevant spatiotemporal specificities. Senescence being pharmacologically targetable, they open the avenue for second primary cancer prevention.


Asunto(s)
Reparación del ADN , Neoplasias Primarias Secundarias , Animales , Carcinogénesis , Transformación Celular Neoplásica , Senescencia Celular , Roturas del ADN de Cadena Simple , Daño del ADN , Ratones
3.
J Biol Chem ; 295(50): 17310-17322, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33037071

RESUMEN

In addition to their well-known role in the control of cellular proliferation and cancer, cell cycle regulators are increasingly identified as important metabolic modulators. Several GWAS have identified SNPs near CDKN2A, the locus encoding for p16INK4a (p16), associated with elevated risk for cardiovascular diseases and type-2 diabetes development, two pathologies associated with impaired hepatic lipid metabolism. Although p16 was recently shown to control hepatic glucose homeostasis, it is unknown whether p16 also controls hepatic lipid metabolism. Using a combination of in vivo and in vitro approaches, we found that p16 modulates fasting-induced hepatic fatty acid oxidation (FAO) and lipid droplet accumulation. In primary hepatocytes, p16-deficiency was associated with elevated expression of genes involved in fatty acid catabolism. These transcriptional changes led to increased FAO and were associated with enhanced activation of PPARα through a mechanism requiring the catalytic AMPKα2 subunit and SIRT1, two known activators of PPARα. By contrast, p16 overexpression was associated with triglyceride accumulation and increased lipid droplet numbers in vitro, and decreased ketogenesis and hepatic mitochondrial activity in vivo Finally, gene expression analysis of liver samples from obese patients revealed a negative correlation between CDKN2A expression and PPARA and its target genes. Our findings demonstrate that p16 represses hepatic lipid catabolism during fasting and may thus participate in the preservation of metabolic flexibility.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Ácidos Grasos/metabolismo , Hígado/metabolismo , Mitocondrias Hepáticas/metabolismo , PPAR alfa/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Ácidos Grasos/genética , Estudio de Asociación del Genoma Completo , Humanos , Gotas Lipídicas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Obesidad/genética , Obesidad/metabolismo , Oxidación-Reducción , PPAR alfa/genética , Sirtuina 1/genética
4.
Int J Oncol ; 54(1): 29-40, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30365153

RESUMEN

Ets-1 transcription factor overexpression in breast cancers is associated with invasive features and is associated with a poor prognosis. Beyond its role in driving carcinoma cell invasion, in this study, we wished to determine whether Ets-1 overexpression in cancer cells promotes angiogenesis by creating a paracrine pro-invasive environment for endothelial cells as well. To address this question, we set up different co-culture models of cancer cells with endothelial cells. Conditioned media from cancer cells induced endothelial cell proliferation, migration and morphogenesis in matrix models. Of note, co-culture assays in three-dimensional matrix models also revealed the reciprocal induction of cancer cell morphogenesis by endothelial cells, in support of an angiocrine action on tumor cells. Ets-1 emerged as a key regulator of the angiogenic potential of breast cancer cells, favoring their ability to induce, in a paracrine manner, the morphogenesis of endothelial cells and also to physically interact with the latter. Nevertheless, Ets-1 overexpression in cancer cells also restrained their chemoattractive potential for endothelial cells both in Boyden chambers and in ex vivo 3D co-cultures. Finally, Ets-1 modulation in breast cancer cells qualitatively altered the angiogenic pattern of experimental in vivo tumors, with a balance between vessel recruitment and intratumoral small capillaries sprouting. Taken together, our data highlight a critical and intriguing role for Ets-1 in the angiogenic potential of breast cancer cells, and reveal another facet of Ets-1 oncogenic activities.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Células Endoteliales/citología , Neovascularización Patológica/genética , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/química , Células Endoteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Neovascularización Patológica/metabolismo , Comunicación Paracrina , Regulación hacia Arriba
5.
Cancer Lett ; 438: 187-196, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30213560

RESUMEN

The incidence of carcinomas highly increases with age. However, the initial steps of the age-related molecular carcinogenic processes remain poorly characterized. We previously showed that normal human epidermal keratinocytes spontaneously and systematically escape from senescence to give rise to preneoplastic emerging cells through a process called post-senescence neoplastic emergence (PSNE). To identify molecular pathways involved in the switch from senescence to pre-transformation, we performed Connectivity Map analyses and DAVID functional annotations followed by hierarchical clustering and multidimensional scaling of the gene expression signature of PSNE cells. We identified endoplasmic reticulum stress related pathways as key regulators of PSNE. Invalidation by RNA interference of the UPR sensors PERK, ATF6α, but not IRE1α, delayed the occurrence of senescence when performed in pre-senescent cells, and increased the PSNE frequency when performed in already senescent cells. Conversely, endoplasmic reticulum stress inducers applied to already senescent cells decreased the frequency of PSNE. In conclusion, these results indicate that the activation of the UPR could protect from the early carcinogenic steps by senescence evasion. This opens new avenues to explore therapeutics that could be useful in decreasing the age-associated tumor incidence.


Asunto(s)
Factor de Transcripción Activador 6/genética , Transformación Celular Neoplásica/genética , Transcriptoma , Respuesta de Proteína Desplegada/genética , eIF-2 Quinasa/genética , Factor de Transcripción Activador 6/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Línea Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/clasificación , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Respuesta de Proteína Desplegada/efectos de los fármacos , eIF-2 Quinasa/metabolismo
6.
Mech Ageing Dev ; 170: 82-91, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28803844

RESUMEN

Senescence is recognized as a cellular state acquired in response to various stresses. It occurs in correlation with the activation of the Unfolded Protein Response (UPR) pathway. However, the UPR targets which might relay the establishment of the senescent phenotype are not known. Herein, we investigated whether the up-regulation of the COX2 (PTGS2) limiting enzyme in the prostaglandin biosynthesis pathway, known to mediate cellular senescence in normal human fibroblasts, could be controlled by the UPR sensors ATF6α, IRE1α and PERK. We found that UPR inducers cause premature senescence through an increase in COX2 expression, and an overproduction of prostaglandin E2 (PGE2) in wild type fibroblasts but not in ATF6α invalidated ones. In replicative senescent fibroblasts, ATF6α and IRE1α silencing abrogated COX2 up-regulation and PGE2 production. The expanded ER and the large cell size characteristics of senescent fibroblasts were both reduced upon the invalidation of COX2 as well as ATF6α. These effects of the ATF6α invalidation were prevented by favoring the import of PGE2, but not just by supplying extracellular PGE2. Taken together, our results support a critical role of ATF6α in the establishment and maintenance of cellular senescence in normal human fibroblasts via the up-regulation of a COX2/PGE2 intracrine pathway.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Senescencia Celular , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Fibroblastos/metabolismo , Transducción de Señal , Respuesta de Proteína Desplegada , Ciclooxigenasa 2/genética , Dinoprostona/genética , Fibroblastos/patología , Humanos
7.
Cell Mol Life Sci ; 74(24): 4471-4509, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28707011

RESUMEN

Senescence is a cell state occurring in vitro and in vivo after successive replication cycles and/or upon exposition to various stressors. It is characterized by a strong cell cycle arrest associated with several molecular, metabolic and morphologic changes. The accumulation of senescent cells in tissues and organs with time plays a role in organismal aging and in several age-associated disorders and pathologies. Moreover, several therapeutic interventions are able to prematurely induce senescence. It is, therefore, tremendously important to characterize in-depth, the mechanisms by which senescence is induced, as well as the precise properties of senescent cells. For historical reasons, senescence is often studied with fibroblast models. Other cell types, however, much more relevant regarding the structure and function of vital organs and/or regarding pathologies, are regrettably often neglected. In this article, we will clarify what is known on senescence of epithelial cells and highlight what distinguishes it from, and what makes it like, replicative senescence of fibroblasts taken as a standard.


Asunto(s)
Adaptación Biológica/fisiología , Carcinogénesis/patología , Senescencia Celular/fisiología , Células Epiteliales/patología , Estrés Fisiológico/fisiología , Animales , Fibroblastos/patología , Humanos
8.
Oncotarget ; 7(42): 67699-67715, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27563820

RESUMEN

Cellular senescence is known as an anti-tumor barrier and is characterized by a number of determinants including cell cycle arrest, senescence associated ß-galactosidase activity and secretion of pro-inflammatory mediators. Senescent cells are also subjected to enlargement, cytoskeleton-mediated shape changes and organelle alterations. However, the underlying molecular mechanisms responsible for these last changes remain still uncharacterized. Herein, we have identified the Unfolded Protein Response (UPR) as a player controlling some morphological aspects of the senescent phenotype. We show that senescent fibroblasts exhibit ER expansion and mild UPR activation, but conserve an ER stress adaptive capacity similar to that of exponentially growing cells. By genetically invalidating the three UPR sensors in senescent fibroblasts, we demonstrated that ATF6α signaling dictates senescence-associated cell shape modifications. We also show that ER expansion and increased secretion of the pro-inflammatory mediator IL6 were partly reversed by silencing ATF6α in senescent cells. Moreover, ATF6α drives the increase of senescence associated-ß-galactosidase activity. Collectively, these findings unveil a novel and central role for ATF6α in the establishment of morphological features of senescence in normal human primary fibroblasts.


Asunto(s)
Factor de Transcripción Activador 6/genética , Senescencia Celular/genética , Fibroblastos/metabolismo , Respuesta de Proteína Desplegada/genética , Factor de Transcripción Activador 6/metabolismo , Adulto , Células Cultivadas , Niño , Dermis/citología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Estrés del Retículo Endoplásmico/genética , Femenino , Fibroblastos/citología , Perfilación de la Expresión Génica/métodos , Humanos , Lactante , Masculino , Microscopía Electrónica de Transmisión , Interferencia de ARN , Transducción de Señal/genética
9.
Nat Commun ; 7: 10399, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26822533

RESUMEN

The main characteristic of senescence is its stability which relies on the persistence of DNA damage. We show that unlike fibroblasts, senescent epithelial cells do not activate an ATM-or ATR-dependent DNA damage response (DDR), but accumulate oxidative-stress-induced DNA single-strand breaks (SSBs). These breaks remain unrepaired because of a decrease in PARP1 expression and activity. This leads to the formation of abnormally large and persistent XRCC1 foci that engage a signalling cascade involving the p38MAPK and leading to p16 upregulation and cell cycle arrest. Importantly, the default in SSB repair also leads to the emergence of post-senescent transformed and mutated precancerous cells. In human-aged skin, XRCC1 foci accumulate in the epidermal cells in correlation with a decline of PARP1, whereas DDR foci accumulate mainly in dermal fibroblasts. These findings point SSBs as a DNA damage encountered by epithelial cells with aging which could fuel the very first steps of carcinogenesis.


Asunto(s)
Senescencia Celular , Roturas del ADN de Cadena Simple , Células Epiteliales/citología , Neoplasias/genética , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/fisiopatología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
10.
Am J Physiol Cell Physiol ; 308(6): C415-25, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25540175

RESUMEN

The endoplasmic reticulum (ER) is a multifunctional organelle critical for the proper folding and assembly of secreted and transmembrane proteins. Perturbations of ER functions cause ER stress, which activates a coordinated system of transcriptional and translational controls called the unfolded protein response (UPR), to cope with accumulation of misfolded proteins and proteotoxicity. It results in ER homeostasis restoration or in cell death. Senescence is a complex cell phenotype induced by several stresses such as telomere attrition, DNA damage, oxidative stress, and activation of some oncogenes. It is mainly characterized by a cell enlargement, a permanent cell-cycle arrest, and the production of a secretome enriched in proinflammatory cytokines and components of the extracellular matrix. Senescent cells accumulate with age in tissues and are suspected to play a role in age-associated diseases. Since senescence is a stress response, the question arises of whether an ER stress could occur concomitantly with senescence and participate in the onset or maintenance of the senescent features. Here, we described the interconnections between the UPR signaling and the different aspects of the cellular senescence programs and discuss the implication of UPR modulations in this context.


Asunto(s)
Senescencia Celular , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Transducción de Señal , Respuesta de Proteína Desplegada , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Retículo Endoplásmico/patología , Humanos , Fenotipo
11.
Int J Cancer ; 135(10): 2317-28, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706481

RESUMEN

Ets-1 overexpression in human breast cancers is associated with invasiveness and poor prognosis. By overexpressing Ets-1 or a dominant negative mutant in MMT breast cancer cells, we previously highlighted the key role of Ets-1 in coordinating multiple invasive features of these cells. Interestingly, we also noticed that Ets-1 decreased the density of breast cancer cells cultured in three-dimensional extracellular matrix gels. The 3D context was instrumental to this phenomenon, as such downregulation was not observed in cells grown on two-dimensional plastic or matrix-coated dishes. Ets-1 overexpression was deleterious to anchorage-independent growth of MMT cells in soft agar, a standard model for in vitro tumorigenicity. The relevance of this mechanism was confirmed in vivo, during primary tumor growth and in a metastatic assay of lung colonization. In these models, Ets-1 was associated with epithelial-to-mesenchymal transition features and modulated the ratio of Ki67-positive cells, while hardly affecting in vivo apoptotic cell death. Finally, siRNA-mediated knockdown of Ets-1 in human breast cancer cell lines also decreased colony growth, both in anchorage-independent assays and 3D extracellular matrix cultures. These in vitro and in vivo observations shed light on an unsuspected facet of Ets-1 in breast tumorigenesis. They show that while promoting malignancy through the acquisition of invasive features, Ets-1 also attenuates breast tumor cell growth and could therefore repress the growth of primary tumors and metastases. This work also demonstrates that 3D models may reveal mechanisms of tumor biology that are cryptic in standard 2D models.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Neoplasias Pulmonares/secundario , Proteína Proto-Oncogénica c-ets-1/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/metabolismo , Técnicas de Cultivo de Célula , Ciclo Celular , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteína Proto-Oncogénica c-ets-1/antagonistas & inhibidores , Proteína Proto-Oncogénica c-ets-1/genética , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Biochim Biophys Acta ; 1831(7): 1217-27, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24046862

RESUMEN

Cyclooxygenase 2 and release of prostaglandin E2 are involved in many responses including inflammation and are upregulated during cellular senescence. However, little is known about the role of lipid inflammatory mediators in senescence. Here, we investigated the mechanism by which the COX-2/PGE2 axis induces senescence. Using the NS398 specific inhibitor of COX-2, we provide evidence that reactive oxygen species by-produced by the COX-2 enzymatic activity are negligible in front of the total senescence-associated oxidative stress. We therefore investigated the role of PGE2 by invalidating the PGE2 synthases downstream of COX-2, or the specific PGE2 receptors, or by applying PGE2 or specific agonists or antagonists. We evaluated the effect on senescence by evaluating the senescence-associated proliferation arrest, the percentage of senescence-associated beta-galactosidase-positive cells, and the expression of senescent molecular markers such as IL-6 and MCP1. We show that PGE2 acting on its EP specific receptors is able to induce both the onset of senescence and the maintenance of the phenotype. It did so only when the PGE2/lactate transporter activity was enhanced, indicating that PGE2 acts on senescence more via the pool of intracellular EP receptors than via those localized at the cell surface. Treatment with agonists, antagonists and silencing of the EP receptors by siRNA revealed that EP3 was the most involved in transducing the intracrine effects of PGE2. Immunofluorescence experiments confirmed that EP3 was more localized in the cytoplasm than at the cell surface. Taken together, these results suggest that COX-2 contributes to the establishment and maintenance of senescence of normal human fibroblasts via an independent-ROS and a dependent-PGE2/EPs intracrine pathway.


Asunto(s)
Senescencia Celular , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Fibroblastos/citología , Transducción de Señal , Línea Celular , Dermis/citología , Fibroblastos/metabolismo , Humanos , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo
13.
PLoS One ; 8(5): e63607, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23675494

RESUMEN

The incidence of carcinoma increases greatly with aging, but the cellular and molecular mechanisms underlying this correlation are only partly known. It is established that senescent fibroblasts promote the malignant progression of already-transformed cells through secretion of inflammatory mediators. We investigated here whether the senescent fibroblast secretome might have an impact on the very first stages of carcinogenesis. We chose the cultured normal primary human epidermal keratinocyte model, because after these cells reach the senescence plateau, cells with transformed and tumorigenic properties systematically and spontaneously emerge from the plateau. In the presence of medium conditioned by autologous senescent dermal fibroblasts, a higher frequency of post-senescence emergence was observed and the post-senescence emergent cells showed enhanced migratory properties and a more marked epithelial-mesenchymal transition. Using pharmacological inhibitors, siRNAs, and blocking antibodies, we demonstrated that the MMP-1 and MMP-2 matrix metalloproteinases, known to participate in late stages of cancer invasion and metastasis, are responsible for this enhancement of early migratory capacity. We present evidence that MMPs act by activating the protease-activated receptor 1 (PAR-1), whose expression is specifically increased in post-senescence emergent keratinocytes. The physiopathological relevance of these results was tested by analyzing MMP activity and PAR-1 expression in skin sections. Both were higher in skin sections from aged subjects than in ones from young subjects. Altogether, our results suggest that during aging, the dermal and epidermal skin compartments might be activated coordinately for initiation of skin carcinoma, via a paracrine axis in which MMPs secreted by senescent fibroblasts promote very early epithelial-mesenchymal transition of keratinocytes undergoing transformation and oversynthesizing the MMP-activatable receptor PAR-1.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Senescencia Celular , Fibroblastos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Comunicación Paracrina , Receptor PAR-1/metabolismo , Piel/metabolismo , Adulto , Movimiento Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Células Cultivadas , Transición Epitelial-Mesenquimal/genética , Femenino , Expresión Génica , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Queratinocitos/metabolismo , Metaloproteinasa 1 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasas de la Matriz/genética , Receptor PAR-1/genética , Piel/patología , Factor de Crecimiento Transformador beta1/farmacología , Adulto Joven
14.
Traffic ; 13(9): 1261-72, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22672335

RESUMEN

The receptor tyrosine kinase Met and its ligand, the hepatocyte growth factor/scatter factor, are essential for embryonic development, whereas deregulation of Met signaling pathways is associated with tumorigenesis and metastasis. The presenilin-regulated intramembrane proteolysis (PS-RIP) is involved in ligand-independent downregulation of Met. This proteolytic process involves shedding of the Met extracellular domain followed by γ-secretase cleavage, generating labile intracellular fragments degraded by the proteasome. We demonstrate here that upon shedding both generated Met N- and C-terminal fragments are degraded directly in the lysosome, with C-terminal fragments escaping γ-secretase cleavage. PS-RIP and lysosomal degradation are complementary, because their simultaneous inhibition induces synergistic accumulation of fragments. Met N-terminal fragments associate with the high-affinity domain of HGF/SF, confirming its decoy activity which could be reduced through their routing to the lysosome at the expense of extracellular release. Finally, the DN30 monoclonal antibody inducing Met shedding promotes receptor degradation through induction of both PS-RIP and the lysosomal pathway. Thus, we demonstrate that Met shedding initiates a novel lysosomal degradation which participates to ligand-independent downregulation of the receptor.


Asunto(s)
Lisosomas/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Línea Celular Tumoral , Perros , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Ratones , Células 3T3 NIH , Presenilinas/metabolismo , Inhibidores de Proteasas/farmacología , Proteolisis , Proteínas Proto-Oncogénicas c-met/genética , ARN Interferente Pequeño
15.
J Biol Chem ; 287(8): 5379-89, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22194601

RESUMEN

The transcriptional repressor HIC1 (Hypermethylated in Cancer 1) is a tumor suppressor gene inactivated in many human cancers including breast carcinomas. In this study, we show that HIC1 is a direct transcriptional repressor of ß-2 adrenergic receptor (ADRB2). Through promoter luciferase activity, chromatin immunoprecipitation (ChIP) and sequential ChIP experiments, we demonstrate that ADRB2 is a direct target gene of HIC1, endogenously in WI-38 cells and following HIC1 re-expression in breast cancer cells. Agonist-mediated stimulation of ADRB2 increases the migration and invasion of highly malignant MDA-MB-231 breast cancer cells but these effects are abolished following HIC1 re-expression or specific down-regulation of ADRB2 by siRNA treatment. Our results suggest that early inactivation of HIC1 in breast carcinomas could predispose to stress-induced metastasis through up-regulation of the ß-2 adrenergic receptor.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores Adrenérgicos beta 2/genética , Estrés Fisiológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores Adrenérgicos beta 2/deficiencia , Estrés Fisiológico/genética
16.
Int J Oncol ; 39(5): 1073-82, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21833469

RESUMEN

The transcription factor Ets-1 is known to be involved in a broad variety of cellular functions such as cell proliferation, migration, invasion, apoptosis and angiogenesis. In nearly all these reports, the full-length Ets-1 (p51) is commonly considered to be the active form and the role of the Ets-1ΔVII splice variant (p42) has not been addressed. Therefore, we studied the functional effects of p42 Ets-1 in comparison to p51 Ets-1 expression in a well-characterized mouse fibroblast cell line. Furthermore, the specific role of Ets-1 was evaluated using mouse fibroblasts with a reduced Ets-1 expression caused by RNAi and compared to fibroblasts with a binding inhibition of the whole ETS transcription factor family by stably overexpressing the ETS DNA binding domain as transdominant-negative mutant. Our results demonstrate that p42 Ets-1 has quite different functions and target genes compared to p51 Ets-1 (e.g. TIMP-4, MMP-3, MMP-9, MMP-13). In some cases (e.g. in cytokine expression) p42 Ets-1 is a functional transcription factor which acts in the same manner as a transdominant-negative approach.


Asunto(s)
Empalme Alternativo/genética , Fibroblastos/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Animales , Movimiento Celular/genética , Proliferación Celular , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Espacio Intracelular/metabolismo , Ratones , Células 3T3 NIH , Transporte de Proteínas , Transcripción Genética
17.
PLoS One ; 5(9): e12712, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20856861

RESUMEN

Senescence is a state of growth arrest resulting mainly from telomere attrition and oxidative stress. It ultimately leads to cell death. We have previously shown that, in keratinocytes, senescence is induced by NF-kappaB activation, MnSOD upregulation and H(2)O(2) overproduction. We have also shown that senescent keratinocytes do not die by apoptosis but as a result of high macroautophagic activity that targets the primary vital cell components. Here, we investigated the mechanisms that activate this autophagic cell death program. We show that corpses occurring at the senescence plateau display oxidatively-damaged mitochondria and nucleus that colocalize with autophagic vacuoles. The occurrence of such corpses was decreased by specifically reducing the H(2)O(2) level with catalase, and, conversely, reproduced by overexpressing MnSOD or applying subtoxic doses of H(2)O(2). This H(2)O(2)-induced cell death did occur through autophagy since it was accompanied by an accumulation of autophagic vesicles as evidenced by Lysotracker staining, LC3 vesiculation and transmission electron microscopy. Most importantly, it was partly abolished by 3-methyladenine, the specific inhibitor of autophagosome formation, and by anti-Atg5 siRNAs. Taken together these results suggest that autophagic cell death is activated in senescent keratinocytes because of the upregulation of MnSOD and the resulting accumulation of oxidative damages to nucleus and mitochondria.


Asunto(s)
Autofagia , Queratinocitos/citología , Queratinocitos/enzimología , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba , Muerte Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Senescencia Celular , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Queratinocitos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Estrés Oxidativo , Superóxido Dismutasa/genética
18.
Cancer Res ; 70(3): 1225-35, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20103638

RESUMEN

Castration resistance in prostate cancer (PCa) constitutes an advanced, aggressive disease with poor prognosis, associated with uncontrolled cell proliferation, resistance to apoptosis, and enhanced invasive potential. The molecular mechanisms involved in the transition of PCa to castration resistance are obscure. Here, we report that the nonselective cationic channel transient receptor potential vanilloid 2 (TRPV2) is a distinctive feature of castration-resistant PCa. TRPV2 transcript levels were higher in patients with metastatic cancer (stage M1) compared with primary solid tumors (stages T2a and T2b). Previous studies of the TRPV2 channel indicated that it is primarily involved in cancer cell migration and not in cell growth. Introducing TRPV2 into androgen-dependent LNCaP cells enhanced cell migration along with expression of invasion markers matrix metalloproteinase (MMP) 9 and cathepsin B. Consistent with the likelihood that TRPV2 may affect cancer cell aggressiveness by influencing basal intracellular calcium levels, small interfering RNA-mediated silencing of TRPV2 reduced the growth and invasive properties of PC3 prostate tumors established in nude mice xenografts, and diminished expression of invasive enzymes MMP2, MMP9, and cathepsin B. Our findings establish a role for TRPV2 in PCa progression to the aggressive castration-resistant stage, prompting evaluation of TRPV2 as a potential prognostic marker and therapeutic target in the setting of advanced PCa.


Asunto(s)
Neoplasias de la Próstata/genética , Interferencia de ARN , Canales Catiónicos TRPV/genética , Andrógenos/metabolismo , Andrógenos/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Microscopía Confocal , Invasividad Neoplásica , Metástasis de la Neoplasia , Orquiectomía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Res ; 69(20): 7917-25, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19826058

RESUMEN

Studies on human fibroblasts have led to viewing senescence as a barrier against tumorigenesis. Using keratinocytes, we show here that partially transformed and tumorigenic cells systematically and spontaneously emerge from senescent cultures. We show that these emerging cells are generated from senescent cells, which are still competent for replication, by an unusual budding-mitosis mechanism. We further present data implicating reactive oxygen species that accumulate during senescence as a potential mutagenic motor of this post-senescence emergence. We conclude that senescence and its associated oxidative stress could be a tumor-promoting state for epithelial cells, potentially explaining why the incidence of carcinogenesis dramatically increases with advanced age.


Asunto(s)
Transformación Celular Neoplásica , Senescencia Celular , Daño del ADN , Neoplasias/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Adenoviridae , Adolescente , Adulto , Elementos Alu , Western Blotting , Proliferación Celular , Células Cultivadas , Ensayo Cometa , Sondas de ADN , Epidermis/metabolismo , Epidermis/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Técnica del Anticuerpo Fluorescente , Humanos , Hibridación in Situ , Cariotipificación , Queratinocitos/metabolismo , Queratinocitos/patología , Persona de Mediana Edad , Neoplasias/metabolismo , Superóxido Dismutasa/metabolismo , Adulto Joven
20.
Biochim Biophys Acta ; 1793(3): 528-39, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19321128

RESUMEN

The physiological role, the mechanisms of activation, as well as the endogenous regulators for the non-selective cationic channel TRPV2 are not known so far. In the present work we report that endogenous lysophospholipids such as lysophosphatidylcholine (LPC) and lysophosphatidylinositol (LPI) induce a calcium influx via TRPV2 channel. This activation is dependent on the length of the side-chain and the nature of the lysophospholipid head-group. TRPV2-mediated calcium uptake stimulated by LPC and LPI occurred via Gq/Go-protein and phosphatidylinositol-3,4 kinase (PI3,4K) signalling. We have shown that the mechanism of TRPV2 activation induced by LPC and LPI is due to the TRPV2 channel translocation to the plasma membrane. The activation of TRPV2 channel by LPC and LPI leads to an increase in the cell migration of the prostate cancer cell line PC3. We have demonstrated that TRPV2 is directly involved in both steady-state and lysophospholipid-stimulated cancer cell migration. Thus, for the first time, we have identified one of the natural regulators of TRPV2 channel, one of the mechanisms of TRPV2 activation and regulation, as well as its pathophysiological role in cancer.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Lisofosfolípidos/farmacología , Neoplasias de la Próstata/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Células CHO , Calcio/metabolismo , Canales de Calcio/metabolismo , Línea Celular , Cricetinae , Cricetulus , Proteínas de Unión al GTP/metabolismo , Humanos , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...