Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 139(3): 384-398, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-34232979

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most frequent lymphoid malignancy affecting adults. The NF-κB transcription factor family is activated by 2 main pathways, the canonical and the alternative NF-κB activation pathway, with different functions. The alternative NF-κB pathway leads to activation of the transcriptionally active RelB NF-κB subunit. Alternative NF-κB activation status and its role in DLBCL pathogenesis remain undefined. Here, we reveal a frequent activation of RelB in a large cohort of DLBCL patients and cell lines, independently of their activated B-cell-like or germinal center B-cell-like subtype. RelB activity defines a new subset of patients with DLBCL and a peculiar gene expression profile and mutational pattern. Importantly, RelB activation does not correlate with the MCD genetic subtype, enriched for activated B-cell-like tumors carrying MYD88L265P and CD79B mutations that cooperatively activate canonical NF-κB, thus indicating that current genetic tools to evaluate NF-κB activity in DLBCL do not provide information on the alternative NF-κB activation. Furthermore, the newly defined RelB-positive subgroup of patients with DLBCL exhibits a dismal outcome after immunochemotherapy. Functional studies revealed that RelB confers DLBCL cell resistance to DNA damage-induced apoptosis in response to doxorubicin, a genotoxic agent used in the front-line treatment of DLBCL. We also show that RelB positivity is associated with high expression of cellular inhibitor of apoptosis protein 2 (cIAP2). Altogether, RelB activation can be used to refine the prognostic stratification of DLBCL and may contribute to subvert the therapeutic DNA damage response in a segment of patients with DLBCL.


Asunto(s)
Linfoma de Células B Grandes Difuso/metabolismo , FN-kappa B/metabolismo , Factor de Transcripción ReIB/metabolismo , Apoptosis , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma de Células B Grandes Difuso/genética , FN-kappa B/genética , Factor de Transcripción ReIB/genética , Activación Transcripcional
2.
Chemphyschem ; 24(12): e202300151, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36973178

RESUMEN

Glutamine is under scrutiny regarding its metabolic deregulation linked to energetic reprogramming in cancer cells. Many analytical techniques have been used to better understand the impact of the metabolism of amino acids on biological processes, however only a few are suited to work with complex samples. Here, we report the use of a general dissolution dynamic nuclear polarization (D-DNP) formulation using an unexpensive radical as a multipurpose tool to study glutamine, with insights from enzymatic modelling to complex metabolic networks and fast imaging. First, hyperpolarized [5-13 C] glutamine is used as molecular probe to study the kinetic action of two enzymes: L-asparaginase that has been used as an anti-metabolic treatment for cancer, and glutaminase. These results are also compared with those acquired with another hyperpolarized amino acid, [1,4-13 C] asparagine. Second, we explored the use of hyperpolarized (HP) substrates to probe metabolic pathways by monitoring metabolic profiles arising from hyperpolarized glutamine in E. coli extracts. Finally, a highly concentrated sample formulation is proposed for the purpose of fast imaging applications. We think that this approach can be extended to formulate other amino acids as well as other metabolites and provide complementary insights into the analysis of metabolic networks.


Asunto(s)
Escherichia coli , Glutamina , Glutamina/análisis , Glutamina/química , Glutamina/metabolismo , Solubilidad , Escherichia coli/metabolismo , Redes y Vías Metabólicas , Aminoácidos/metabolismo , Isótopos de Carbono
3.
J Proteome Res ; 21(4): 1041-1051, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35119866

RESUMEN

Ultrahigh-resolution NMR has recently attracted considerable attention in the field of complex samples analysis. Indeed, the implementation of broadband homonuclear decoupling techniques has allowed us to greatly simplify crowded 1H spectra, yielding singlets for almost every proton site from the analyzed molecules. Pure shift methods have notably shown to be particularly suitable for deciphering mixtures of metabolites in biological samples. Here, we have successfully implemented a new pure shift pulse sequence based on the PSYCHE method, which incorporates a block for solvent suppression that is suitable for metabolomics analysis. The resulting experiment allows us to record ultrahigh-resolution 1D NOESY 1H spectra of biofluids with suppression of the water signal, which is a crucial step for highlighting metabolite mixtures in an aqueous phase. We have successfully recorded pure shift spectra on extracellular media of diffuse large B-cell lymphoma (DLBCL) cells. Despite a lower sensitivity, the resolution of pure shift data was found to be better than that of the standard approach, which provides a more detailed vision of the exo-metabolome. The statistical analyses carried out on the resulting metabolic profiles allow us to successfully highlight several metabolic pathways affected by these drugs. Notably, we show that Kidrolase plays a major role in the metabolic pathways of this DLBCL cell line.


Asunto(s)
Linfoma de Células B Grandes Difuso , Agua , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Espectroscopía de Resonancia Magnética/métodos , Metaboloma , Metabolómica/métodos
4.
Eur J Immunol ; 50(7): 972-985, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32012260

RESUMEN

Several drugs targeting members of the TNF superfamily or TNF receptor superfamily (TNFRSF) are widely used in medicine or are currently being tested in therapeutic trials. However, their mechanism of action remains poorly understood. Here, we explored the effects of TNFRSF co-stimulation on murine Foxp3+ regulatory T cell (Treg) biology, as they are pivotal modulators of immune responses. We show that engagement of TNFR2, 4-1BB, GITR, and DR3, but not OX40, increases Treg proliferation and survival. Triggering these TNFRSF in Tregs induces similar changes in gene expression patterns, suggesting that they engage common signal transduction pathways. Among them, we identified a major role of canonical NF-κB. Importantly, TNFRSF co-stimulation improves the ability of Tregs to suppress colitis. Our data demonstrate that stimulation of discrete TNFRSF members enhances Treg activation and function through a shared mechanism. Consequently, therapeutic effects of drugs targeting TNFRSF or their ligands may be mediated by their effect on Tregs.


Asunto(s)
Activación de Linfocitos , FN-kappa B/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Animales , Ratones , Ratones Noqueados , FN-kappa B/genética , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal/genética , Linfocitos T Reguladores/citología
5.
J Immunol ; 198(4): 1423-1428, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28093523

RESUMEN

Hypoxia upregulates the core pluripotency factors NANOG, SOX2, and OCT4, associated with tumor aggressiveness and resistance to conventional anticancer treatments. We have previously reported that hypoxia-induced NANOG contributed in vitro to tumor cell resistance to autologous-specific CTL and in vivo to the in situ recruitment of immune-suppressive cells. In this study, we investigated the mechanisms underlying NANOG-mediated tumor cell resistance to specific lysis under hypoxia. We demonstrated the tumor-promoting effect of hypoxia on tumor initiation into immunodeficient mice using human non-small lung carcinoma cells. We next showed a link between NANOG and autophagy activation under hypoxia because inhibition of NANOG decreased autophagy in tumor cells. Chromatin immunoprecipitation and luciferase reporter assays revealed a direct binding of NANOG to a transcriptionally active site in a BNIP3L enhancer sequence. These data establish a new link between the pluripotency factor NANOG and autophagy involved in resistance to CTL under hypoxia.


Asunto(s)
Autofagia , Hipoxia de la Célula , Elementos de Facilitación Genéticos , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Proteína Homeótica Nanog/metabolismo , Regiones Promotoras Genéticas , Animales , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Interferencia de ARN , Regulación hacia Arriba
6.
J Chem Inf Model ; 57(2): 223-233, 2017 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-28004927

RESUMEN

NF-κB is a major transcription factor whose activation is triggered through two main activation pathways: the canonical pathway involving disruption of IκB-α/NF-κB complexes and the alternative pathway whose activation relies on the inducible proteolysis of the inhibitory protein p100. One central step controlling p100 processing consists in the interaction of the E3 ubiquitin ligase ß-TrCP with p100, thereby leading to its ubiquitinylation and subsequent either complete degradation or partial proteolysis by the proteasome. However, the interaction mechanism between p100 and ß-TrCP is still poorly defined. In this work, a diphosphorylated 21-mer p100 peptide model containing the phosphodegron motif was used to characterize the interaction with ß-TrCP by NMR. In parallel, docking simulations were performed in order to obtain a model of the 21P-p100/ß-TrCP complex. Saturation transfer difference (STD) experiments were performed in order to highlight the residues of p100 involved in the interaction with the ß-TrCP protein. These results highlighted the importance of pSer865 and pSer869 residues in the interaction with ß-TrCP and particularly the Tyr867 that fits inside the hydrophobe ß-TrCP cavity with the Arg474 guanidinium group. Four other arginines, Arg285, Arg410, Arg431, and Arg521, were found essential in the stabilization of p100 on the ß-TrCP surface. Importantly, the requirement for these five arginine residues of ß-TrCP for the interaction with p100 was further confirmed in vivo, thereby validating the docking model through a biological approach.


Asunto(s)
Simulación del Acoplamiento Molecular , Subunidad p52 de NF-kappa B/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Secuencia de Aminoácidos , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Mutación , Subunidad p52 de NF-kappa B/química , Unión Proteica , Conformación Proteica , Proteínas con Repetición de beta-Transducina/química , Proteínas con Repetición de beta-Transducina/genética
7.
Proc Natl Acad Sci U S A ; 111(41): 14794-9, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25267645

RESUMEN

TNFα is a potent cytokine that plays a critical role in numerous cellular processes, particularly immune and inflammatory responses, programmed cell death, angiogenesis, and cell migration. Thus, understanding the molecular mechanisms that mediate TNFα-induced cellular responses is a crucial issue. It is generally accepted that global DNA binding activity of the NF-κB avian reticuloendotheliosis viral (v-rel) oncogene related B (RelB) subunit is not induced upon TNFα treatment in fibroblasts, despite its TNFα-induced nuclear accumulation. Here, we demonstrate that RelB plays a critical role in promoting fibroblast migration upon prolonged TNFα treatment. We identified the two kinases IκB kinase α (IKKα) and IκB kinase ß (IKKß) as RelB interacting partners whose activation by TNFα promotes RelB phosphorylation at serine 472. Once phosphorylated on serine 472, nuclear RelB dissociates from its interaction with the inhibitory protein IκBα and binds to the promoter of critical migration-associated genes, such as the matrix metallopeptidase 3 (MMP3). Further, we show that RelB serine 472 phosphorylation status controls MMP3 expression and promigration activity downstream of TNF receptors. Our findings provide new insights into the regulation of RelB activity and reveal a novel link between selective NF-κB target gene expression and cellular response in response to TNFα.


Asunto(s)
Movimiento Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Quinasa I-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Transcripción ReIB/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Fibroblastos/efectos de los fármacos , Células HEK293 , Humanos , Proteínas I-kappa B/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Inhibidor NF-kappaB alfa , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
8.
Blood ; 123(4): 509-19, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24184684

RESUMEN

Loss of hematopoietic stem cell (HSC) function and increased risk of developing hematopoietic malignancies are severe and concerning complications of anticancer radiotherapy and chemotherapy. We have previously shown that thrombopoietin (TPO), a critical HSC regulator, ensures HSC chromosomal integrity and function in response to γ-irradiation by regulating their DNA-damage response. TPO directly affects the double-strand break (DSB) repair machinery through increased DNA-protein kinase (DNA-PK) phosphorylation and nonhomologous end-joining (NHEJ) repair efficiency and fidelity. This effect is not shared by other HSC growth factors, suggesting that TPO triggers a specific signal in HSCs facilitating DNA-PK activation upon DNA damage. The discovery of these unique signaling pathways will provide a means of enhancing TPO-desirable effects on HSCs and improving the safety of anticancer DNA agents. We show here that TPO specifically triggers Erk and nuclear factor κB (NF-κB) pathways in mouse hematopoietic stem and progenitor cells (HSPCs). Both of these pathways are required for a TPO-mediated increase in DSB repair. They cooperate to induce and activate the early stress-response gene, Iex-1 (ier3), upon DNA damage. Iex-1 forms a complex with pERK and the catalytic subunit of DNA-PK, which is necessary and sufficient to promote TPO-increased DNA-PK activation and NHEJ DSB repair in both mouse and human HSPCs.


Asunto(s)
Reparación del ADN , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica , Células Madre Hematopoyéticas/citología , Proteínas Inmediatas-Precoces/metabolismo , FN-kappa B/metabolismo , Trombopoyetina/metabolismo , Transporte Activo de Núcleo Celular , Animales , Antineoplásicos/química , Dominio Catalítico , Roturas del ADN de Doble Cadena , Daño del ADN , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Transducción de Señal , Células Madre/citología
9.
Blood ; 122(23): 3713-22, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-24089328

RESUMEN

Monogenic interleukin-10 (IL-10) and IL-10 receptor (IL-10R) deficiencies cause very early onset severe inflammatory bowel disease. Here, we report that 5 patients with an IL-10R1 (n = 1) or IL-10R2 (n = 4) deficiency developed B-cell non-Hodgkin lymphoma between the ages of 5 and 6 years (which was recurrent in 1 patient). These lymphomas had some of the characteristics of diffuse large B-cell lymphomas and contained monoclonal, Epstein-Barr virus-negative germinal center B cells. The tumors displayed a remarkably homogeneous signature, with original activation of the nuclear factor κB pathway and a decrease in intratumor T-cell infiltration. Hence, IL-10R deficiency is associated with a high risk of developing B-cell lymphoma. Our results revealed an unexpected role of the IL-10R pathway in lymphomagenesis.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-10/deficiencia , Subunidad alfa del Receptor de Interleucina-10/genética , Subunidad beta del Receptor de Interleucina-10/deficiencia , Subunidad beta del Receptor de Interleucina-10/genética , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Adolescente , Edad de Inicio , Niño , Preescolar , Femenino , Perfilación de la Expresión Génica , Genes rel , Predisposición Genética a la Enfermedad , Centro Germinal/inmunología , Centro Germinal/patología , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-10/metabolismo , Linfoma de Células B/etiología , Linfoma de Células B Grandes Difuso/etiología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Masculino , Mutación , FN-kappa B/metabolismo , Linaje , Transducción de Señal
10.
J Immunol ; 191(12): 5802-6, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24227785

RESUMEN

Emerging evidence suggests a link between tumor hypoxia and immune suppression. In this study, we investigated the role of hypoxia-induced Nanog, a stemness-associated transcription factor, in immune suppression. We observed that hypoxia-induced Nanog correlated with the acquisition of stem cell-like properties in B16-F10 cells. We further show that Nanog was selectively induced in hypoxic areas of B16-F10 tumors. Stable short hairpin RNA-mediated depletion of Nanog, combined with melanocyte differentiation Ag tyrosinase-related protein-2 peptide-based vaccination, resulted in complete inhibition of B16-F10 tumor growth. Nanog targeting significantly reduced immunosuppressive cells (regulatory T cells and macrophages) and increased CD8(+) T effector cells in tumor bed in part by modulating TGF-ß1 production. Additionally, Nanog regulated TGF-ß1 under hypoxia by directly binding the TGF-ß1 proximal promoter. Collectively, our data establish a novel functional link between hypoxia-induced Nanog and TGF-ß1 regulation and point to a major role of Nanog in hypoxia-driven immunosuppression.


Asunto(s)
Hipoxia de la Célula/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/fisiología , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/inmunología , Melanoma Experimental/inmunología , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/citología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/fisiología , Escape del Tumor/inmunología , Animales , Línea Celular Tumoral , Terapia Genética , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Inmunoterapia , Oxidorreductasas Intramoleculares/inmunología , Linfopoyesis , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Proteína Homeótica Nanog , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/inmunología , Fragmentos de Péptidos/inmunología , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño , Esferoides Celulares , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Escape del Tumor/genética , Microambiente Tumoral , Regulación hacia Arriba , Vacunación
11.
EMBO J ; 29(3): 619-31, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19959994

RESUMEN

In response to stress, cells start transcriptional and transcription-independent programs that can lead to adaptation or death. Here, we show that multiple inducers of autophagy, including nutrient depletion, trigger the activation of the IKK (IkappaB kinase) complex that is best known for its essential role in the activation of the transcription factor NF-kappaB by stress. Constitutively active IKK subunits stimulated autophagy and transduced multiple signals that operate in starvation-induced autophagy, including the phosphorylation of AMPK and JNK1. Genetic inhibition of the nuclear translocation of NF-kappaB or ablation of the p65/RelA NF-kappaB subunit failed to suppress IKK-induced autophagy, indicating that IKK can promote the autophagic pathway in an NF-kappaB-independent manner. In murine and human cells, knockout and/or knockdown of IKK subunits (but not that of p65) prevented the induction of autophagy in response to multiple stimuli. Moreover, the knockout of IKK-beta suppressed the activation of autophagy by food deprivation or rapamycin injections in vivo, in mice. Altogether, these results indicate that IKK has a cardinal role in the stimulation of autophagy by physiological and pharmacological stimuli.


Asunto(s)
Autofagia/fisiología , Quinasa I-kappa B/fisiología , Animales , Autofagia/genética , Células Cultivadas , Células HeLa , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , FN-kappa B/genética , FN-kappa B/metabolismo , Células 3T3 NIH , Transducción de Señal/fisiología
12.
Front Immunol ; 15: 1379777, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38504985

RESUMEN

CD8+ T cells are critical mediators of pathogen clearance and anti-tumor immunity. Although signaling pathways leading to the activation of NF-κB transcription factors have crucial functions in the regulation of immune responses, the CD8+ T cell-autonomous roles of the different NF-κB subunits, are still unresolved. Here, we investigated the function of the ubiquitously expressed transcription factor RelA in CD8+ T-cell biology using a novel mouse model and gene-edited human cells. We found that CD8+ T cell-specific ablation of RelA markedly altered the transcriptome of ex vivo stimulated cells, but maintained the proliferative capacity of both mouse and human cells. In contrast, in vivo experiments showed that RelA deficiency did not affect the CD8+ T-cell response to acute viral infection or transplanted tumors. Our data suggest that in CD8+ T cells, RelA is dispensable for their protective activity in pathological contexts.


Asunto(s)
Neoplasias , Virosis , Animales , Humanos , Ratones , Linfocitos T CD8-positivos/metabolismo , Neoplasias/metabolismo , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Virosis/metabolismo
13.
medRxiv ; 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38798321

RESUMEN

IKKα, encoded by CHUK , is crucial in the non-canonical NF-κB pathway and part of the IKK complex activating the canonical pathway alongside IKKß. Absence of IKKα cause fetal encasement syndrome in human, fatal in utero, while an impaired IKKα-NIK interaction was reported in a single patient and cause combined immunodeficiency. Here, we describe compound heterozygous variants in the kinase domain of IKKα in a female patient with hypogammaglobulinemia, recurrent lung infections, and Hay-Wells syndrome-like features. We showed that both variants were loss-of-function. Non-canonical NF-κB activation was profoundly diminished in stromal and immune cells while the canonical pathway was partially impaired. Reintroducing wild-type CHUK restored non-canonical NF-κB activation. The patient had neutralizing autoantibodies against type I IFN, akin to non-canonical NF-κB pathway deficiencies. Thus, this is the first case of bi-allelic CHUK mutations disrupting IKKα kinase function, broadening non-canonical NF-κB defect understanding and suggesting IKKα's role in canonical NF-κB target gene expression in human.

14.
Blood ; 117(6): 1917-27, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21139082

RESUMEN

Chronic lymphocytic leukemia (CLL) is characterized by a clonal accumulation of mature neoplastic B cells that are resistant to apoptosis. Aiolos, a member of the Ikaros family of zinc-finger transcription factors, plays an important role in the control of mature B lymphocyte differentiation and maturation. In this study, we showed that Aiolos expression is up-regulated in B-CLL cells. This overexpression does not implicate isoform imbalance or disturb Aiolos subcellular localization. The chromatin status at the Aiolos promoter in CLL is defined by the demethylation of DNA and an enrichment of euchromatin associated histone markers, such as the dimethylation of the lysine 4 on histone H3. These epigenetic modifications should allow its upstream effectors, such as nuclear factor-κB, constitutively activated in CLL, to gain access to promoter, resulting up-regulation of Aiolos. To determine the consequences of Aiolos deregulation in CLL, we analyzed the effects of Aiolos overexpression or down-regulation on apoptosis. Aiolos is involved in cell survival by regulating the expression of some Bcl-2 family members. Our results strongly suggest that Aiolos deregulation by epigenetic modifications may be a hallmark of CLL.


Asunto(s)
Epigénesis Genética , Factor de Transcripción Ikaros/genética , Leucemia Linfocítica Crónica de Células B/genética , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Apoptosis/fisiología , Linfocitos B/metabolismo , Linfocitos B/patología , Secuencia de Bases , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Cromatina/genética , Cromatina/metabolismo , Islas de CpG , Metilación de ADN , Cartilla de ADN/genética , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Factor de Transcripción Ikaros/antagonistas & inhibidores , Factor de Transcripción Ikaros/metabolismo , Técnicas In Vitro , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Persona de Mediana Edad , Modelos Biológicos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Fracciones Subcelulares/metabolismo
15.
Nat Cancer ; 4(3): 344-364, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36732635

RESUMEN

Metabolic rewiring is often considered an adaptive pressure limiting metastasis formation; however, some nutrients available at distant organs may inherently promote metastatic growth. We find that the lung and liver are lipid-rich environments. Moreover, we observe that pre-metastatic niche formation increases palmitate availability only in the lung, whereas a high-fat diet increases it in both organs. In line with this, targeting palmitate processing inhibits breast cancer-derived lung metastasis formation. Mechanistically, breast cancer cells use palmitate to synthesize acetyl-CoA in a carnitine palmitoyltransferase 1a-dependent manner. Concomitantly, lysine acetyltransferase 2a expression is promoted by palmitate, linking the available acetyl-CoA to the acetylation of the nuclear factor-kappaB subunit p65. Deletion of lysine acetyltransferase 2a or carnitine palmitoyltransferase 1a reduces metastasis formation in lean and high-fat diet mice, and lung and liver metastases from patients with breast cancer show coexpression of both proteins. In conclusion, palmitate-rich environments foster metastases growth by increasing p65 acetylation, resulting in a pro-metastatic nuclear factor-kappaB signaling.


Asunto(s)
Lisina Acetiltransferasas , FN-kappa B , Ratones , Animales , FN-kappa B/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Acetilación , Acetilcoenzima A/metabolismo , Palmitatos , Lisina Acetiltransferasas/metabolismo
16.
J Biol Chem ; 286(37): 32277-88, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21784860

RESUMEN

NF-κB transcription factors are pivotal players in controlling inflammatory and immune responses, as well as cell proliferation and apoptosis. Aberrant regulation of NF-κB and the signaling pathways that regulate its activity have been involved in various pathologies, particularly cancers, as well as inflammatory and autoimmune diseases. NF-κB activation is tightly regulated by the IκB kinase (IKK) complex, which is composed of two catalytic subunits IKKα and IKKß, and a regulatory subunit IKKγ/NEMO. Although IKKα and IKKß share structural similarities, IKKα has been shown to have distinct biological functions. However, the molecular mechanisms that modulate IKKα activity have not yet been fully elucidated. To understand better the regulation of IKKα activity, we purified IKKα-associated proteins and identified ABIN-2. Here, we demonstrate that IKKα and IKKß both interact with ABIN-2 and impair its constitutive degradation by the proteasome. Nonetheless, ABIN-2 enhances IKKα- but not IKKß-mediated NF-κB activation by specifically inducing IKKα autophosphorylation and kinase activity. Furthermore, we found that ABIN-2 serine 146 is critical for the ABIN-2-dependent IKKα transcriptional up-regulation of specific NF-κB target genes. These results imply that ABIN-2 acts as a positive regulator of NF-κB-dependent transcription by activating IKKα.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Transcripción Genética/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células HEK293 , Células HeLa , Humanos , Quinasa I-kappa B/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , FN-kappa B/genética , Fosforilación/fisiología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Regulación hacia Arriba/fisiología
17.
Blood ; 116(20): 4240-50, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-20671123

RESUMEN

The FOXO transcription factors are involved in multiple signaling pathways and have tumor-suppressor functions. In acute myeloid leukemia (AML), deregulation of oncogenic kinases, including Akt, extra-signal-regulated kinase, or IκB kinase, is frequently observed, which may potentially inactivate FOXO activity. We therefore investigated the mechanism underlying the regulation of FOXO3a, the only FOXO protein constantly expressed in AML blast cells. We show that in both primary AML samples and in a MV4-11/FOXO3a-GFP cell line, FOXO3a is in a constant inactive state due to its cytoplasmic localization, and that neither PI3K/Akt nor extra-signal-regulated kinase-specific inhibition resulted in its nuclear translocation. In contrast, the anti-Nemo peptide that specifically inhibits IKK activity was found to induce FOXO3a nuclear localization in leukemic cells. Furthermore, an IKK-insensitive FOXO3a protein mutated at S644 translocated into the nucleus and activated the transcription of the Fas-L and p21(Cip1) genes. This, in turn, inhibited leukemic cell proliferation and induced apoptosis. These results thus indicate that IKK activity maintains FOXO3a in the cytoplasm and establishes an important role of FOXO3a inactivation in the proliferation and survival of AML cells. The restoration of FOXO3a activity by interacting with its subcellular distribution may thus represent a new attractive therapeutic strategy for AML.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Quinasa I-kappa B/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apoptosis/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteína Forkhead Box O3 , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Mutantes/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Serina/metabolismo , Relación Estructura-Actividad
18.
Curr Top Microbiol Immunol ; 349: 97-114, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21267708

RESUMEN

NF-κB transcription factors are critical regulators of many biological processes such as innate and adaptive immune responses, inflammation, cell proliferation and programmed cell death. This versatility necessitates a highly complex and tightly coordinated control of the signaling pathways leading to their activation. Here, we review the role of proteolysis in the regulation of NF-κB activity, more specifically the contribution of the well-known ubiquitin-proteasome system and the involvement of proteolytic activity of caspases and calpains.


Asunto(s)
FN-kappa B/fisiología , Animales , Calpaína/fisiología , Caspasas/fisiología , Humanos , Quinasa I-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Transducción de Señal
19.
Biomedicines ; 10(2)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35203557

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma in adults and reveals distinct genetic and metabolic signatures. NF-κB transcription factor family is involved in diverse biological processes enabling tumor development and resistance to anticancer-therapy through activation of its two main pathways, the canonical and the alternative NF-κB pathways, the main actor of the latter being the RelB NF-kB subunit. RelB DNA binding activity is frequently activated in DLBCL patients and cell lines. RelB activation defines a new DLBCL subgroup with dismal outcome upon immunochemotherapy, and RelB confers DLBCL cell resistance to DNA damage. However, whether RelB can impact on DLBCL cell metabolism and survival upon metabolic stress is unknown. Here, we reveal that RelB controls DLBCL oxidative energetic metabolism. Accordingly, RelB inhibition reduce DLBCL mitochondrial ATP production, and sensitizes DLBCL cells to apoptosis induced by Metformin and L-asparaginase (®Kidrolase), two FDA approved antimetabolic drugs targeting mitochondrial metabolism. RelB also confers DLBCL cell resistance to glutamine deprivation, an essential amino acid that feeds the TCA cycle. Taken together, our findings uncover a new role for RelB in the regulation of DLBCL cell metabolism and DLBCL cell survival upon metabolic stress.

20.
Biomedicines ; 10(2)2022 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-35203450

RESUMEN

Gastric cancer (GC) is one of the major causes of cancer-related mortality worldwide. The vast majority of GC cases are adenocarcinomas including intestinal and diffuse GC. The incidence of diffuse GCs, often associated with poor overall survival, has constantly increased in USA and Europe The molecular basis of diffuse GC aggressivity remains unclear. Using mRNA from diffuse and intestinal GC tumor samples of a Western cohort, this study reports the expression level of the immunomodulatory aryl-hydrocarbon receptor (AhR), and genes involved in immune suppression (PD1, PD-L1, PD-L2) and the early steps of tryptophan metabolism (IDO1, IDO2, TDO2). Strongly increased expression of IDO1 (p < 0.001) and PD1 (p < 0.003) was observed in the intestinal sub-type. The highest expression of IDO1 and PDL1 correlated with early clinical stage and absence of lymphatic invasion (×25 p = 0.004, ×3 p = 0.04, respectively). Our results suggest that kynurenine, produced by tryptophan catabolism, and AhR activation play a central role in creating an immunosuppressive environment. Correspondingly, as compared to intestinal GCs, expression levels of IDO1-TDO2 and PD-L1 were less prominent in diffuse GCs which also had less infiltration of immune cells, suggesting an inactive immune response in the advanced diffuse GC. Confirmation of these patterns of gene expression will require a larger cohort of early and advanced stages of diffuse GC samples.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA