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










Base de datos
Intervalo de año de publicación
1.
Sci Immunol ; 9(92): eadi0042, 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38306418

RESUMEN

Familial hemophagocytic lymphohistiocytosis (FHL) is an inherited, often fatal immune deficiency characterized by severe systemic hyperinflammation. Although allogeneic bone marrow transplantation can be curative, more effective therapies are urgently needed. FHL is caused by inactivating mutations in proteins that regulate cellular immunity. Here, we used an adeno-associated virus-based CRISPR-Cas9 system with an inhibitor of nonhomologous end joining to repair such mutations in potentially long-lived T cells ex vivo. Repaired CD8 memory T cells efficiently cured lethal hyperinflammation in a mouse model of Epstein-Barr virus-triggered FHL2, a subtype caused by perforin-1 (Prf1) deficiency. Furthermore, repair of PRF1 and Munc13-4 (UNC13D)-whose deficiency causes the FHL subtype FHL3-in mutant memory T cells from two critically ill patients with FHL restored T cell cytotoxicity. These results provide a starting point for the treatment of genetic T cell immune dysregulation syndromes with repaired autologous T cells.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Linfohistiocitosis Hemofagocítica , Animales , Ratones , Humanos , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/terapia , Sistemas CRISPR-Cas , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/terapia , Células T de Memoria , Herpesvirus Humano 4 , Proteínas de la Membrana/genética
2.
Nat Commun ; 15(1): 414, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38195569

RESUMEN

Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) drives viral B cell transformation and oncogenesis. LMP1's transforming activity depends on its C-terminal activation region 2 (CTAR2), which induces NF-κB and JNK by engaging TNF receptor-associated factor 6 (TRAF6). The mechanism of TRAF6 recruitment to LMP1 and its role in LMP1 signalling remains elusive. Here we demonstrate that TRAF6 interacts directly with a viral TRAF6 binding motif within CTAR2. Functional and NMR studies supported by molecular modeling provide insight into the architecture of the LMP1-TRAF6 complex, which differs from that of CD40-TRAF6. The direct recruitment of TRAF6 to LMP1 is essential for NF-κB activation by CTAR2 and the survival of LMP1-driven lymphoma. Disruption of the LMP1-TRAF6 complex by inhibitory peptides interferes with the survival of EBV-transformed B cells. In this work, we identify LMP1-TRAF6 as a critical virus-host interface and validate this interaction as a potential therapeutic target in EBV-associated cancer.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Linfoma de Células B , Humanos , Herpesvirus Humano 4 , Factor 6 Asociado a Receptor de TNF , Infecciones por Virus de Epstein-Barr/complicaciones , FN-kappa B , Transformación Celular Neoplásica , Transformación Celular Viral
3.
Cancer Cell ; 41(7): 1327-1344.e10, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37352862

RESUMEN

Gastric neuroendocrine carcinomas (G-NEC) are aggressive malignancies with poorly understood biology and a lack of disease models. Here, we use genome sequencing to characterize the genomic landscapes of human G-NEC and its histologic variants. We identify global and subtype-specific alterations and expose hitherto unappreciated gains of MYC family members in a large part of cases. Genetic engineering and lineage tracing in mice delineate a model of G-NEC evolution, which defines MYC as a critical driver and positions the cancer cell of origin to the neuroendocrine compartment. MYC-driven tumors have pronounced metastatic competence and display defined signaling addictions, as revealed by large-scale genetic and pharmacologic screening of cell lines and organoid resources. We create global maps of G-NEC dependencies, highlight critical vulnerabilities, and validate therapeutic targets, including candidates for clinical drug repurposing. Our study gives comprehensive insights into G-NEC biology.


Asunto(s)
Carcinoma Neuroendocrino , Tumores Neuroendocrinos , Neoplasias Gástricas , Humanos , Animales , Ratones , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Modelos Moleculares , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/genética
4.
Nat Immunol ; 24(2): 295-308, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36604548

RESUMEN

It has been shown that innate immune responses can adopt adaptive properties such as memory. Whether T cells utilize innate immune signaling pathways to diversify their repertoire of effector functions is unknown. Gasdermin E (GSDME) is a membrane pore-forming molecule that has been shown to execute pyroptotic cell death and thus to serve as a potential cancer checkpoint. In the present study, we show that human T cells express GSDME and, surprisingly, that this expression is associated with durable viability and repurposed for the release of the alarmin interleukin (IL)-1α. This property was restricted to a subset of human helper type 17 T cells with specificity for Candida albicans and regulated by a T cell-intrinsic NLRP3 inflammasome, and its engagement of a proteolytic cascade of successive caspase-8, caspase-3 and GSDME cleavage after T cell receptor stimulation and calcium-licensed calpain maturation of the pro-IL-1α form. Our results indicate that GSDME pore formation in T cells is a mechanism of unconventional cytokine release. This finding diversifies our understanding of the functional repertoire and mechanistic equipment of T cells and has implications for antifungal immunity.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Células Th17 , Humanos , Caspasa 1/metabolismo , Gasderminas , Inmunidad Innata , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis
5.
Front Immunol ; 14: 1331730, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38169736

RESUMEN

Introduction: Epstein-Barr virus (EBV) infection in humans is associated with a wide range of diseases including malignancies of different origins, most prominently B cells. Several EBV latent genes are thought to act together in B cell immortalization, but a minimal set of EBV genes sufficient for transformation remains to be identified. Methods: Here, we addressed this question by transducing human peripheral B cells from EBV-negative donors with retrovirus expressing the latent EBV genes encoding Latent Membrane Protein (LMP) 1 and 2A and Epstein-Barr Nuclear Antigen (EBNA) 2. Results: LMP1 together with EBNA2, but not LMP1 alone or in combination with LMP2A was able to transform human primary B cells. LMP1/EBNA2-immortalized cell lines shared surface markers with EBV-transformed lymphoblastoid cell lines (LCLs). They showed sustained growth for more than 60 days, albeit at a lower growth rate than EBV-transformed LCLs. LMP1/EBNA2-immortalized cell lines generated tumors when transplanted subcutaneously into severely immunodeficient NOG mice. Conclusion: Our results identify a minimal set of EBV proteins sufficient for B cell transformation.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Humanos , Animales , Ratones , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/genética , Proteínas Virales/metabolismo , Linfocitos B , Transformación Celular Neoplásica/genética
6.
Front Immunol ; 11: 602868, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33343574

RESUMEN

A highly recurrent somatic L265P mutation in the TIR domain of the signaling adapter MYD88 constitutively activates NF-κB. It occurs in nearly all human patients with Waldenström's macroglobulinemia (WM), a B cell malignancy caused by IgM-expressing cells. Here, we introduced an inducible leucine to proline point mutation into the mouse Myd88 locus, at the orthologous position L252P. When the mutation was introduced early during B cell development, B cells developed normally. However, IgM-expressing plasma cells accumulated with age in spleen and bone, leading to more than 20-fold elevated serum IgM titers. When introduced into germinal center B cells in the context of an immunization, the Myd88L252P mutation caused prolonged persistence of antigen-specific serum IgM and elevated numbers of antigen-specific IgM plasma cells. Myd88L252P-expressing B cells switched normally, but plasma cells expressing other immunoglobulin isotypes did not increase in numbers, implying that IgM expression may be required for the observed cellular expansion. In order to test whether the Myd88L252P mutation can cause clonal expansions, we introduced it into a small fraction of CD19-positive B cells. In this scenario, five out of five mice developed monoclonal IgM serum paraproteins accompanied by an expansion of clonally related plasma cells that expressed mostly hypermutated VDJ regions. Taken together, our data suggest that the Myd88L252P mutation is sufficient to promote aberrant survival and expansion of IgM-expressing plasma cells which in turn can cause IgM monoclonal gammopathy of undetermined significance (MGUS), the premalignant condition that precedes WM.


Asunto(s)
Linfocitos B/metabolismo , Marcación de Gen , Inmunoglobulina M/sangre , Gammopatía Monoclonal de Relevancia Indeterminada/genética , Factor 88 de Diferenciación Mieloide/genética , Células Plasmáticas/metabolismo , Mutación Puntual , Animales , Linfocitos B/inmunología , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Predisposición Genética a la Enfermedad , Inmunoglobulina M/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Gammopatía Monoclonal de Relevancia Indeterminada/sangre , Gammopatía Monoclonal de Relevancia Indeterminada/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Paraproteínas/metabolismo , Fenotipo , Células Plasmáticas/inmunología
7.
Proc Natl Acad Sci U S A ; 117(25): 14421-14432, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32522871

RESUMEN

Epstein-Barr virus (EBV) is a B cell transforming virus that causes B cell malignancies under conditions of immune suppression. EBV orchestrates B cell transformation through its latent membrane proteins (LMPs) and Epstein-Barr nuclear antigens (EBNAs). We here identify secondary mutations in mouse B cell lymphomas induced by LMP1, to predict and identify key functions of other EBV genes during transformation. We find aberrant activation of early B cell factor 1 (EBF1) to promote transformation of LMP1-expressing B cells by inhibiting their differentiation to plasma cells. EBV EBNA3A phenocopies EBF1 activities in LMP1-expressing B cells, promoting transformation while inhibiting differentiation. In cells expressing LMP1 together with LMP2A, EBNA3A only promotes lymphomagenesis when the EBNA2 target Myc is also overexpressed. Collectively, our data support a model where proproliferative activities of LMP1, LMP2A, and EBNA2 in combination with EBNA3A-mediated inhibition of terminal plasma cell differentiation critically control EBV-mediated B cell lymphomagenesis.


Asunto(s)
Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/patogenicidad , Linfoma de Células B/patología , Células Plasmáticas/patología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Fibroblastos , Herpesvirus Humano 4/metabolismo , Humanos , Linfoma de Células B/virología , Ratones , Ratones Noqueados , Células Plasmáticas/virología , Cultivo Primario de Células , Transactivadores/genética , Transactivadores/metabolismo , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/metabolismo
8.
Cell Rep ; 28(13): 3510-3522.e5, 2019 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-31553918

RESUMEN

Mutations accumulating in hematopoietic stem and progenitor cells (HSPCs) during development can cause severe hematological disorders. Modeling these mutations in mice is essential for understanding their functional consequences. Here, we describe an efficient CRISPR/Cas9-based system to knock in and repair genes in mouse HSPCs. CRISPR/Cas9 ribonucleoproteins, in combination with recombinant adeno-associated virus (rAAV)-DJ donor templates, led to gene knockin efficiencies of up to 30% in the Lmnb1 and Actb loci of mouse HSPCs in vitro. The targeted HSPCs engraft and reconstitute all immune cell lineages in the recipient mice. Using this approach, we corrected a neomycin-disrupted Rag2 gene. The Rag2-corrected HSPCs restore B and T cell development in vivo, confirming the functionality of the approach. Our method provides an efficient strategy to study gene function in the hematopoietic system and model hematological disorders in vivo, without the need for germline mutagenesis.


Asunto(s)
Sistemas CRISPR-Cas/genética , Técnicas de Sustitución del Gen/métodos , Células Madre Hematopoyéticas/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Ratones
9.
Cell Metab ; 30(3): 539-555.e11, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31257153

RESUMEN

Epstein-Barr virus (EBV) causes Burkitt, Hodgkin, and post-transplant B cell lymphomas. How EBV remodels metabolic pathways to support rapid B cell outgrowth remains largely unknown. To gain insights, primary human B cells were profiled by tandem-mass-tag-based proteomics at rest and at nine time points after infection; >8,000 host and 29 viral proteins were quantified, revealing mitochondrial remodeling and induction of one-carbon (1C) metabolism. EBV-encoded EBNA2 and its target MYC were required for upregulation of the central mitochondrial 1C enzyme MTHFD2, which played key roles in EBV-driven B cell growth and survival. MTHFD2 was critical for maintaining elevated NADPH levels in infected cells, and oxidation of mitochondrial NADPH diminished B cell proliferation. Tracing studies underscored contributions of 1C to nucleotide synthesis, NADPH production, and redox defense. EBV upregulated import and synthesis of serine to augment 1C flux. Our results highlight EBV-induced 1C as a potential therapeutic target and provide a new paradigm for viral onco-metabolism.


Asunto(s)
Aminohidrolasas/metabolismo , Linfocitos B/metabolismo , Linfocitos B/virología , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/metabolismo , Ácido Fólico/metabolismo , Herpesvirus Humano 4/metabolismo , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Enzimas Multifuncionales/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Femenino , Glucólisis , Células HEK293 , Humanos , Activación de Linfocitos , Mitocondrias/metabolismo , NADP/biosíntesis , Oxidación-Reducción , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Serina/biosíntesis
10.
Blood ; 132(25): 2670-2683, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30333121

RESUMEN

Forkhead box class O1 (FOXO1) acts as a tumor suppressor in solid tumors. The oncogenic phosphoinositide-3-kinase (PI3K) pathway suppresses FOXO1 transcriptional activity by enforcing its nuclear exclusion upon AKT-mediated phosphorylation. We show here abundant nuclear expression of FOXO1 in Burkitt lymphoma (BL), a germinal center (GC) B-cell-derived lymphoma whose pathogenesis is linked to PI3K activation. Recurrent FOXO1 mutations, which prevent AKT targeting and lock the transcription factor in the nucleus, are used by BL to circumvent mutual exclusivity between PI3K and FOXO1 activation. Using genome editing in human and mouse lymphomas in which MYC and PI3K cooperate synergistically in tumor development, we demonstrate proproliferative and antiapoptotic activity of FOXO1 in BL and identify its nuclear localization as an oncogenic event in GC B-cell-derived lymphomagenesis.


Asunto(s)
Linfocitos B , Linfoma de Burkitt , Núcleo Celular , Transformación Celular Neoplásica , Proteína Forkhead Box O1 , Centro Germinal , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Linfoma de Burkitt/genética , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Edición Génica , Centro Germinal/metabolismo , Centro Germinal/patología , Humanos , Ratones , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Blood ; 132(9): 924-934, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30002144

RESUMEN

To date, little is known about the interaction between (pre-)malignant B cells and T cells. We generated transgenic mice that allow B cell-specific induction of the oncogene SV40 large T-antigen (TAg) to analyze the role of oncogene-specific T cells during sporadic B-cell lymphoma development. Constitutive TAg expression in CD19-Cre × LoxP-Tag mice resulted in TAg-tolerant CD8+ T cells and development of B-cell lymphomas. In contrast, CD19-CreERT2 × LoxP-Tag mice retained TAg-competent CD8+ T cells at time of oncogene induction and TAg expression in few B cells of adult mice resulted in exceptionally rare lymphoma formation late in life. Increased lymphoma incidence in the absence of TAg-specific T cells suggested T cell-mediated inhibition of lymphoma progression. However, TAg-initiated B cells were not eliminated by T cells and detected long term. Our results demonstrate a failure of the immune system to eradicate lymphoma-initiating B cells, retaining the risk of lymphoma development.


Asunto(s)
Antígenos Transformadores de Poliomavirus/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Linfoma de Células B/inmunología , Animales , Antígenos Transformadores de Poliomavirus/genética , Linfocitos B/patología , Linfocitos T CD8-positivos/patología , Linfoma de Células B/genética , Linfoma de Células B/patología , Ratones , Ratones Noqueados
12.
Proc Natl Acad Sci U S A ; 113(48): 13821-13826, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27856754

RESUMEN

Epstein-Barr Virus (EBV) infects human B cells and drives them into continuous proliferation. Two key viral factors in this process are the latent membrane proteins LMP1 and LMP2A, which mimic constitutively activated CD40 receptor and B-cell receptor signaling, respectively. EBV-infected B cells elicit a powerful T-cell response that clears the infected B cells and leads to life-long immunity. Insufficient immune surveillance of EBV-infected B cells causes life-threatening lymphoproliferative disorders, including mostly germinal center (GC)-derived B-cell lymphomas. We have modeled acute EBV infection of naive and GC B cells in mice through timed expression of LMP1 and LMP2A. Although lethal when induced in all B cells, induction of LMP1 and LMP2A in just a small fraction of naive B cells initiated a phase of rapid B-cell expansion followed by a proliferative T-cell response, clearing the LMP-expressing B cells. Interfering with T-cell activity prevented clearance of LMP-expressing B cells. This was also true for perforin deficiency, which in the human causes a life-threatening EBV-related immunoproliferative syndrome. LMP expression in GC B cells impeded the GC reaction but, upon loss of T-cell surveillance, led to fatal B-cell expansion. Thus, timed expression of LMP1 together with LMP2A in subsets of mouse B cells allows one to study major clinically relevant features of human EBV infection in vivo, opening the way to new therapeutic approaches.


Asunto(s)
Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Proteínas de la Matriz Viral/genética , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Antígenos CD40/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Regulación Viral de la Expresión Génica , Centro Germinal/inmunología , Centro Germinal/metabolismo , Herpesvirus Humano 4/patogenicidad , Humanos , Ratones , Perforina/deficiencia , Perforina/genética , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T/virología , Proteínas de la Matriz Viral/biosíntesis
13.
BMC Biotechnol ; 16: 4, 2016 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-26772810

RESUMEN

BACKGROUND: The CRISPR/Cas9 system is increasingly used for gene inactivation in mouse zygotes, but homology-directed mutagenesis and use of inbred embryos are less established. In particular, Rosa26 knock-in alleles for the insertion of transgenes in a genomic 'safe harbor' site, have not been produced. Here we applied CRISPR/Cas9 for the knock-in of 8-11 kb inserts into Rosa26 of C57BL/6 zygotes. RESULTS: We found that 10-20 % of live pups derived from microinjected zygotes were founder mutants, without apparent off-target effects, and up to 50 % knock-in embryos were recovered upon coinjection of Cas9 mRNA and protein. Using this approach, we established a new mouse line for the Cre/loxP-dependent expression of Cas9. CONCLUSIONS: Altogether, our protocols and resources support the fast and direct generation of new Rosa26 knock-in alleles and of Cas9-mediated in vivo gene editing in the widely used C57BL/6 inbred strain.


Asunto(s)
Sistemas CRISPR-Cas/genética , Técnicas de Sustitución del Gen/métodos , ARN no Traducido/genética , Animales , Clonación Molecular , Embrión de Mamíferos , Ratones , Ratones Endogámicos C57BL , Microinyecciones
14.
Artículo en Inglés | MEDLINE | ID: mdl-24241423

RESUMEN

Epstein-Barr virus (EBV) is a γ herpes virus endemic in humans and transforming human B lymphocytes. It causes a variety of human pathologies ranging from infectious mononucleosis upon acute infection to EBV-driven B-cell lymphomas. In humans, EBV-infected cells are under powerful immune surveillance by T and NK cells. If this immune surveillance is compromised as in immunosuppressed (AIDS- or posttransplantation) patients, the virus can spread from rare, EBV-containing cells and cause life-threatening pathologies. We have found that EBV immune surveillance and lymphomagenesis can be modeled in mice by targeted expression of key EBV proteins in the B-cell lineage. As EBV does not infect mouse B cells and mice have thus not coevolved with the virus, EBV exploits basic modes of the host immune response to optimize its coexistence with the host.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Vigilancia Inmunológica , Enfermedad Aguda , Animales , Linfocitos B/virología , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Linfoma de Células B/inmunología , Ratones , Transducción de Señal , Linfocitos T/inmunología , Proteínas de la Matriz Viral/metabolismo
15.
Autophagy ; 8(2): 265-7, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22361669

RESUMEN

Autophagy allows cells to survive under conditions of nutrient deprivation. We have demonstrated that autophagy inhibitors are synthetically lethal with NFκB inhibitors in B-cell lymphomas because the NFκB pathway promotes survival by increasing glucose import. When NFκB is inhibited in B-cell lymphoma, glucose import decreases and cells become sensitive to perturbations in mitochondrial metabolism and autophagy. Thus, combined inhibition of autophagy and NFκB drives cells into metabolic crisis accelerating cell death.


Asunto(s)
Autofagia , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , FN-kappa B/antagonistas & inhibidores , Transporte Biológico , Membrana Celular/metabolismo , Supervivencia Celular , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Linfocitos/metabolismo , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/enzimología , Modelos Biológicos , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transcripción Genética
16.
Cancer Res ; 71(23): 7291-300, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21987722

RESUMEN

All cancer cells require increased nutrient uptake to support proliferation. In this study, we investigated the signals that govern glucose uptake in B-cell lymphomas and determined that the inhibitor of NF-κB-kinase ß (IKKß) induced glucose transporter-1 (GLUT1) membrane trafficking in both viral and spontaneous B-cell lymphomas. IKKß induced AKT activity, whereas IKKß-driven NF-κB transcription was required for GLUT1 surface localization downstream of AKT. Activated NF-κB promoted AKT-mediated phosphorylation of the GLUT1 regulator, AKT substrate of 160kD (AS160), but was not required for AKT phosphorylation of the mTOR regulator Tuberous Sclerosis 2 (TSC2). In Epstein-Barr virus-transformed B cells, NF-κB inhibition repressed glucose uptake and induced caspase-independent cell death associated with autophagy. After NF-κB inhibition, an alternate carbon source ameliorated both autophagy and cell death, whereas autophagy inhibitors specifically accelerated cell death. Taken together, the results indicate that NF-κB signaling establishes a metabolic program supporting proliferation and apoptosis resistance by driving glucose import.


Asunto(s)
Transportador de Glucosa de Tipo 1/metabolismo , Quinasa I-kappa B/genética , Linfoma de Células B/genética , FN-kappa B/genética , Proteínas Proto-Oncogénicas c-akt/genética , Apoptosis/genética , Autofagia/genética , Caspasas/metabolismo , Muerte Celular/genética , Procesos de Crecimiento Celular/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 1/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/metabolismo , Linfoma de Células B/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/biosíntesis , FN-kappa B/metabolismo , Fosforilación/genética , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo
17.
Proc Natl Acad Sci U S A ; 108(19): 7808-13, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21518914

RESUMEN

EBV nuclear antigen 2 (EBNA2) and EBV nuclear antigen LP (EBNALP) are critical for B-lymphocyte transformation to lymphoblastoid cell lines (LCLs). EBNA2 activates transcription through recombination signal-binding immunoglobulin κJ region (RBPJ), a transcription factor associated with NCoR repressive complexes, and EBNALP is implicated in repressor relocalization. EBNALP coactivation with EBNA2 was found to dominate over NCoR repression. EBNALP associated with NCoR and dismissed NCoR, NCoR and RBPJ, or NCoR, RBPJ, and EBNA2 from matrix-associated deacetylase (MAD) bodies. In non-EBV-infected BJAB B lymphoma cells that stably express EBNA2, EBNALP, or EBNA2 and EBNALP, EBNALP was associated with hairy and enhancer of split 1 (hes1), cd21, cd23, and arginine and glutamate-rich 1 (arglu1) enhancer or promoter DNA and was associated minimally with coding DNA. With the exception of RBPJ at the arglu1 enhancer, NCoR and RBPJ were significantly decreased at enhancer and promoter sites in EBNALP or EBNA2 and EBNALP BJAB cells. EBNA2 DNA association was unaffected by EBNALP, and EBNALP was unaffected by EBNA2. EBNA2 markedly increased RBPJ at enhancer sites without increasing NCoR. EBNALP further increased hes1 and arglu1 RNA levels with EBNA2 but did not further increase cd21 or cd23 RNA levels. EBNALP in which the 45 C-terminal residues critical for transformation and transcriptional activation were deleted associated with NCoR but was deficient in dismissing NCoR from MAD bodies and from enhancer and promoter sites. These data strongly support a model in which EBNA2 association with NCoR-deficient RBPJ enhances transcription and EBNALP dismisses NCoR and RBPJ repressive complexes from enhancers to coactivate hes1 and arglu1 but not cd21 or cd23.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 2 de Receptor Nuclear/metabolismo , Proteínas Virales/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular , Transformación Celular Viral/genética , ADN/genética , ADN/metabolismo , Elementos de Facilitación Genéticos , Antígenos Nucleares del Virus de Epstein-Barr/genética , Regulación Viral de la Expresión Génica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Proteínas de Homeodominio/genética , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/genética , Regiones Promotoras Genéticas , Receptores de Complemento 3d/genética , Receptores de IgE/genética , Factor de Transcripción HES-1 , Proteínas Virales/genética
18.
J Am Chem Soc ; 130(47): 16003-10, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-18975872

RESUMEN

The remarkable stability of glycals under oxidative conditions becomes apparent by their redox data in solution, computed HOMO energies, and behavior on the addition of electrophilic radicals generated in the presence of cerium(IV) ammonium nitrate. Oxidation potentials up to 2.03 V vs ferrocene were obtained, which are exceptionally high for cyclic enol ethers but correlate nicely with the reaction times of the radical reactions. Protecting groups have a strong influence on the oxidation stability and HOMO energies of glycals as E(ox) is shifted from O-silyl over O-benzyl to O-acetyl by more than 500 mV. Interestingly, this effect must be transmitted through sigma-bonds, even up to the para-position of a benzoate group, as verified by a wide variation of remote substituents in the carbohydrate. Favorable interactions of the sigma*-orbital of the adjacent C-O bond with the HOMO of the double bond are proposed as a mechanistic rationale, which might be important for the redox behavior of other allylic systems. Finally, donors and acceptors in the 1-position exert the strongest influence on the oxidation stability, shifting the potentials by almost 1 V and resulting in different follow-up reactions of the cerium(IV)-mediated additions of malonates. It is the remarkable oxidation stability of glycals that makes them valuable building blocks in carbohydrate chemistry.

19.
Chemistry ; 13(36): 10152-67, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17955558

RESUMEN

2-C-malonyl carbohydrates were synthesized in only few steps and high yields by radical additions of malonates to glycals. For the first time, the undesired formation of nitrates was completely suppressed with anhydrous cerium ammonium nitrate (CAN) as oxidizing agent. A coherent explanation for the high stereoselectivities of the additions to gluco-configured glycals was provided by variation of the substituents in the 3-position. We established steric effects for the face selectivity, and electronic effects strongly influence the reactivity of the double bonds. The scope and limitation of transition-metal-mediated radical reactions in the synthesis of 2-C-branched carbohydrates was thoroughly investigated. Thus, unsaturated disaccharides and benzyl-protected glycals were used as substrates for the first time. Finally, the 2-C-malonyl carbohydrates were transformed into various products by decarboxylation, saponification and reduction, which afforded interesting precursors for C-disaccharides. In this paper we describe the syntheses of more than 40 new 2-C-analogues of carbohydrates, which were isolated in high yields in analytically pure form. Therefore, the transition-metal-mediated radical addition of malonates to glycals offers a simple and convenient entry to such important carbohydrate derivatives.


Asunto(s)
Carbohidratos/síntesis química , Malonatos/química , Benceno/química , Carbohidratos/química , Radicales Libres/química , Espectroscopía de Resonancia Magnética , Estructura Molecular , Oxidación-Reducción , Estereoisomerismo
20.
Chem Commun (Camb) ; (22): 2624-5, 2004 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-15543311

RESUMEN

Only three steps are required for the convenient synthesis of 2-C-branched glyco-amino acids from glycals with good yields and stereoselectivities.


Asunto(s)
Glicina/análogos & derivados , Glicina/síntesis química , Glicosilación , Modelos Químicos , Modelos Moleculares , Estructura Molecular , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...