Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Hepatology ; 78(4): 1050-1063, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36089330

RESUMEN

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death. The NF-κB transcription factor family subunit c-Rel is typically protumorigenic; however, it has recently been reported as a tumor suppressor. Here, we investigated the role of c-Rel in HCC. APPROACH AND RESULTS: Histological and transcriptional studies confirmed expression of c-Rel in human patients with HCC, but low c-Rel expression correlated with increased tumor cell proliferation and mutational burden and was associated with advanced disease. In vivo , global ( Rel-/- ) and epithelial specific ( RelAlb ) c-Rel knockout mice develop more tumors, with a higher proliferative rate and increased DNA damage, than wild-type (WT) controls 30 weeks after N-diethylnitrosamine injury. However, tumor burden was comparable when c-Rel was deleted in hepatocytes once tumors were established, suggesting c-Rel signaling is important for preventing HCC initiation after genotoxic injury, rather than for HCC progression. In vitro , Rel-/- hepatocytes were more susceptible to genotoxic injury than WT controls. ATM-CHK2 DNA damage response pathway proteins were suppressed in Rel-/- hepatocytes following genotoxic injury, suggesting that c-Rel is required for effective DNA repair. To determine if c-Rel inhibition sensitizes cancer cells to chemotherapy, by preventing repair of chemotherapy-induced DNA damage, thus increasing tumor cell death, we administered single or combination doxorubicin and IT-603 (c-Rel inhibitor) therapy in an orthotopic HCC model. Indeed, combination therapy was more efficacious than doxorubicin alone. CONCLUSION: Hepatocyte c-Rel signaling limits genotoxic injury and subsequent HCC burden. Inhibiting c-Rel as an adjuvant therapy increased the effectiveness of DNA damaging agents and reduced HCC growth.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Ratones , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Daño del ADN , Doxorrubicina/farmacología , Hepatocitos/metabolismo , Neoplasias Hepáticas/metabolismo , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo
2.
Biochem J ; 479(11): 1121-1126, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35647902

RESUMEN

Numerous studies, published over many years, have established the key role that the IκB kinase (IKK) subunits, α and ß, play in regulating the Nuclear Factor κB (NF-κB) pathway. This research generally concluded that their functions can be separated, with IKKß being the critical regulator of the canonical NF-κB pathway, while IKKα functions as the key activating kinase for the non-canonical pathway. However, other roles for these kinases have been described and several reports concluded that this separation of their functions may not always be the case. This commentary discusses the recent report by Biochem J. 479, 305-325, who elegantly demonstrate that in KRAS driven colorectal cancer cell lines, IKKα is an important regulator of the canonical NF-κB pathway. As is so often the case with trying to understand the complexity of NF-κB signalling, cellular context is everything.


Asunto(s)
Quinasa I-kappa B , FN-kappa B , Línea Celular , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas , Transducción de Señal
3.
Biochem J ; 479(19): 2115-2130, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36240068

RESUMEN

Claspin is an adaptor protein required for ATR-dependent phosphorylation of CHK1 during S-phase following DNA replication stress. Claspin expression is highly variable in cancer, with low levels frequently correlating with poor patient survival. To learn more about the biological consequences of reduced Claspin expression and its effects on tumorigenesis, we investigated mice with a heterozygous knockout of the Clspn gene. Claspin haploinsufficiency resulted in reduced female fertility and a maternally inherited defect in oocyte meiosis I cell cycle progression. Furthermore, aged Clspn+/- mice developed spontaneous lymphoid hyperplasia and increased susceptibility to non-alcoholic fatty liver disease. Importantly, we demonstrate a tumour suppressor role for Claspin. Reduced Claspin levels result in increased liver damage and tumourigenesis in the DEN model of hepatocellular carcinoma. These data reveal that Clspn haploinsufficiency has widespread unanticipated biological effects and establishes the importance of Claspin as a regulatory node controlling tumorigenesis and multiple disease aetiologies.


Asunto(s)
Replicación del ADN , Haploinsuficiencia , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Femenino , Fertilidad/genética , Hiperplasia , Ratones , Fosforilación
4.
Biochem J ; 479(19): 2063-2086, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36240066

RESUMEN

Previously, we discovered that deletion of c-Rel in the Eµ-Myc mouse model of lymphoma results in earlier onset of disease, a finding that contrasted with the expected function of this NF-κB subunit in B-cell malignancies. Here we report that Eµ-Myc/cRel-/- cells have an unexpected and major defect in the CHK1 pathway. Total and phospho proteomic analysis revealed that Eµ-Myc/cRel-/- lymphomas highly resemble wild-type (WT) Eµ-Myc lymphomas treated with an acute dose of the CHK1 inhibitor (CHK1i) CCT244747. Further analysis demonstrated that this is a consequence of Eµ-Myc/cRel-/- lymphomas having lost expression of CHK1 protein itself, an effect that also results in resistance to CCT244747 treatment in vivo. Similar down-regulation of CHK1 protein levels was also seen in CHK1i resistant U2OS osteosarcoma and Huh7 hepatocellular carcinoma cells. Further investigation revealed that the deubiquitinase USP1 regulates CHK1 proteolytic degradation and that its down-regulation in our model systems is responsible, at least in part, for these effects. We demonstrate that treating WT Eµ-Myc lymphoma cells with the USP1 inhibitor ML323 was highly effective at reducing tumour burden in vivo. Targeting USP1 activity may thus be an alternative therapeutic strategy in MYC-driven tumours.


Asunto(s)
Linfoma , Proteínas Proto-Oncogénicas c-myc , Aminopiridinas , Animales , Enzimas Desubicuitinizantes , Linfoma/metabolismo , Linfoma/patología , Ratones , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteómica , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirimidinas
5.
Biochem J ; 479(19): 2131-2151, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36240067

RESUMEN

The development of resistance and the activation of bypass pathway signalling represents a major problem for the clinical application of protein kinase inhibitors. While investigating the effect of either a c-Rel deletion or RelAT505A phosphosite knockin on the Eµ-Myc mouse model of B-cell lymphoma, we discovered that both NF-κB subunit mutations resulted in CHK1 inhibitor resistance, arising from either loss or alteration of CHK1 activity, respectively. However, since Eµ-Myc lymphomas depend on CHK1 activity to cope with high levels of DNA replication stress and consequent genomic instability, it was not clear how these mutant NF-κB subunit lymphomas were able to survive. To understand these survival mechanisms and to identify potential compensatory bypass signalling pathways in these lymphomas, we applied a multi-omics strategy. With c-Rel-/- Eµ-Myc lymphomas we observed high levels of Phosphatidyl-inositol 3-kinase (PI3K) and AKT pathway activation. Moreover, treatment with the PI3K inhibitor Pictilisib (GDC-0941) selectively inhibited the growth of reimplanted c-Rel-/- and RelAT505A, but not wild type (WT) Eµ-Myc lymphomas. We also observed up-regulation of a RHO/RAC pathway gene expression signature in both Eµ-Myc NF-κB subunit mutation models. Further investigation demonstrated activation of the RHO/RAC effector p21-activated kinase (PAK) 2. Here, the PAK inhibitor, PF-3758309 successfully overcame resistance of RelAT505A but not WT lymphomas. These findings demonstrate that up-regulation of multiple bypass pathways occurs in CHK1 inhibitor resistant Eµ-Myc lymphomas. Consequently, drugs targeting these pathways could potentially be used as either second line or combinatorial therapies to aid the successful clinical application of CHK1 inhibitors.


Asunto(s)
Linfoma , Fosfatidilinositol 3-Quinasas , Animales , Inositol , Linfoma/tratamiento farmacológico , Linfoma/genética , Linfoma/metabolismo , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Regulación hacia Arriba , Quinasas p21 Activadas/genética
6.
Int J Mol Sci ; 24(12)2023 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-37373413

RESUMEN

Introducing or correcting disease-causing mutations through genome editing in human pluripotent stem cells (hPSCs) followed by tissue-specific differentiation provide sustainable models of multiorgan diseases, such as cystic fibrosis (CF). However, low editing efficiency resulting in extended cell culture periods and the use of specialised equipment for fluorescence activated cell sorting (FACS) make hPSC genome editing still challenging. We aimed to investigate whether a combination of cell cycle synchronisation, single-stranded oligodeoxyribonucleotides, transient selection, manual clonal isolation, and rapid screening can improve the generation of correctly modified hPSCs. Here, we introduced the most common CF mutation, ΔF508, into the CFTR gene, using TALENs into hPSCs, and corrected the W1282X mutation using CRISPR-Cas9, in human-induced PSCs. This relatively simple method achieved up to 10% efficiency without the need for FACS, generating heterozygous and homozygous gene edited hPSCs within 3-6 weeks in order to understand genetic determinants of disease and precision medicine.


Asunto(s)
Edición Génica , Células Madre Pluripotentes , Humanos , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Células Madre Pluripotentes/metabolismo , Mutación , Heterocigoto
7.
Biochem J ; 478(3): 533-551, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33438746

RESUMEN

Different types of DNA damage can initiate phosphorylation-mediated signalling cascades that result in stimulus specific pro- or anti-apoptotic cellular responses. Amongst its many roles, the NF-κB transcription factor RelA is central to these DNA damage response pathways. However, we still lack understanding of the co-ordinated signalling mechanisms that permit different DNA damaging agents to induce distinct cellular outcomes through RelA. Here, we use label-free quantitative phosphoproteomics to examine the temporal effects of exposure of U2OS cells to either etoposide (ETO) or hydroxyurea (HU) by monitoring the phosphorylation status of RelA and its protein binding partners. Although few stimulus-specific differences were identified in the constituents of phosphorylated RelA interactome after exposure to these DNA damaging agents, we observed subtle, but significant, changes in their phosphorylation states, as a function of both type and duration of treatment. The DNA double strand break (DSB)-inducing ETO invoked more rapid, sustained responses than HU, with regulated targets primarily involved in transcription, cell division and canonical DSB repair. Kinase substrate prediction of ETO-regulated phosphosites suggest abrogation of CDK and ERK1 signalling, in addition to the known induction of ATM/ATR. In contrast, HU-induced replicative stress mediated temporally dynamic regulation, with phosphorylated RelA binding partners having roles in rRNA/mRNA processing and translational initiation, many of which contained a 14-3-3ε binding motif, and were putative substrates of the dual specificity kinase CLK1. Our data thus point to differential regulation of key cellular processes and the involvement of distinct signalling pathways in modulating DNA damage-specific functions of RelA.


Asunto(s)
Daño del ADN , Procesamiento Proteico-Postraduccional , Factor de Transcripción ReIA/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias Óseas/patología , Línea Celular Tumoral , Cromatografía Liquida , Secuencia de Consenso , Roturas del ADN de Doble Cadena , Replicación del ADN , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/metabolismo , Etopósido/farmacología , Humanos , Hidroxiurea/farmacología , Osteosarcoma/patología , Fosforilación , Mapas de Interacción de Proteínas , Proteínas Quinasas/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem , Factores de Tiempo
8.
Mol Cell ; 49(5): 922-33, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23375502

RESUMEN

The ARF tumor suppressor is a central component of the cellular defense against oncogene activation in mammals. p14ARF activates p53 by binding and inhibiting HDM2, resulting, inter alia, in increased transcription and expression of the cyclin-dependent kinase inhibitor p21 and consequent cell-cycle arrest. We analyzed the effect of p14ARF induction on nucleolar protein dynamics using SILAC mass spectrometry and have identified the human Formin-2 (FMN2) protein as a component of the p14ARF tumor suppressor pathway. We show that FMN2 is increased upon p14ARF induction at both the mRNA and the protein level via a NF-κB-dependent mechanism that is independent of p53. FMN2 enhances expression of the cell-cycle inhibitor p21 by preventing its degradation. FMN2 is also induced by activation of other oncogenes, hypoxia, and DNA damage. These results identify FMN2 as a crucial component in the regulation of p21 and consequent oncogene/stress-induced cell-cycle arrest in human cells.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas del Tejido Nervioso/genética , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Nucléolo Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Espectrometría de Masas , FN-kappa B/genética , Proteínas del Tejido Nervioso/metabolismo , Proteómica , ARN Mensajero/metabolismo , Proteína p14ARF Supresora de Tumor/genética , Proteína p14ARF Supresora de Tumor/metabolismo
9.
Mol Cell ; 45(1): 1-3, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22244324

RESUMEN

The importance of parallel signaling pathways controlling NF-κB subunit posttranslational modifications is demonstrated by Sen et al. (2012), who reveal that RelA (p65) sulfhydration, at its highly conserved cysteine 38 residue, regulates association with the coactivator RPS3, DNA binding, and antiapoptotic gene expression.

10.
BMC Microbiol ; 19(1): 68, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30922226

RESUMEN

BACKGROUND: Thermal regulation of gene expression occurs in many microorganisms, and is mediated via several typical mechanisms. Yersinia pestis is the causative agent of the plague and spreads by zoonotic transfer from fleas to mammalian blood with a concomitant rapid temperature change, from ambient to 37 °C, which induces the expression of capsular antigen (Caf1) that inhibits phagocytosis. Caf1 is formed into long polymeric fimbriae by a periplasmic chaperone (Caf1M) and outer membrane usher (Caf1A). All three are encoded on an operon regulated by an AraC-type transcription factor Caf1R. The aim of this study was to determine the role of Caf1R in the thermal control of caf1 operon gene expression. RESULTS: PCR analysis of cDNA demonstrated that the genes of the operon are transcribed as a single polycistronic mRNA. Bioinformatic analysis, supported by deletion mutagenesis, then revealed a region containing the promoter of this polycistronic transcript that was critical for Caf1 protein expression. Caf1R was found to be essential for Caf1 protein production. Finally, RT-PCR analysis and western blot experiments showed large, Caf1R dependent increases in caf1 operon transcripts upon a shift in temperature from 25 °C to 35 °C. CONCLUSIONS: The results show that thermal control of Caf1 polymer production is established at the transcriptional level, in a Caf1R dependent manner. This gives us new insights into how a virulent pathogen evades destruction by the immune system by detecting and responding to environmental changes.


Asunto(s)
Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Temperatura , Factores de Transcripción/genética , Yersinia pestis/genética , Regulación Bacteriana de la Expresión Génica , Evasión Inmune , Operón
11.
BMC Genomics ; 19(1): 813, 2018 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-30419821

RESUMEN

BACKGROUND: The cellular response to ionizing radiation involves activation of p53-dependent pathways and activation of the atypical NF-κB pathway. The crosstalk between these two transcriptional networks include (co)regulation of common gene targets. Here we looked for novel genes potentially (co)regulated by p53 and NF-κB using integrative genomics screening in human osteosarcoma U2-OS cells irradiated with a high dose (4 and 10 Gy). Radiation-induced expression in cells with silenced TP53 or RELA (coding the p65 NF-κB subunit) genes was analyzed by RNA-Seq while radiation-enhanced binding of p53 and RelA in putative regulatory regions was analyzed by ChIP-Seq, then selected candidates were validated by qPCR. RESULTS: We identified a subset of radiation-modulated genes whose expression was affected by silencing of both TP53 and RELA, and a subset of radiation-upregulated genes where radiation stimulated binding of both p53 and RelA. For three genes, namely IL4I1, SERPINE1, and CDKN1A, an antagonistic effect of the TP53 and RELA silencing was consistent with radiation-enhanced binding of both p53 and RelA. This suggested the possibility of a direct antagonistic (co)regulation by both factors: activation by NF-κB and inhibition by p53 of IL4I1, and activation by p53 and inhibition by NF-κB of CDKN1A and SERPINE1. On the other hand, radiation-enhanced binding of both p53 and RelA was observed in a putative regulatory region of the RRAD gene whose expression was downregulated both by TP53 and RELA silencing, which suggested a possibility of direct (co)activation by both factors. CONCLUSIONS: Four new candidates for genes directly co-regulated by NF-κB and p53 were revealed.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/genética , Radiación Ionizante , Sitios de Unión , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/radioterapia , Cromatina/genética , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , L-Aminoácido Oxidasa/genética , L-Aminoácido Oxidasa/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Osteosarcoma/patología , Osteosarcoma/radioterapia , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
12.
Mol Cell ; 38(4): 524-38, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20513428

RESUMEN

NF-kappaB and p53 are important regulators of the cellular response to stress. Here, we identify the Skp2 gene as being both an NF-kappaB and p53 target after DNA damage. However, Skp2 expression can be either induced or repressed in a manner requiring both the p52 NF-kappaB subunit and p53, with subsequent effects on autophagy, apoptosis, and p53 function. This process is regulated by the Akt(PKB)/GSK3beta pathway. When Akt is active, GSK3beta is repressed, allowing p52 and p53 to cooperatively induce Skp2 expression. However, if Akt is inactive, GSK3beta phosphorylates p52 at Ser 222. This modification disrupts p52 homodimer/Bcl-3 complexes and facilitates transcriptional repression by p52/-c-Rel. The Skp2 promoter therefore integrates signaling through the NF-kappaB, p53, and Akt/GSK3beta pathways to regulate cell fate in response to DNA damage.


Asunto(s)
Apoptosis , Autofagia , Glucógeno Sintasa Quinasa 3/metabolismo , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Proteína p53 Supresora de Tumor/metabolismo , Daño del ADN , Glucógeno Sintasa Quinasa 3 beta , Humanos , Transducción de Señal
15.
PLoS Genet ; 10(9): e1004642, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25255445

RESUMEN

There are two major pathways leading to induction of NF-κB subunits. The classical (or canonical) pathway typically leads to the induction of RelA or c-Rel containing complexes, and involves the degradation of IκBα in a manner dependent on IκB kinase (IKK) ß and the IKK regulatory subunit NEMO. The alternative (or non-canonical) pathway, involves the inducible processing of p100 to p52, leading to the induction of NF-κB2(p52)/RelB containing complexes, and is dependent on IKKα and NF-κB inducing kinase (NIK). Here we demonstrate that in primary human fibroblasts, the alternative NF-κB pathway subunits NF-κB2 and RelB have multiple, but distinct, effects on the expression of key regulators of the cell cycle, reactive oxygen species (ROS) generation and protein stability. Specifically, following siRNA knockdown, quantitative PCR, western blot analyses and chromatin immunoprecipitation (ChIP) show that NF-κB2 regulates the expression of CDK4 and CDK6, while RelB, through the regulation of genes such as PSMA5 and ANAPC1, regulates the stability of p21WAF1 and the tumour suppressor p53. These combine to regulate the activity of the retinoblastoma protein, Rb, leading to induction of polycomb protein EZH2 expression. Moreover, our ChIP analysis demonstrates that EZH2 is also a direct NF-κB target gene. Microarray analysis revealed that in fibroblasts, EZH2 antagonizes a subset of p53 target genes previously associated with the senescent cell phenotype, including DEK and RacGAP1. We show that this pathway provides the major route of crosstalk between the alternative NF-κB pathway and p53, a consequence of which is to suppress cell senescence. Importantly, we find that activation of NF-κB also induces EZH2 expression in CD40L stimulated cells from Chronic Lymphocytic Leukemia patients. We therefore propose that this pathway provides a mechanism through which microenvironment induced NF-κB can inhibit tumor suppressor function and promote tumorigenesis.


Asunto(s)
Senescencia Celular/genética , FN-kappa B/metabolismo , Complejo Represivo Polycomb 2/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Ligando de CD40/agonistas , Ligando de CD40/metabolismo , Análisis por Conglomerados , Proteína Potenciadora del Homólogo Zeste 2 , Activación Enzimática , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Modelos Biológicos , Subunidad p52 de NF-kappa B/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Unión Proteica , Estabilidad Proteica , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción ReIB/metabolismo , Transcripción Genética , Transcriptoma
16.
Trends Biochem Sci ; 37(8): 317-24, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22626470

RESUMEN

Among the characteristics acquired by many tumour cells is a shift from using oxidative phosphorylation to using glycolysis for ATP production. Although the nuclear factor (NF)-κB family of transcriptional regulators have important roles in tumorigenesis, their ability to function as regulators of metabolism has only been recently investigated. This has revealed the importance of crosstalk between NF-κB, the p53 tumour suppressor and other crucial cell signalling pathways. This review discusses the mechanisms through which NF-κB regulates tumour cell metabolism and the important role of p53 in determining the consequences of NF-κB activity. It also proposes a model in which NF-κB contributes to the shift to glycolytic ATP production through regulation of both nuclear and mitochondrial gene expression.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Glucólisis , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/genética , FN-kappa B/genética , Fosforilación Oxidativa , Transporte de Proteínas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/genética
17.
Br J Cancer ; 114(1): 1-6, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26757421

RESUMEN

When the genes encoding NF-κB subunits were first isolated, their homology to the previously identified c-Rel proto-oncogene and its viral homologue v-Rel was clear. This provided the first indication that these transcription factors also had a role in cancer. Because of its homology to v-Rel, which transforms chicken B cells together with the important role c-Rel can have as a regulator of B- and T-cell proliferation, most attention has focussed on its role in B-cell lymphomas, where the REL gene is frequently amplified. However, a growing number of reports now indicate that c-Rel has important functions in many solid tumours, although studies in mice suggest it may not always function as an oncogene. Moreover, c-Rel is a critical regulator of fibrosis, which provides an environment for tumour development in many settings. Overall, c-Rel is emerging as a complex regulator of tumorigenesis, and there is still much to learn about its functions in human malignancies and the response to cancer therapies.


Asunto(s)
Neoplasias/etiología , Proteínas Proto-Oncogénicas c-rel/fisiología , Animales , Fibrosis , Genes p53/fisiología , Genes rel/fisiología , Humanos , Linfoma de Células B/etiología , Ratones , Neoplasias/terapia , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-rel/química
18.
J Proteome Res ; 13(5): 2478-94, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24654937

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive malignancy characterized by an excessive resistance to all known anticancer therapies, a still largely elusive phenomenon. To identify original mechanisms, we have explored the role of post-translational modifications (PTMs) mediated by members of the ubiquitin family. Although alterations of these pathways have been reported in different cancers, no methodical search for these kinds of anomalies has been performed so far. Therefore, we studied the ubiquitin-, Nedd8-, and SUMO1-specific proteomes of a pancreatic cancer cell line (MiaPaCa-2) and identified changes induced by gemcitabine, the standard PDAC's chemotherapeutic drug. These PTMs profiles contained both known major substrates of all three modifiers as well as original ones. Gemcitabine treatment altered the PTM profile of proteins involved in various biological functions, some known cancer associated genes, many potentially cancer-associated genes, and several cancer-signaling networks, including canonical and noncanonical WNT and PI3K/Akt/MTOR pathways. Some of these altered PTMs formed groups of functionally and physically associated proteins. Importantly, we could validate the gemcitabine-induced PTMs variations of relevant candidates and we could demonstrate the biological significance of such altered PTMs by studying in detail the sumoylation of SNIP1, one of these new targets.


Asunto(s)
Antineoplásicos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Western Blotting , Línea Celular Tumoral , Cromatografía Liquida , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microscopía Fluorescente , Proteína NEDD8 , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem , Ubiquitina/genética , Ubiquitinas/genética , Ubiquitinas/metabolismo , Proteína Wnt1/metabolismo , Gemcitabina
19.
Biochem Soc Trans ; 42(1): 76-81, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24450631

RESUMEN

The NF-κB (nuclear factor κB) transcription factor family is a pleiotropic regulator of many cellular pathways, providing a mechanism for the cell to respond to a wide variety of stimuli and environmental challenges. It is not surprising therefore that an important component of NF-κB's function includes regulation of the cell cycle. However, this aspect of its behaviour is often overlooked and receives less attention than its ability to induce inflammatory gene expression. In the present article, we provide an updated review of the current state of our knowledge about integration of NF-κB activity with cell cycle regulation, including newly characterized direct and indirect target genes in addition to the mechanisms through which NF-κB itself can be regulated by the cell cycle.


Asunto(s)
Mitosis , FN-kappa B/fisiología , Animales , Ciclina D1/genética , Ciclina D1/metabolismo , Regulación de la Expresión Génica , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Transcripción Genética
20.
Hepatology ; 57(2): 817-28, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22996371

RESUMEN

UNLABELLED: Phosphorylation of the RelA subunit at serine 536 (RelA-P-Ser536) is important for hepatic myofibroblast survival and is mechanistically implicated in liver fibrosis. Here, we show that a cell-permeable competing peptide (P6) functions as a specific targeted inhibitor of RelA-P-Ser536 in vivo and exerts an antifibrogenic effect in two progressive liver disease models, but does not impair hepatic inflammation or innate immune responses after lipopolysaccharide challenge. Using kinase assays and western blotting, we confirm that P6 is a substrate for the inhibitory kappa B kinases (IKKs), IKKα and IKKß, and, in human hepatic myofibroblasts, P6 prevents RelA-P-Ser536, but does not affect IKK activation of IκBα. We demonstrate that RelA-P-Ser536 is a feature of human lung and skin fibroblasts, but not lung epithelial cells, in vitro and is present in sclerotic skin and diseased lungs of patients suffering from idiopathic pulmonary fibrosis. CONCLUSION: RelA-P-Ser536 may be a core fibrogenic regulator of fibroblast phenotype.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Cirrosis Hepática/prevención & control , Fragmentos de Péptidos/farmacología , Factor de Transcripción ReIA/antagonistas & inhibidores , Adulto , Animales , Intoxicación por Tetracloruro de Carbono/tratamiento farmacológico , Fibroblastos/metabolismo , Humanos , Quinasa I-kappa B/fisiología , Lipopolisacáridos/farmacología , Masculino , Ratones , Fragmentos de Péptidos/metabolismo , Fosforilación , Serina , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIA/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda