Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell ; 146(6): 918-30, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21925315

RESUMEN

Inhibitors of DNA binding (IDs) antagonize basic-helix-loop-helix (bHLH) transcription factors to inhibit differentiation and maintain stem cell fate. ID ubiquitination and proteasomal degradation occur in differentiated tissues, but IDs in many neoplasms appear to escape degradation. We show that the deubiquitinating enzyme USP1 promotes ID protein stability and stem cell-like characteristics in osteosarcoma. USP1 bound, deubiquitinated, and thereby stabilized ID1, ID2, and ID3. A subset of primary human osteosarcomas coordinately overexpressed USP1 and ID proteins. USP1 knockdown in osteosarcoma cells precipitated ID protein destabilization, cell-cycle arrest, and osteogenic differentiation. Conversely, ectopic USP1 expression in mesenchymal stem cells stabilized ID proteins, inhibited osteoblastic differentiation, and enhanced proliferation. Consistent with USP1 functioning in normal mesenchymal stem cells, USP1-deficient mice were osteopenic. Our observations implicate USP1 in preservation of the stem cell state that characterizes osteosarcoma and identify USP1 as a target for differentiation therapy.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Neoplásicas/citología , Osteosarcoma/patología , Animales , Proteínas de Arabidopsis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteasas Ubiquitina-Específicas , Ubiquitinación
2.
Immunity ; 44(3): 609-621, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26944201

RESUMEN

Targeted inhibition of mitogen-activated protein kinase (MAPK) kinase (MEK) can induce regression of tumors bearing activating mutations in the Ras pathway but rarely leads to tumor eradication. Although combining MEK inhibition with T-cell-directed immunotherapy might lead to more durable efficacy, T cell responses are themselves at least partially dependent on MEK activity. We show here that MEK inhibition did profoundly block naive CD8(+) T cell priming in tumor-bearing mice, but actually increased the number of effector-phenotype antigen-specific CD8(+) T cells within the tumor. MEK inhibition protected tumor-infiltrating CD8(+) T cells from death driven by chronic TCR stimulation while sparing cytotoxic activity. Combining MEK inhibition with anti-programmed death-ligand 1 (PD-L1) resulted in synergistic and durable tumor regression even where either agent alone was only modestly effective. Thus, despite the central importance of the MAP kinase pathway in some aspects of T cell function, MEK-targeted agents can be compatible with T-cell-dependent immunotherapy.


Asunto(s)
Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma/terapia , Neoplasias del Colon/terapia , Inmunoterapia , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis , Azetidinas/administración & dosificación , Azetidinas/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Carcinoma/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Sinergismo Farmacológico , Quimioterapia , Quimioterapia Combinada , Quinasas MAP Reguladas por Señal Extracelular , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Trasplante de Neoplasias , Piperidinas/administración & dosificación , Piperidinas/farmacología
3.
Nature ; 510(7505): 407-411, 2014 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-24919154

RESUMEN

Inhibitors against the p110δ isoform of phosphoinositide-3-OH kinase (PI(3)K) have shown remarkable therapeutic efficacy in some human leukaemias. As p110δ is primarily expressed in leukocytes, drugs against p110δ have not been considered for the treatment of solid tumours. Here we report that p110δ inactivation in mice protects against a broad range of cancers, including non-haematological solid tumours. We demonstrate that p110δ inactivation in regulatory T cells unleashes CD8(+) cytotoxic T cells and induces tumour regression. Thus, p110δ inhibitors can break tumour-induced immune tolerance and should be considered for wider use in oncology.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Neoplasias/enzimología , Neoplasias/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Activación Enzimática/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Ratones , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología
5.
Nature ; 471(7336): 110-4, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21368834

RESUMEN

Microtubules have pivotal roles in fundamental cellular processes and are targets of antitubulin chemotherapeutics. Microtubule-targeted agents such as Taxol and vincristine are prescribed widely for various malignancies, including ovarian and breast adenocarcinomas, non-small-cell lung cancer, leukaemias and lymphomas. These agents arrest cells in mitosis and subsequently induce cell death through poorly defined mechanisms. The strategies that resistant tumour cells use to evade death induced by antitubulin agents are also unclear. Here we show that the pro-survival protein MCL1 (ref. 3) is a crucial regulator of apoptosis triggered by antitubulin chemotherapeutics. During mitotic arrest, MCL1 protein levels decline markedly, through a post-translational mechanism, potentiating cell death. Phosphorylation of MCL1 directs its interaction with the tumour-suppressor protein FBW7, which is the substrate-binding component of a ubiquitin ligase complex. The polyubiquitylation of MCL1 then targets it for proteasomal degradation. The degradation of MCL1 was blocked in patient-derived tumour cells that lacked FBW7 or had loss-of-function mutations in FBW7, conferring resistance to antitubulin agents and promoting chemotherapeutic-induced polyploidy. Additionally, primary tumour samples were enriched for FBW7 inactivation and elevated MCL1 levels, underscoring the prominent roles of these proteins in oncogenesis. Our findings suggest that profiling the FBW7 and MCL1 status of tumours, in terms of protein levels, messenger RNA levels and genetic status, could be useful to predict the response of patients to antitubulin chemotherapeutics.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Moduladores de Tubulina/farmacología , Tubulina (Proteína)/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Resistencia a Antineoplásicos , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Fibroblastos , Humanos , Ratones , Mitosis/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Paclitaxel/farmacología , Farmacogenética , Fosforilación/efectos de los fármacos , Poliploidía , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/deficiencia , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Vincristina/farmacología
6.
Nature ; 463(7277): 103-7, 2010 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20023629

RESUMEN

MCL1 is essential for the survival of stem and progenitor cells of multiple lineages, and is unique among pro-survival BCL2 family members in that it is rapidly turned over through the action of ubiquitin ligases. B- and mantle-cell lymphomas, chronic myeloid leukaemia, and multiple myeloma, however, express abnormally high levels of MCL1, contributing to chemoresistance and disease relapse. The mechanism of MCL1 overexpression in cancer is not well understood. Here we show that the deubiquitinase USP9X stabilizes MCL1 and thereby promotes cell survival. USP9X binds MCL1 and removes the Lys 48-linked polyubiquitin chains that normally mark MCL1 for proteasomal degradation. Increased USP9X expression correlates with increased MCL1 protein in human follicular lymphomas and diffuse large B-cell lymphomas. Moreover, patients with multiple myeloma overexpressing USP9X have a poor prognosis. Knockdown of USP9X increases MCL1 polyubiquitination, which enhances MCL1 turnover and cell killing by the BH3 mimetic ABT-737. These results identify USP9X as a prognostic and therapeutic target, and they show that deubiquitinases may stabilize labile oncoproteins in human malignancies.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Poliubiquitina/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN , Docetaxel , Etopósido/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Semivida , Humanos , Lisina/metabolismo , Ratones , Ratones SCID , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias/diagnóstico , Nitrofenoles/farmacología , Fosforilación/efectos de la radiación , Piperazinas/farmacología , Pronóstico , Unión Proteica/efectos de la radiación , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Interferencia de ARN , Sulfonamidas/farmacología , Taxoides/farmacología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/genética , Ubiquitinación , Rayos Ultravioleta , Ensayos Antitumor por Modelo de Xenoinjerto
7.
MAbs ; 15(1): 2229101, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37639687

RESUMEN

The antibody-drug conjugate (ADC) field has undergone a renaissance, with substantial recent developmental investment and subsequent drug approvals over the past 6 y. In November 2022, ElahereTM became the latest ADC to be approved by the US Food and Drug Administration (FDA). To date, over 260 ADCs have been tested in the clinic against various oncology indications. Here, we review the clinical landscape of ADCs that are currently FDA approved (11), agents currently in clinical trials but not yet approved (164), and candidates discontinued following clinical testing (92). These clinically tested ADCs are further analyzed by their targeting tumor antigen(s), linker, payload choices, and highest clinical stage achieved, highlighting limitations associated with the discontinued drug candidates. Lastly, we discuss biologic engineering modifications preclinically demonstrated to improve the therapeutic index that if incorporated may increase the proportion of molecules that successfully transition to regulatory approval.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Estados Unidos , Inmunoconjugados/uso terapéutico , Anticuerpos Monoclonales , United States Food and Drug Administration
8.
Cancer Cell ; 2(2): 139-48, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12204534

RESUMEN

Epigenetic regulation of gene expression significantly influences cell growth and differentiation. Here we show that epigenetic silencing of Fas determines tumor growth in vivo and apoptotic sensitivity in vitro. In established tumors with epigenetically repressed Fas, restoration of Fas activity either by transfection of fas or treatment with Trichostatin A (TSA), an inhibitor of histone deacetylase, suppresses tumor growth and restores chemosensitivity. The TSA-dependent chemosensitivity and tumor growth control require both tumor Fas and the host NK (natural killer) cell functions. This work demonstrates the importance of epigenetic modification of Fas in tumor progression and immune evasion, and emphasizes the essential interplay between Fas and innate immunity in the control of chemoresistant tumors.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Receptor fas/metabolismo , Animales , Apoptosis , Diferenciación Celular , División Celular/efectos de los fármacos , Transformación Celular Neoplásica , Progresión de la Enfermedad , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Ácidos Hidroxámicos/farmacología , Inmunidad Innata , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias/metabolismo , Neoplasias/patología , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Receptor fas/genética
9.
Dev Cell ; 2(3): 331-41, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11879638

RESUMEN

Cellular differentiation involves transcriptional responses to environmental stimuli. Adipocyte differentiation is inhibited under hypoxic conditions, indicating that oxygen (O(2)) is an important physiological regulator of adipogenesis. Hypoxia inhibits PPAR gamma 2 nuclear hormone receptor transcription, and overexpression of PPAR gamma 2 or C/EBP beta stimulates adipogenesis under hypoxia. Mouse embryonic fibroblasts deficient in hypoxia-inducible transcription factor 1 alpha (HIF-1 alpha) are refractory to hypoxia-mediated inhibition of adipogenesis. The HIF-1-regulated gene DEC1/Stra13, a member of the Drosophila hairy/Enhancer of split transcription repressor family, represses PPAR gamma 2 promoter activation and functions as an effector of hypoxia-mediated inhibition of adipogenesis. These data indicate that an O(2)-sensitive signaling mechanism regulates adipogenesis. Thus, agents that regulate HIF-1 activity or O(2) sensing may be used to inhibit adipogenesis and control obesity.


Asunto(s)
Adipocitos/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Proteínas Nucleares/genética , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Células 3T3 , Adipocitos/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/fisiología , Hipoxia de la Célula/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Proteínas Nucleares/metabolismo , Oxígeno/farmacología
10.
Nat Med ; 19(2): 202-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23291630

RESUMEN

Proteins in the B cell CLL/lymphoma 2 (BCL-2) family are key regulators of the apoptotic process. This family comprises proapoptotic and prosurvival proteins, and shifting the balance toward the latter is an established mechanism whereby cancer cells evade apoptosis. The therapeutic potential of directly inhibiting prosurvival proteins was unveiled with the development of navitoclax, a selective inhibitor of both BCL-2 and BCL-2-like 1 (BCL-X(L)), which has shown clinical efficacy in some BCL-2-dependent hematological cancers. However, concomitant on-target thrombocytopenia caused by BCL-X(L) inhibition limits the efficacy achievable with this agent. Here we report the re-engineering of navitoclax to create a highly potent, orally bioavailable and BCL-2-selective inhibitor, ABT-199. This compound inhibits the growth of BCL-2-dependent tumors in vivo and spares human platelets. A single dose of ABT-199 in three patients with refractory chronic lymphocytic leukemia resulted in tumor lysis within 24 h. These data indicate that selective pharmacological inhibition of BCL-2 shows promise for the treatment of BCL-2-dependent hematological cancers.


Asunto(s)
Antineoplásicos/farmacología , Plaquetas/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas/farmacología , Compuestos de Anilina/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Perros , Femenino , Células HeLa , Humanos , Ratones , Ratones SCID , Proteínas Proto-Oncogénicas c-bcl-2/química , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína bcl-X/antagonistas & inhibidores
11.
Sci Signal ; 5(216): ra22, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22434933

RESUMEN

Tumor necrosis factor (TNF) family members are essential for the development and proper functioning of the immune system. TNF receptor (TNFR) signaling is mediated through the assembly of protein signaling complexes that activate the nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in a ubiquitin-dependent manner. The cellular inhibitor of apoptosis (c-IAP) proteins c-IAP1 and c-IAP2 are E3 ubiquitin ligases that are recruited to TNFR signaling complexes through their constitutive association with the adaptor protein TNFR-associated factor 2 (TRAF2). We demonstrated that c-IAP1 and c-IAP2 were required for canonical activation of NF-κB and MAPK by members of the TNFR family. c-IAPs were required for the recruitment of inhibitor of κB kinase ß (IKKß), the IKK regulatory subunit NF-κB essential modulator (NEMO), and RBCK1/Hoil1-interacting protein (HOIP) to TNFR signaling complexes and the induction of gene expression by TNF family members. In contrast, TNFRs that stimulated the noncanonical NF-κB pathway triggered translocation of c-IAPs, TRAF2, and TRAF3 from the cytosol to membrane fractions, which led to their proteasomal and lysosomal degradation. Finally, we established that signaling by B cell-activating factor receptor 3 induced the cytosolic depletion of TRAF3, which enabled noncanonical NF-κB activation. These results define c-IAP proteins as critical regulators of the activation of NF-κB and MAPK signaling pathways by members of the TNFR superfamily.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/inmunología , Factores de Necrosis Tumoral/metabolismo , Western Blotting , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Silenciador del Gen , Humanos , Quinasa I-kappa B/metabolismo , Proteínas Inhibidoras de la Apoptosis/inmunología , Transporte de Proteínas , ARN Interferente Pequeño/genética , Receptores de Interleucina-4/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Factores de Necrosis Tumoral/inmunología , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas
12.
Mol Cancer Ther ; 10(12): 2340-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21914853

RESUMEN

The ability of a cancer cell to avoid apoptosis is crucial to tumorigenesis and can also contribute to chemoresistance. The Bcl-2 family of prosurvival proteins (Bcl-2, Bcl-X(L), Bcl-w, Mcl-1, and A1) plays a key role in these processes. We previously reported the discovery of ABT-263 (navitoclax), a potent small-molecule inhibitor of Bcl-2, Bcl-X(L), and Bcl-w. While navitoclax exhibits single-agent activity in tumors dependent on Bcl-2 or Bcl-X(L) for survival, the expression of Mcl-1 has been shown to confer resistance to navitoclax, most notably in solid tumors. Thus, therapeutic agents that can downregulate or neutralize Mcl-1 are predicted to synergize potently with navitoclax. Here, we report the activity of navitoclax in combination with 19 clinically relevant agents across a panel of 46 human solid tumor cell lines. Navitoclax broadly enhanced the activity of multiple therapeutic agents in vitro and enhanced efficacy of both docetaxel and erlotinib in xenograft models. The ability of navitoclax to synergize with docetaxel or erlotinib corresponded to an altered sensitivity of the mitochondria toward navitoclax, which was associated with the downmodulation of Mcl-1 and/or upregulation of Bim. These data provide a rationale to interrogate these combinations clinically.


Asunto(s)
Compuestos de Anilina/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Sulfonamidas/farmacología , Compuestos de Anilina/administración & dosificación , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Sinergismo Farmacológico , Femenino , Células HCT116 , Células Hep G2 , Humanos , Células K562 , Masculino , Ratones , Neoplasias/patología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas/administración & dosificación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína bcl-X/antagonistas & inhibidores
13.
J Biol Chem ; 280(49): 40599-608, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16227629

RESUMEN

Apo2 ligand/tumor necrosis factor (TNF)-related apoptosis-inducing ligand (Apo2L/TRAIL) mainly activates programmed cell death through caspases. By contrast, TNF primarily induces gene transcription through the inhibitor of kappaB kinase (IKK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase pathways. Apo2L/TRAIL also can stimulate these kinases, albeit less strongly; however, the underlying mechanisms of this stimulation and its relation to apoptosis are not well understood. Here we show that Apo2L/TRAIL activates kinase pathways by promoting the association of a secondary signaling complex, subsequent to assembly of a primary, death-inducing signaling complex (DISC). The secondary complex retained the DISC components FADD and caspase-8, but recruited several factors involved in kinase activation by TNF, namely, RIP1, TRAF2, and NEMO/IKKgamma. Secondary complex formation required Fas-associated death domain (FADD), as well as caspase-8 activity. Apo2L/TRAIL stimulation of JNK and p38 further depended on RIP1 and TRAF2, whereas IKK activation required NEMO. Apo2L/TRAIL induced secretion of interleukin-8 and monocyte chemoattractant protein-1, augmenting macrophage migration. Thus, Apo2L/TRAIL and TNF organize common molecular determinants in distinct signaling complexes to stimulate similar kinase pathways. One function of kinase stimulation by Apo2L/TRAIL may be to promote phagocytic engulfment of apoptotic cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/farmacología , Glicoproteínas de Membrana/farmacología , Proteínas Quinasas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Caspasa 8 , Caspasas/fisiología , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Activación Enzimática/efectos de los fármacos , Proteína de Dominio de Muerte Asociada a Fas , Humanos , Quinasa I-kappa B/metabolismo , Interleucina-8/metabolismo , Cinética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/fisiología , Fosforilación , ARN Interferente Pequeño , Transducción de Señal/fisiología , Factor 2 Asociado a Receptor de TNF/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Cell ; 123(5): 931-44, 2005 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-16325585

RESUMEN

Innate immunity is the first line of defense against infection, protecting the host during the development of adaptive immunity and critically affecting the nature of the adaptive response. We show that, in contrast to tumor necrosis factor alpha (TNF-alpha), the related protein TWEAK attenuates the transition from innate to adaptive mechanisms. TWEAK-/- mice had overabundant natural killer (NK) cells and displayed hypersensitivity to bacterial endotoxin, with their innate immune cells producing excess interferon (IFN)-gamma and interleukin (IL)-12. TWEAK inhibited stimulation of the transcriptional activator STAT-1 and induced p65 nuclear factor (NF)-kappaB association with histone deacetylase 1, repressing cytokine production. TWEAK-/- mice developed oversized spleens with expanded memory and T helper 1 (TH1) subtype cells upon aging and mounted stronger innate and adaptive TH1-based responses against tumor challenge. Thus, TWEAK suppresses production of IFN-gamma and IL-12, curtailing the innate response and its transition to adaptive TH1 immunity.


Asunto(s)
Inmunidad Celular/fisiología , Inmunidad Innata/fisiología , Factor de Necrosis Tumoral alfa/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Células Cultivadas , Citocina TWEAK , Endotoxinas/inmunología , Histona Desacetilasa 1 , Histona Desacetilasas/metabolismo , Humanos , Hipersensibilidad/inmunología , Interferón gamma/inmunología , Interleucina-12/inmunología , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Ratones , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Transcripción STAT1/metabolismo , Bazo/anatomía & histología , Bazo/inmunología , Subgrupos de Linfocitos T/inmunología , Receptor de TWEAK , Factor de Transcripción ReIA/metabolismo , Factores de Necrosis Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA