Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Nat Commun ; 15(1): 7173, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39169003

RESUMEN

Plasma growth differentiation factor-15 (GDF-15) levels increase with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) but the underlying mechanism remains poorly defined. Using male mouse models of obesity and MASLD, and biopsies from carefully-characterized patients regarding obesity, type 2 diabetes (T2D) and MASLD status, we identify adipose tissue (AT) as the key source of GDF-15 at onset of obesity and T2D, followed by liver during the progression towards metabolic dysfunction-associated steatohepatitis (MASH). Obesity and T2D increase GDF15 expression in AT through the accumulation of macrophages, which are the main immune cells expressing GDF15. Inactivation of Gdf15 in macrophages reduces plasma GDF-15 concentrations and exacerbates obesity in mice. During MASH development, Gdf15 expression additionally increases in hepatocytes through stress-induced TFEB and DDIT3 signaling. Together, these results demonstrate a dual contribution of AT and liver to GDF-15 production in metabolic diseases and identify potential therapeutic targets to raise endogenous GDF-15 levels.


Asunto(s)
Tejido Adiposo , Diabetes Mellitus Tipo 2 , Hígado Graso , Factor 15 de Diferenciación de Crecimiento , Hepatocitos , Macrófagos , Obesidad , Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Animales , Obesidad/metabolismo , Obesidad/patología , Hepatocitos/metabolismo , Masculino , Macrófagos/metabolismo , Ratones , Humanos , Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Hígado Graso/metabolismo , Hígado Graso/patología , Ratones Endogámicos C57BL , Hígado/metabolismo , Hígado/patología , Modelos Animales de Enfermedad , Transducción de Señal
2.
Nucleic Acids Res ; 50(22): 12768-12789, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36477312

RESUMEN

Genotoxic agents, that are used in cancer therapy, elicit the reprogramming of the transcriptome of cancer cells. These changes reflect the cellular response to stress and underlie some of the mechanisms leading to drug resistance. Here, we profiled genome-wide changes in pre-mRNA splicing induced by cisplatin in breast cancer cells. Among the set of cisplatin-induced alternative splicing events we focused on COASY, a gene encoding a mitochondrial enzyme involved in coenzyme A biosynthesis. Treatment with cisplatin induces the production of a short isoform of COASY lacking exons 4 and 5, whose depletion impedes mitochondrial function and decreases sensitivity to cisplatin. We identified RBM39 as a major effector of the cisplatin-induced effect on COASY splicing. RBM39 also controls a genome-wide set of alternative splicing events partially overlapping with the cisplatin-mediated ones. Unexpectedly, inactivation of RBM39 in response to cisplatin involves its interaction with the AP-1 family transcription factor c-Jun that prevents RBM39 binding to pre-mRNA. Our findings therefore uncover a novel cisplatin-induced interaction between a splicing regulator and a transcription factor that has a global impact on alternative splicing and contributes to drug resistance.


Asunto(s)
Empalme Alternativo , Cisplatino , Resistencia a Antineoplásicos , Proteínas de Unión al ARN , Factores de Transcripción , Empalme Alternativo/genética , Cisplatino/farmacología , Cisplatino/metabolismo , Daño del ADN , Proteínas Nucleares/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Línea Celular Tumoral , Humanos , Animales
3.
J Photochem Photobiol B ; 222: 112258, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34399205

RESUMEN

Photodynamic therapy (PDT) is an approved therapeutic approach and an alternative to conventional chemotherapy for the treatment of several types of cancer with the advantages of reducing the side effects and developing resistance mechanisms. Here, was evaluated the photosensitization capabilities of 5,10,15,20-tetrakis[4-(pyridinium-1-yl-methyl)phenyl]porphyrin (3), its N-confused isomer (4) and of the neutral precursors (1) and (2) and the results were compared with the ones obtained with the cationic 5,10,15,20-tetrakis(1-methylpyridinium-4-yl)porphyrin (TMPyP). Both regular porphyrin derivatives 1 and 3 showed higher efficiency to generate singlet oxygen than TMPyP. The PDT assays towards MCF-7 cells under red light irradiation (λ > 640 nm, 23.7 mW cm-2) demonstrated that the cationic porphyrin 3 is an efficient photosensitizer to kill MCF-7 breast cancer cells. The study of the cell death mechanisms induced by the photodynamic process showed that the studied porphyrin 3 and TMPyP caused cell death by autophagic flux and necrosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología , Apoptosis/efectos de la radiación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Femenino , Humanos , Luz , Células MCF-7 , Microscopía Confocal , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/química , Porfirinas/uso terapéutico , Oxígeno Singlete/metabolismo
4.
Nutrients ; 12(12)2020 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-33302552

RESUMEN

Growth differentiation factor-15 (GDF-15) and its receptor GFRAL are both involved in the development of obesity and insulin resistance. Plasmatic GDF-15 level increases with obesity and is positively associated with disease progression. Despite macrophages have been recently suggested as a key source of GDF-15 in obesity, little is known about the regulation of GDF-15 in these cells. In the present work, we sought for potential pathophysiological activators of GDF15 expression in human macrophages and identified saturated fatty acids (SFAs) as strong inducers of GDF15 expression and secretion. SFAs increase GDF15 expression through the induction of an ER stress and the activation of the PERK/eIF2/CHOP signaling pathway in both PMA-differentiated THP-1 cells and in primary monocyte-derived macrophages. The transcription factor CHOP directly binds to the GDF15 promoter region and regulates GDF15 expression. Unlike SFAs, unsaturated fatty acids do not promote GDF15 expression and rather inhibit both SFA-induced GDF15 expression and ER stress. These results suggest that free fatty acids may be involved in the control of GDF-15 and provide new molecular insights about how diet and lipid metabolism may regulate the development of obesity and T2D.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Ácidos Grasos/farmacología , Factor 15 de Diferenciación de Crecimiento/metabolismo , Macrófagos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/metabolismo , Supervivencia Celular/efectos de los fármacos , Dieta , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ácidos Grasos no Esterificados , Ácidos Grasos Insaturados , Regulación de la Expresión Génica/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Metabolismo de los Lípidos , Obesidad/metabolismo , ARN Interferente Pequeño , Células THP-1
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(7): 1017-1030, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30953761

RESUMEN

NLRP3 inflammasome plays a key role in Western diet-induced systemic inflammation and was recently shown to mediate long-lasting trained immunity in myeloid cells. Saturated fatty acids (SFAs) are sterile triggers able to induce the assembly of the NLRP3 inflammasome in macrophages, leading to IL-1ß secretion while unsaturated ones (UFAs) prevent SFAs-mediated NLRP3 activation. Unlike previous studies using LPS-primed bone marrow derived macrophages, we do not see any ROS or IRE-1α involvement in SFAs-mediated NLRP3 activation in human monocytes-derived macrophages. Rather we show that SFAs need to enter the cells and to be activated into acyl-CoA to lead to NLRP3 activation in human macrophages. However, their ß-oxidation is dispensable. Instead, they are channeled towards phospholipids but redirected towards lipid droplets containing triacylglycerol in the presence of UFAs. Lipidomic analyses and Laurdan fluorescence experiments demonstrate that SFAs induce a dramatic saturation of phosphatidylcholine (PC) correlated with a loss of membrane fluidity, both events inhibited by UFAs. The silencing of CCTα, the key enzyme in PC synthesis, prevents SFA-mediated NLRP3 activation, demonstrating the essential role of the de novo PC synthesis. This SFA-induced membrane remodeling promotes a disruption of the plasma membrane Na, K-ATPase, instigating a K+ efflux essential and sufficient for NLRP3 activation. This work opens novel therapeutic avenues to interfere with Western diet-associated diseases such as those targeting the glycerolipid pathway.


Asunto(s)
Ácidos Grasos/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Potasio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Transporte Biológico , Células Cultivadas , Humanos , Inflamasomas/metabolismo , Fosfolípidos/metabolismo
6.
Biochem Pharmacol ; 153: 217-229, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29371029

RESUMEN

Overexpression of the ubiquitous type II melanoma antigen-D2 (MAGED2) in numerous types of cancer suggests that this protein contributes to carcinogenesis, a well-documented characteristic of other MAGE proteins. Modification of MAGED2 intracellular localization during cell cycle phases and following treatment with camptothecin (CPT) and phosphorylation by ATM/ATR following ionizing irradiation led us to investigate the molecular functions of MAGED2 in the cellular response to DNA damage. Cell cycle regulators, cell cycle progression, and bromodeoxyuridine (BrdU) incorporation were compared between MAGED2-sufficient and -depleted U2OS cells following exposure to CPT. At 24 h post-CPT removal, MAGED2-depleted cells had lower levels of p21 and p27, and there was an increase in S phase BrdU-positive cells with a concurrent decrease in cells in G2. These cell cycle modifications were p21-independent, but ATR-, SKP2-, and CDC20-dependent. Importantly, while MAGED2 depletion reduced CHK2 phosphorylation after 8 h of CPT treatment, it enhanced and prolonged CHK1 phosphorylation after a 24 h recovery period, indicating sustained ATR activation. MAGED2 depletion had no impact on cell survival under our experimental conditions. In summary, our data indicate that MAGED2 reduced CPT-related replicative stress, suggesting a role for this protein in genomic stability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Daño del ADN/fisiología , Reparación del ADN/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas de Ciclo Celular/deficiencia , Supervivencia Celular/fisiología , Células HeLa , Humanos
7.
J Innate Immun ; 9(2): 162-180, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27855374

RESUMEN

The cell wall of mycobacteria is characterised by glycolipids composed of different classes of mycolic acids (MAs; alpha-, keto-, and methoxy-) and sugars (trehalose, glucose, and arabinose). Studies using mutant Mtb strains have shown that the structure of MAs influences the inflammatory potential of these glycolipids. As mutant Mtb strains possess a complex mixture of glycolipids, we analysed the inflammatory potential of single classes of mycolate esters of the Mtb cell wall using 38 different synthetic analogues. Our results show that synthetic trehalose dimycolate (TDM) and trehalose, glucose, and arabinose monomycolates (TMM, GMM, and AraMM) activate bone marrow-derived dendritic cells in terms of the production of pro-inflammatory cytokines (IL-6 and TNF-α) and reactive oxygen species, upregulation of costimulatory molecules, and activation of NLRP3 inflammasome by a mechanism dependent on Mincle. These findings demonstrate that Mincle receptor can also recognise pentose esters and seem to contradict the hypothesis that production of GMM is an escape mechanism used by pathogenic mycobacteria to avoid recognition by the innate immune system. Finally, our experiments indicate that TMM and GMM, as well as TDM, can promote Th1 and Th17 responses in mice in an OVA immunisation model, and that further analysis of their potential as novel adjuvants for subunit vaccines is warranted.


Asunto(s)
Pared Celular/inmunología , Células Dendríticas/fisiología , Glucolípidos/inmunología , Inflamación/inmunología , Mycobacterium tuberculosis/inmunología , Ácidos Micólicos/química , Tuberculosis/inmunología , Adyuvantes Inmunológicos , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular , Células Cultivadas , Ésteres/química , Glucosa , Glucolípidos/síntesis química , Inflamasomas/metabolismo , Interleucina-6/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trehalosa/química , Trehalosa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 113-124, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27984090

RESUMEN

Glioblastomas are the deadliest type of brain cancer and are frequently associated with poor prognosis and a high degree of recurrence despite removal by surgical resection and treatment by chemo- and radio-therapy. Photodynamic therapy (PDT) is a treatment well known to induce mainly necrotic and apoptotic cell death in solid tumors. 5-Aminolevulinic acid (5-ALA)-based PDT was recently shown to sensitize human glioblastoma cells (LN-18) to a RIP3 (Receptor Interacting Protein 3)-dependent cell death which is counter-acted by activation of autophagy. These promising results led us to investigate the pathways involved in cell death and survival mechanisms occurring in glioblastoma following PDT. In the present study, we describe a new TSC2 (Tuberous Sclerosis 2)-dependent survival pathway implicating MK2 (MAPKAPK2) kinase and 14-3-3 proteins which conducts to the activation of a pro-survival autophagy. Moreover, we characterized a new RIP3/TSC2 complex where RIP3 is suggested to promote cell death by targeting TSC2-dependent survival pathway. These results highlight (i) a new role of TSC2 to protect glioblastoma against PDT-induced cell death and (ii) TSC2 and 14-3-3 as new RIP3 partners.


Asunto(s)
Proteínas 14-3-3/genética , Ácido Aminolevulínico/farmacología , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Neuroglía/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Proteínas 14-3-3/antagonistas & inhibidores , Proteínas 14-3-3/metabolismo , Ácido Aminolevulínico/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Luz , Neuroglía/metabolismo , Neuroglía/patología , Fotoquimioterapia , Fármacos Fotosensibilizantes/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo
9.
Biochem Pharmacol ; 114: 14-21, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27238288

RESUMEN

The atypical chemokine receptor CXCR7/ACKR3 binds two endogenous chemokines, CXCL12 and CXCL11, and is upregulated in many cancers or following infection by several cancer-inducing viruses, including HHV-8. ACKR3 is a ligand-scavenging receptor and does not activate the canonical G protein pathways but was proposed to trigger ß-arrestin-dependent signaling. Here, we identified the human herpesvirus 8-encoded CC chemokine vCCL2/vMIP-II as a third high-affinity ligand for ACKR3. vCCL2 acted as partial ACKR3 agonist, inducing ß-arrestin recruitment to the receptor, subsequent reduction of its surface levels and its delivery to endosomes. In addition, ACKR3 reduced vCCL2-triggered MAP kinase and PI3K/Akt signaling through other chemokine receptors. Our data suggest that ACKR3 acts as a scavenger receptor for vCCL2, regulating its availability and activity toward human receptors, thereby likely controlling its function in HHV-8 infection. Our study provides new insights into the complex crosstalk between viral chemokines and host receptors as well as into the biology of ACKR3, this atypical and still enigmatic receptor.


Asunto(s)
Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Herpesvirus Humano 8/metabolismo , Receptores CXCR/agonistas , Animales , Unión Competitiva , Células CHO , Línea Celular Tumoral , Quimiocina CCL2/genética , Quimiocina CXCL12/metabolismo , Quimiocinas/genética , Cricetulus , Endosomas/metabolismo , Citometría de Flujo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Humanos , Ligandos , Receptor Cross-Talk , Receptores CXCR/genética , Transfección , Arrestina beta 2/metabolismo
10.
Biochim Biophys Acta ; 1863(4): 581-95, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26705694

RESUMEN

Melanoma antigen D2 (MAGE-D2) is recognized as a cancer diagnostic marker; however, it has poorly characterized functions. Here, we established its intracellular localization and shuttling during cell cycle progression and in response to cellular stress. In normal conditions, MAGE-D2 is present in the cytoplasm, nucleoplasm, and nucleoli. Within the latter, MAGE-D2 is mostly found in the granular and the dense fibrillar components, and it interacts with nucleolin. Transfection of MAGE-D2 deletion mutants demonstrated that Δ203-254 leads to confinement of MAGE-D2 to the cytoplasm, while Δ248-254 prevents its accumulation in nucleoli but still allows its presence in the nucleoplasm. Consequently, this short sequence belongs to a nucleolar localization signal. MAGE-D2 deletion does not alter the nucleolar organization or rRNA levels. However, its intracellular localization varies with the cell cycle in a different kinetic than nucleolin. After genotoxic and nucleolar stresses, MAGE-D2 is excluded from nucleoli and concentrates in the nucleoplasm. We demonstrated that its camptothecin-related delocalization results from two distinct events: a rapid nucleolar release and a slower phospho-ERK-dependent cytoplasm to nucleoplasm translocation, which results from an increased flux from the cytoplasm to nucleoplasm. In conclusion, MAGE-D2 is a dynamic protein whose shuttling properties could suggest a role in cell cycle regulation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos de Neoplasias/metabolismo , Ciclo Celular/fisiología , Estrés Fisiológico/fisiología , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Células MCF-7 , Transporte de Proteínas , Células Tumorales Cultivadas
11.
BMC Cancer ; 15: 227, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25884497

RESUMEN

BACKGROUND: Modification of splicing by chemotherapeutic drugs has usually been evaluated on a limited number of pre-mRNAs selected for their recognized or potential importance in cell proliferation or apoptosis. However, the pathways linking splicing alterations to the efficiency of cancer therapy remain unclear. METHODS: Next-generation sequencing was used to analyse the transcriptome of breast carcinoma cells treated by cisplatin. Pharmacological inhibitors, RNA interference, cells deficient in specific signalling pathways, RT-PCR and FACS analysis were used to investigate how the anti-cancer drug cisplatin affected alternative splicing and the cell death pathway. RESULTS: We identified 717 splicing events affected by cisplatin, including 245 events involving cassette exons. Gene ontology analysis indicates that cell cycle, mRNA processing and pre-mRNA splicing were the main pathways affected. Importantly, the cisplatin-induced splicing alterations required class I PI3Ks P110ß but not components such as ATM, ATR and p53 that are involved in the DNA damage response. The siRNA-mediated depletion of the splicing regulator SRSF4, but not SRSF6, expression abrogated many of the splicing alterations as well as cell death induced by cisplatin. CONCLUSION: Many of the splicing alterations induced by cisplatin are caused by SRSF4 and they contribute to apoptosis in a process requires class I PI3K.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Precursores del ARN/genética , Empalme del ARN/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Línea Celular Tumoral , Biología Computacional , Daño del ADN , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Factores de Empalme Serina-Arginina , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Photochem Photobiol Sci ; 14(8): 1510-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25656235

RESUMEN

The response of tumors to photodynamic therapy (PDT) largely varies depending upon the intensity of the stress created in the cancer cells and also in the local environment. Singlet oxygen has been demonstrated, in many instances, to be the primary reactive oxygen species generated by PDT and responsible for most of the cellular effects. Cancer cells have developed various sensors which activate signalling pathways in response to PDT, and the nature of the activated pathway varies with the PDT stress intensity. At low doses of PDT, signalling pathways allow cancer cells to both proliferate and switch on pro-survival responses such as autophagy. Above a certain level of PDT stress intensity, cancer cells cannot cope with the heavy damage, and signalling pathways leading to cell death are activated. Two types of regulated cell death have been shown to be induced by PDT: apoptosis and necrosis. Signalling pathways activating NF-κB transcription factors have the peculiar characteristic of being activated both at low and high doses of PDT. These pathways coordinate the cross-talk between the immune system via the release of cytokines and chemokines and an anti-cell death response via the control of apoptosis and necrosis. Therefore, NF-κB induced by PDT appears to play a positive role in educating the immune system to fight tumors but also a negative role in helping cancer cells to survive the stress generated by singlet oxygen. This is why NF-κB cannot easily be considered as a pharmacological target whose inhibition will favor tumor cell eradication by PDT.


Asunto(s)
FN-kappa B/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Fotoquimioterapia , Animales , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Humanos , Fotoquimioterapia/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
13.
Arthritis Res Ther ; 16(1): R58, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24572376

RESUMEN

INTRODUCTION: Recent evidence suggests that tissue accumulation of senescent p16INK4a-positive cells during the life span would be deleterious for tissue functions and could be the consequence of inherent age-associated disorders. Osteoarthritis (OA) is characterized by the accumulation of chondrocytes expressing p16INK4a and markers of the senescence-associated secretory phenotype (SASP), including the matrix remodeling metalloproteases MMP1/MMP13 and pro-inflammatory cytokines interleukin-8 (IL-8) and IL-6. Here, we evaluated the role of p16INK4a in the OA-induced SASP and its regulation by microRNAs (miRs). METHODS: We used IL-1-beta-treated primary OA chondrocytes cultured in three-dimensional setting or mesenchymal stem cells differentiated into chondrocyte to follow p16INK4a expression. By transient transfection experiments and the use of knockout mice, we validate p16INK4a function in chondrocytes and its regulation by one miR identified by means of a genome-wide miR-array analysis. RESULTS: p16INK4a is induced upon IL-1-beta treatment and also during in vitro chondrogenesis. In the mouse model, Ink4a locus favors in vivo the proportion of terminally differentiated chondrocytes. When overexpressed in chondrocytes, p16INK4a is sufficient to induce the production of the two matrix remodeling enzymes, MMP1 and MMP13, thus linking senescence with OA pathogenesis and bone development. We identified miR-24 as a negative regulator of p16INK4a. Accordingly, p16INK4a expression increased while miR-24 level was repressed upon IL-1-beta addition, in OA cartilage and during in vitro terminal chondrogenesis. CONCLUSIONS: We disclosed herein a new role of the senescence marker p16INK4a and its regulation by miR-24 during OA and terminal chondrogenesis.


Asunto(s)
Condrocitos/patología , Condrogénesis/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , MicroARNs/metabolismo , Osteoartritis/metabolismo , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Western Blotting , Diferenciación Celular/fisiología , Senescencia Celular/fisiología , Condrocitos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoartritis/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
14.
Free Radic Biol Med ; 67: 292-303, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24269898

RESUMEN

Emerging evidence indicates that oxidative stress instigates the formation of ubiquitin (Ub) aggregates, substrates of autophagy, through a process requiring the ubiquitin binding adaptors p62/SQSTM1 and NBR1. Here, we have investigated the role of p62 and NBR1 in cell survival after hypericin-mediated photodynamic therapy (Hyp-PDT), a procedure known to incite robust reactive oxygen species (ROS)-based endoplasmic reticulum stress and autophagy pathways. We found that Hyp-PDT stimulated the formation of p62- and NBR1-associated Ub aggregates in normal and cancer cells, which were ultimately removed by autophagy, through a mechanism partially regulated by p38(MAPK). In line with this, genetic or pharmacological p38(MAPK) inhibition reduced p62 and NBR1 levels and aggregate formation and impaired Nrf2 activation, thus increasing photo-oxidative stress and cell death. p62-deficient cells, or cells lacking p62 and with reduced levels of NBR1 (through siRNA knockdown), also displayed reduced aggregate formation but exhibited attenuated ROS levels, reduced caspase activation, and improved survival after Hyp-PDT. The increased resistance to photo-oxidative stress exhibited by cells lacking p62 and/or NBR1 was overruled by the inhibition of p38(MAPK), which restored cytotoxic ROS levels, thus indicating the relevance of this signal in the control of cell viability. Taken together these findings provide evidence that in photodynamically treated cells a p38(MAPK)-regulated pathway coordinates the p62/NBR1-mediated clearance of cytosolic aggregates and mitigates PDT-induced proteotoxicity. They also reveal that a functional p38(MAPK)-Nrf2 signal is required to keep ROS levels in check and protect against PDT-induced proteotoxicity, independent of aggregate formation.


Asunto(s)
Autofagia/genética , Fibroblastos/metabolismo , Factor 2 Relacionado con NF-E2/genética , Proteínas/genética , Proteínas de Unión al ARN/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Animales , Antracenos , Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Línea Celular , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Luz , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Perileno/análogos & derivados , Perileno/farmacología , Agregado de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/metabolismo , Proteolisis , Proteínas de Unión al ARN/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Ubiquitina/genética , Ubiquitina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Lipid Res ; 54(11): 2998-3008, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24006511

RESUMEN

The NLRP3 inflammasome is involved in many obesity-associated diseases, such as type 2 diabetes, atherosclerosis, and gouty arthritis, through its ability to induce interleukin (IL)-1ß release. The molecular link between obesity and inflammasome activation is still unclear, but free fatty acids have been proposed as one triggering event. Here we reported opposite effects of saturated fatty acids (SFAs) compared with unsaturated fatty acids (UFAs) on NLRP3 inflammasome in human monocytes/macrophages. Palmitate and stearate, both SFAs, triggered IL-1ß secretion in a caspase-1/ASC/NLRP3-dependent pathway. Unlike SFAs, the UFAs oleate and linoleate did not lead to IL-1ß secretion. In addition, they totally prevented the IL-1ß release induced by SFAs and, with less efficiency, by a broad range of NLRP3 inducers, including nigericin, alum, and monosodium urate. UFAs did not affect the transcriptional effect of SFAs, suggesting a specific effect on the NLRP3 activation. These results provide a new anti-inflammatory mechanism of UFAs by preventing the activation of the NLRP3 inflammasome and, therefore, IL-1ß processing. By this way, UFAs might play a protective role in NLRP3-associated diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Ácidos Grasos Insaturados/farmacología , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Línea Celular , Humanos , Interleucina-1beta/metabolismo , Macrófagos/citología , Monocitos/citología , Proteína con Dominio Pirina 3 de la Familia NLR , Palmitatos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estearatos/farmacología
16.
J Biomed Nanotechnol ; 9(8): 1432-40, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23926811

RESUMEN

This study investigates the use of a new biocompatible block copolymer poly(2-(dimethylamino)ethyl methacrylate-N-(morpholino)ethyl methacrylate (PDMAEMA-b-PMEMA) for the delivery of a particular antisense oligonucleotide targeting E6 gene from human papilloma virus. This antisense oligonucleotide was derivatized with a polyazaaromatic Ru(II) complex which, under visible illumination, is able to produce an irreversible crosslink with the complementary targeted sequence. The purpose of this study is to determine whether by the use of a suitable transfection agent, it is possible to increase the efficiency of the antisense oligonucleotide targeting E6 gene, named Ru-P-4. In a recent study, we showed that Oligofectamine transfected Ru-P-4 antisense oligonucleotide failed to inhibit efficiently the growth of cervical cancer cell line SiHa, contrarily to the Ru-P-6 antisense oligonucleotide, another sequence also targeting the E6 gene. The ability of PDMAEMA-b-PMEMA to form polyplexes with optimal physicochemical characteristics was investigated first. Then the ability of the PDMAEMA-b-PMEMA/Ru-P-4 antisense oligonucleotide polyplexes to transfect two keratinocyte cell lines (SiHa and HaCat) and the capacity of polyplexes to inhibit HPV16+ cervical cancer cell growth was evaluated. PDMAEMA-b-PMEMA base polyplexes at the optimal molar ratio of polymer nitrogen atoms to DNA phosphates (N/P), were able to deliver Ru-P-4 antisense oligonucleotide and to induce a higher growth inhibition in human cervical cancer SiHa cells, compared to other formulations based on Oligofectamine.


Asunto(s)
Sistemas de Liberación de Medicamentos , Metilmetacrilatos/química , Morfolinos/química , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos/administración & dosificación , Proteínas Oncogénicas Virales/genética , Proteínas Represoras/genética , Rutenio/química , Células Cultivadas , Femenino , Terapia Genética/instrumentación , Terapia Genética/métodos , Papillomavirus Humano 16/efectos de los fármacos , Papillomavirus Humano 16/genética , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/virología , Ensayo de Materiales , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Interferencia de ARN , Proteínas Represoras/antagonistas & inhibidores , Transfección/instrumentación , Transfección/métodos
17.
PLoS Pathog ; 9(3): e1003234, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23555249

RESUMEN

Exposure to hepatitis C virus (HCV) typically results in chronic infection that leads to progressive liver disease ranging from mild inflammation to severe fibrosis and cirrhosis as well as primary liver cancer. HCV triggers innate immune signaling within the infected hepatocyte, a first step in mounting of the adaptive response against HCV infection. Persistent inflammation is strongly associated with liver tumorigenesis. The goal of our work was to investigate the initiation of the inflammatory processes triggered by HCV viral proteins in their host cell and their possible link with HCV-related liver cancer. We report a dramatic upregulation of the lymphotoxin signaling pathway and more specifically of lymphotoxin-ß in tumors of the FL-N/35 HCV-transgenic mice. Lymphotoxin expression is accompanied by activation of NF-κB, neosynthesis of chemokines and intra-tumoral recruitment of mononuclear cells. Spectacularly, IKKß inactivation in FL-N/35 mice drastically reduces tumor incidence. Activation of lymphotoxin-ß pathway can be reproduced in several cellular models, including the full length replicon and HCV-infected primary human hepatocytes. We have identified NS5B, the HCV RNA dependent RNA polymerase, as the viral protein responsible for this phenotype and shown that pharmacological inhibition of its activity alleviates activation of the pro-inflammatory pathway. These results open new perspectives in understanding the inflammatory mechanisms linked to HCV infection and tumorigenesis.


Asunto(s)
Hepacivirus/enzimología , Neoplasias Hepáticas/metabolismo , Linfocitos/inmunología , Linfotoxina beta/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Animales , Línea Celular , Quimiocinas/metabolismo , Quimiotaxis de Leucocito , Hepacivirus/patogenicidad , Hepatocitos/metabolismo , Hepatocitos/patología , Hepatocitos/virología , Interacciones Huésped-Patógeno , Humanos , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Hígado/metabolismo , Hígado/patología , Hígado/virología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Activación de Linfocitos , Linfocitos/virología , Masculino , Ratones , Ratones Transgénicos , FN-kappa B , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Transducción de Señal , Regulación hacia Arriba , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/metabolismo
18.
J Virol ; 87(5): 2868-81, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269791

RESUMEN

The role of the tegument during the herpesvirus lytic cycle is still not clearly established, particularly at the late phase of infection, when the newly produced viral particles need to be fully assembled before being released from the infected cell. The varicella-zoster virus (VZV) protein coded by open reading frame (ORF) 9 (ORF9p) is an essential tegument protein, and, even though its mRNA is the most expressed during the productive infection, little is known about its functions. Using a GalK positive/negative selection technique, we modified a bacterial artificial chromosome (BAC) containing the complete VZV genome to create viruses expressing mutant versions of ORF9p. We showed that ORF9p is hyperphosphorylated during the infection, especially through its interaction with the viral Ser/Thr kinase ORF47p; we identified a consensus site within ORF9p recognized by ORF47p and demonstrated its importance for ORF9p phosphorylation. Strikingly, an ultrastructural analysis revealed that the mutation of this consensus site (glutamate 85 to arginine) strongly affects viral assembly and release, reproducing the ORF47 kinase-dead VZV phenotype. It also slightly diminishes the infectivity toward immature dendritic cells. Taken together, our results identify ORF9p as a new viral substrate of ORF47p and suggest a determinant role of this phosphorylation for viral infectivity, especially during the process of viral particle formation and egress.


Asunto(s)
Herpesvirus Humano 3/metabolismo , Proteínas Virales/metabolismo , Liberación del Virus , Línea Celular Tumoral , Cromosomas Artificiales Bacterianos , Células Dendríticas/inmunología , Células HEK293 , Herpesvirus Humano 3/fisiología , Herpesvirus Humano 3/ultraestructura , Humanos , Mutación , Sistemas de Lectura Abierta , Fosforilación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Virales/genética , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo , Virión/fisiología , Virión/ultraestructura , Ensamble de Virus , Replicación Viral
19.
Autophagy ; 8(9): 1312-24, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22889744

RESUMEN

Although reactive oxygen species (ROS) have been reported to evoke different autophagic pathways, how ROS or their secondary products modulate the selective clearance of oxidatively damaged organelles is less explored. To investigate the signaling role of ROS and the impact of their compartmentalization in autophagy pathways, we used murine fibrosarcoma L929 cells overexpressing different antioxidant enzymes targeted to the cytosol or mitochondria and subjected them to photodynamic (PD) stress with the endoplasmic reticulum (ER)-associated photosensitizer hypericin. We show that following apical ROS-mediated damage to the ER, predominantly cells overexpressing mitochondria-associated glutathione peroxidase 4 (GPX4) and manganese superoxide dismutase (SOD2) displayed attenuated kinetics of autophagosome formation and overall cell death, as detected by computerized time-lapse microscopy. Consistent with a primary ER photodamage, kinetics and colocalization studies revealed that photogenerated ROS induced an initial reticulophagy, followed by morphological changes in the mitochondrial network that preceded clearance of mitochondria by mitophagy. Overexpression of cytosolic and mitochondria-associated GPX4 retained the tubular mitochondrial network in response to PD stress and concomitantly blocked the progression toward mitophagy. Preventing the formation of phospholipid hydroperoxides and H(2)O(2) in the cytosol as well as in the mitochondria significantly reduced cardiolipin peroxidation and apoptosis. All together, these results show that in response to apical ER photodamage ROS propagate to mitochondria, which in turn amplify ROS production, thereby contributing to two antagonizing processes, mitophagy and apoptosis.


Asunto(s)
Autofagia/efectos de los fármacos , Mitocondrias/metabolismo , Orgánulos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Antracenos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Orgánulos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Perileno/análogos & derivados , Perileno/farmacología , Factores de Tiempo
20.
PLoS One ; 7(7): e41005, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815893

RESUMEN

SHIP-1 is an inositol phosphatase predominantly expressed in hematopoietic cells. Over the ten past years, SHIP-1 has been described as an important regulator of immune functions. Here, we characterize a new inhibitory function for SHIP-1 in NOD2 signaling. NOD2 is a crucial cytoplasmic bacterial sensor that activates proinflammatory and antimicrobial responses upon bacterial invasion. We observed that SHIP-1 decreases NOD2-induced NF-κB activation in macrophages. This negative regulation relies on its interaction with XIAP. Indeed, we observed that XIAP is an essential mediator of the NOD2 signaling pathway that enables proper NF-κB activation in macrophages. Upon NOD2 activation, SHIP-1 C-terminal proline rich domain (PRD) interacts with XIAP, thereby disturbing the interaction between XIAP and RIP2 in order to decrease NF-κB signaling.


Asunto(s)
FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Células Epiteliales/citología , Regulación de la Expresión Génica , Humanos , Sistema Inmunológico , Inflamación , Inositol Polifosfato 5-Fosfatasas , Macrófagos/citología , Macrófagos/metabolismo , Modelos Biológicos , Monocitos/citología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Estructura Terciaria de Proteína , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA