Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell ; 146(6): 918-30, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925315

ABSTRACT

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.


Subject(s)
Endopeptidases/metabolism , Inhibitor of Differentiation Proteins/metabolism , Mesenchymal Stem Cells/cytology , Neoplastic Stem Cells/cytology , Osteosarcoma/pathology , Animals , Arabidopsis Proteins , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Knockdown Techniques , Humans , Mice , Ubiquitin-Specific Proteases , Ubiquitination
2.
Immunity ; 44(3): 609-621, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26944201

ABSTRACT

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.


Subject(s)
B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma/therapy , Colonic Neoplasms/therapy , Immunotherapy , Animals , Antibodies, Monoclonal/administration & dosage , Apoptosis , Azetidines/administration & dosage , Azetidines/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Carcinoma/immunology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Colonic Neoplasms/immunology , Drug Synergism , Drug Therapy , Drug Therapy, Combination , Extracellular Signal-Regulated MAP Kinases , Humans , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , Neoplasm Transplantation , Piperidines/administration & dosage , Piperidines/pharmacology
3.
Nature ; 510(7505): 407-411, 2014 06 19.
Article in English | MEDLINE | ID: mdl-24919154

ABSTRACT

Inhibitors against the p110ƎĀ“ isoform of phosphoinositide-3-OH kinase (PI(3)K) have shown remarkable therapeutic efficacy in some human leukaemias. As p110ƎĀ“ is primarily expressed in leukocytes, drugs against p110ƎĀ“ have not been considered for the treatment of solid tumours. Here we report that p110ƎĀ“ inactivation in mice protects against a broad range of cancers, including non-haematological solid tumours. We demonstrate that p110ƎĀ“ inactivation in regulatory T cells unleashes CD8(+) cytotoxic T cells and induces tumour regression. Thus, p110ƎĀ“ inhibitors can break tumour-induced immune tolerance and should be considered for wider use in oncology.


Subject(s)
Enzyme Inhibitors/pharmacology , Immune Tolerance/drug effects , Neoplasms/enzymology , Neoplasms/immunology , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Agents/pharmacology , Enzyme Activation/drug effects , Immune Tolerance/immunology , Mice , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology
5.
Nature ; 471(7336): 110-4, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368834

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin Modulators/pharmacology , Tubulin/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Drug Resistance, Neoplasm , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Fibroblasts , Humans , Mice , Mitosis/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Paclitaxel/pharmacology , Pharmacogenetics , Phosphorylation/drug effects , Polyploidy , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Vincristine/pharmacology
6.
Nature ; 463(7277): 103-7, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20023629

ABSTRACT

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.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Polyubiquitin/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival , DNA Damage , Docetaxel , Etoposide/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Half-Life , Humans , Lysine/metabolism , Mice , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasms/diagnosis , Nitrophenols/pharmacology , Phosphorylation/radiation effects , Piperazines/pharmacology , Prognosis , Protein Binding/radiation effects , Protein Stability , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Sulfonamides/pharmacology , Taxoids/pharmacology , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/genetics , Ubiquitination , Ultraviolet Rays , Xenograft Model Antitumor Assays
7.
MAbs ; 15(1): 2229101, 2023.
Article in English | MEDLINE | ID: mdl-37639687

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Immunoconjugates , United States , Immunoconjugates/therapeutic use , Antibodies, Monoclonal , United States Food and Drug Administration
8.
Cancer Cell ; 2(2): 139-48, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12204534

ABSTRACT

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.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplasms/immunology , fas Receptor/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Division/drug effects , Cell Transformation, Neoplastic , Disease Progression , Flow Cytometry , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Hydroxamic Acids/pharmacology , Immunity, Innate , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Time Factors , Transfection , Tumor Cells, Cultured , fas Receptor/genetics
9.
Dev Cell ; 2(3): 331-41, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11879638

ABSTRACT

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.


Subject(s)
Adipocytes/physiology , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , 3T3 Cells , Adipocytes/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/physiology , Cell Hypoxia/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Nuclear Proteins/metabolism , Oxygen/pharmacology
10.
Nat Med ; 19(2): 202-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23291630

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Platelets/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Hematologic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Survival/drug effects , Dogs , Female , HeLa Cells , Humans , Mice , Mice, SCID , Proto-Oncogene Proteins c-bcl-2/chemistry , Tumor Burden , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors
11.
Sci Signal ; 5(216): ra22, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22434933

ABSTRACT

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 Ɵ (IKKƟ), 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.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction/immunology , Tumor Necrosis Factors/metabolism , Blotting, Western , Carrier Proteins/metabolism , Cell Line, Tumor , Gene Silencing , Humans , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/immunology , Protein Transport , RNA, Small Interfering/genetics , Receptors, Interleukin-4/metabolism , TNF Receptor-Associated Factor 2/metabolism , Tumor Necrosis Factors/immunology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases
12.
Mol Cancer Ther ; 10(12): 2340-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21914853

ABSTRACT

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.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Sulfonamides/pharmacology , Aniline Compounds/administration & dosage , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Drug Synergism , Female , HCT116 Cells , Hep G2 Cells , Humans , K562 Cells , Male , Mice , Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors
13.
J Biol Chem ; 280(49): 40599-608, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16227629

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Membrane Glycoproteins/pharmacology , Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adaptor Proteins, Signal Transducing/physiology , Animals , Caspase 8 , Caspases/physiology , Cell Line, Tumor , Chemokine CCL2/metabolism , Death Domain Receptor Signaling Adaptor Proteins , Enzyme Activation/drug effects , Fas-Associated Death Domain Protein , Humans , I-kappa B Kinase/metabolism , Interleukin-8/metabolism , Kinetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , NF-kappa B/physiology , Phosphorylation , RNA, Small Interfering , Signal Transduction/physiology , TNF Receptor-Associated Factor 2/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Cell ; 123(5): 931-44, 2005 Dec 02.
Article in English | MEDLINE | ID: mdl-16325585

ABSTRACT

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.


Subject(s)
Immunity, Cellular/physiology , Immunity, Innate/physiology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factors/immunology , Animals , Cells, Cultured , Cytokine TWEAK , Endotoxins/immunology , Histone Deacetylase 1 , Histone Deacetylases/metabolism , Humans , Hypersensitivity/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , STAT1 Transcription Factor/metabolism , Spleen/anatomy & histology , Spleen/immunology , T-Lymphocyte Subsets/immunology , TWEAK Receptor , Transcription Factor RelA/metabolism , Tumor Necrosis Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL