Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Bone Marrow Transplant ; 59(5): 637-646, 2024 May.
Article in English | MEDLINE | ID: mdl-38361117

ABSTRACT

The ruxolitinib compassionate use (CU) program offered ruxolitinib to patients ≥2 years of age with confirmed steroid-resistant acute or chronic graft-versus-host disease (aGvHD and cGvHD, respectively). Data from 1180 patients (n = 775, 370 and 35 with cGvHD, aGvHD, and non-specified GvHD, respectively) were analyzed. Most patients had severe cGvHD (56%) or stage III/IV aGvHD (70%) disease and had previously received corticosteroids ( > 80%); ruxolitinib was requested primarily as a second-/third-line option. Patients <12 and ≥12 years old most often received the recommended ruxolitinib doses (5 mg twice daily [BID] and 10 mg BID, respectively); however, 23% and 30% of ≥12 year olds with cGvHD and aGvHD, respectively, received the lower dose of 5 mg BID. Notably, corticosteroid usage decreased with ruxolitinib treatment; at the initial ruxolitinib request, 81% and 91% of patients with cGvHD and aGvHD, respectively, were receiving corticosteroids whereas at resupply, 62% and 64%, respectively, were receiving corticosteroids. Eighty two percent of evaluable patients with cGvHD had a complete or partial response to treatment and 56% of evaluable patients with aGvHD had a best response of grade 0/I. These findings demonstrate the rapid and positive effects of ruxolitinib in patients with GvHD in a real-world setting.


Subject(s)
Compassionate Use Trials , Graft vs Host Disease , Nitriles , Pyrazoles , Pyrimidines , Humans , Graft vs Host Disease/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Male , Female , Adult , Adolescent , Child , Middle Aged , Aged , Young Adult , Child, Preschool , Hematopoietic Stem Cell Transplantation
2.
Arch Virol ; 164(8): 2049-2059, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31123965

ABSTRACT

Avipoxviruses (APVs) are large DNA viruses that are detected widely in many species of birds. Little information is available regarding genetic variations in these host-specific viruses. In the present study, nine canarypox virus and five pigeonpox virus isolates were collected from northeastern Iran and isolated via the chorioallantoic membrane of chicken embryos. Further investigations were conducted using analysis of virus growth in chicken embryo fibroblasts, histopathology, electron microscopy, and molecular techniques such as polymerase chain reaction (PCR) combined with sequencing and phylogenetic analysis to investigate variations in the highly conserved P4b gene of poxviruses. Virus replication and pock lesions were evident, and microscopic examination revealed eosinophilic intracytoplasmic inclusion bodies and biconcave enveloped virus particles with randomly arranged surface filaments, which are characteristic features of poxviruses. PCR results confirmed the presence of an APV-specific 578-bp fragment in all of the samples. Sequence analysis and phylogenetic analysis of 578-bp P4b fragments of eight isolates confirmed that our canary and pigeon isolates clustered with previously reported isolates. The similarity between the nucleotide sequences of most of our isolates and those isolated previously in other countries could be due to the high degree of conservation of these fragments. However, the FZRC6V isolate from a canary in this study did not have a canarypox virus origin according to the sequence analysis, and might have originated from cross-infection with different strains of avipoxviruses.


Subject(s)
Avipoxvirus/genetics , Canarypox virus/genetics , Poxviridae Infections/virology , Animals , Bird Diseases/virology , Cells, Cultured , Chickens/virology , Conserved Sequence/genetics , Cross Infection/virology , Fibroblasts/virology , Genes, Viral/genetics , Genetic Variation/genetics , Iran , Phylogeny , Poultry Diseases/virology , Poxviridae/genetics
3.
J Virol ; 86(2): 726-37, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22072751

ABSTRACT

X-linked inhibitor of apoptosis (XIAP) is a potent antagonist of caspase 3-, 7-, and 9-dependent apoptotic activities that functions as an E3 ubiquitin ligase, and it targets caspases for degradation. In this study, we demonstrate that Sendai virus (SeV) infection results in the IKKε- or TBK1-mediated phosphorylation of XIAP in vivo at Ser430, resulting in Lys(48)-linked autoubiquitination at Lys322/328 residues, followed by the subsequent proteasomal degradation of XIAP. Interestingly, IKKε expression and XIAP turnover increases SeV-triggered mitochondrion-dependent apoptosis via the release of caspase 3, whereas TBK1 expression does not increase apoptosis. Interestingly, phosphorylation also regulates XIAP interaction with the transcription factor IRF3, suggesting a role in IRF3-Bax-mediated apoptosis. Our findings reveal a novel function of IKKε as a regulator of the virus-induced triggering of apoptosis via the phosphorylation-dependent turnover of XIAP.


Subject(s)
Apoptosis , I-kappa B Kinase/metabolism , Respirovirus Infections/metabolism , Respirovirus Infections/physiopathology , Sendai virus/physiology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Amino Acid Motifs , Cell Line , Humans , I-kappa B Kinase/genetics , Phosphorylation , Respirovirus Infections/virology , Sendai virus/genetics , X-Linked Inhibitor of Apoptosis Protein/chemistry , X-Linked Inhibitor of Apoptosis Protein/genetics
4.
J Virol ; 85(3): 1224-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21084468

ABSTRACT

The rapid induction of type I interferon (IFN) is essential for establishing innate antiviral responses. During infection, cytoplasmic viral RNA is sensed by two DExD/H box RNA helicases, RIG-I and MDA5, ultimately driving IFN production. Here, we demonstrate that purified genomic RNA from HIV-1 induces a RIG-I-dependent type I IFN response. Both the dimeric and monomeric forms of HIV-1 were sensed by RIG-I, but not MDA5, with monomeric RNA, usually found in defective HIV-1 particles, acting as a better inducer of IFN than dimeric RNA. However, despite the presence of HIV-1 RNA in the de novo infection of monocyte-derived macrophages, HIV-1 replication did not lead to a substantial induction of IFN signaling. We demonstrate the existence of an evasion mechanism based on the inhibition of the RIG-I sensor through the action of the HIV-1 protease (PR). Indeed, the ectopic expression of PR resulted in the inhibition of IFN regulatory factor 3 (IRF-3) phosphorylation and decreased expression of IFN and interferon-stimulated genes. A downregulation of cytoplasmic RIG-I levels occurred in cells undergoing a single-cycle infection with wild-type provirus BH10 but not in cells transfected with a protease-deficient provirus, BH10-PR(-). Cellular fractionation and confocal microscopy studies revealed that RIG-I translocated from the cytosol to an insoluble fraction during the de novo HIV-1 infection of monocyte-derived macrophages, in the presence of PR. The loss of cytoplasmic RIG-I was prevented by the lysosomal inhibitor E64, suggesting that PR targets RIG-I to the lysosomes. This study reveals a novel PR-dependent mechanism employed by HIV-1 to counteract the early IFN response to viral RNA in infected cells.


Subject(s)
DEAD-box RNA Helicases/antagonists & inhibitors , HIV Protease/metabolism , HIV-1/immunology , HIV-1/pathogenicity , Immune Evasion , Interferons/antagonists & inhibitors , Signal Transduction , Cells, Cultured , DEAD Box Protein 58 , Humans , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferons/immunology , Macrophages/immunology , Macrophages/virology , Protein Binding , RNA, Viral/immunology , Receptors, Immunologic
5.
J Mol Cell Biol ; 2(3): 110-2, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20022884

ABSTRACT

The cytosolic DNA sensing pathway has remained poorly defined thus far. A recent study by Ishikawa et al. demonstrates that STING is essential for DNA-mediated type I IFN production and host defence against DNA pathogens.


Subject(s)
DNA Viruses/physiology , Immunity, Innate , Interferon-beta/biosynthesis , Membrane Proteins/metabolism , RNA Viruses/physiology , Animals , Endoplasmic Reticulum/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Signal Transduction
6.
PLoS Pathog ; 5(11): e1000650, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19893624

ABSTRACT

The primary role of the innate immune response is to limit the spread of infectious pathogens, with activation of Toll-like receptor (TLR) and RIG-like receptor (RLR) pathways resulting in a pro-inflammatory response required to combat infection. Limiting the activation of these signaling pathways is likewise essential to prevent tissue injury in the host. Triad3A is an E3 ubiquitin ligase that interacts with several components of TLR signaling and modulates TLR activity. In the present study, we demonstrate that Triad3A negatively regulates the RIG-I RNA sensing pathway through Lys48-linked, ubiquitin-mediated degradation of the tumor necrosis factor receptor-associated factor 3 (TRAF3) adapter. Triad3A was induced following dsRNA exposure or virus infection and decreased TRAF3 levels in a dose-dependent manner; moreover, Triad3A expression blocked IRF-3 activation by Ser-396 phosphorylation and inhibited the expression of type 1 interferon and antiviral genes. Lys48-linked ubiquitination of TRAF3 by Triad3A increased TRAF3 turnover, whereas reduction of Triad3A expression by stable shRNA expression correlated with an increase in TRAF3 protein expression and enhancement of the antiviral response following VSV or Sendai virus infection. Triad3A and TRAF3 physically interacted together, and TRAF3 residues Y440 and Q442--previously shown to be important for association with the MAVS adapter--were also critical for Triad3A. Point mutation of the TRAF-Interacting-Motif (TIM) of Triad3A abrogated its ability to interact with TRAF3 and modulate RIG-I signaling. TRAF3 appears to undergo sequential ubiquitin "immuno-editing" following virus infection that is crucial for regulation of RIG-I-dependent signaling to the antiviral response. Thus, Triad3A represents a versatile E3 ubiquitin ligase that negatively regulates RIG-like receptor signaling by targeting TRAF3 for degradation following RNA virus infection.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/physiology , RNA Virus Infections/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism , Binding Sites , Cell Line , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Binding , RNA, Double-Stranded , TNF Receptor-Associated Factor 3/genetics , Transcriptional Activation , Transfection
7.
Semin Immunol ; 21(4): 215-22, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19539500

ABSTRACT

Viral and microbial pathogens contain specific motifs or pathogen-associated molecular patterns (PAMPs) that are recognized by cell surface- and endosome-associated Toll-like receptors (TLRs). RNA virus infection is also detected through TLR-independent mechanisms. Early viral replicative intermediates are detected by two recently characterized cystolic viral RNA receptors-RIG-I and MDA-5. Both are DExDH/box RNA helicases, and RIG-I specifically recognizes 5'-triphosphate containing viral RNA and transmits signals that induce type I interferon-mediated host immunity against virus infection. In this review, we will focus on RIG-I-like receptor (RLR) signal transduction and the regulatory mechanisms - ubiquitination, deubiquitination, ISGylation - underlying this important host response.


Subject(s)
DEAD-box RNA Helicases/immunology , DEAD-box RNA Helicases/metabolism , RNA Virus Infections/immunology , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , DEAD-box RNA Helicases/chemistry , Humans , RNA, Viral/immunology , Signal Transduction , Ubiquitination
8.
PLoS One ; 4(5): e5500, 2009.
Article in English | MEDLINE | ID: mdl-19430534

ABSTRACT

BACKGROUND: Interferon regulatory factor (IRF) family members have been implicated as critical transcription factors that function in immune response, hematopoietic differentiation and cell growth regulation. Activation of IRF-5 results in the production of pro-inflammatory cytokines such as TNFalpha, IL6 and IL12p40, as well as type I interferons. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we identify a G202C (position relative to translation start codon) missense-mutation transcript of IRF-5 in transformed B and T cell lines, which were either infected or non-infected by viruses, and peripheral blood from ATL or CLL patients. The mutated transcript encodes a novel protein in which the sixty-eighth amino acid, Alanine, is substituted by Proline (IRF-5P68) in the DNA binding domain of IRF-5. IRF-5P68 phenotype results in a complete loss of its DNA-binding activity and functions as a dominant negative molecule through interacting with wild type IRF-5. Co-expression of IRF-5P68 inhibits MyD88-mediated IRF-5 transactivation. Moreover, Toll-like receptor (TLR)-dependent IL6 and IL12P40 production induced by lipopolysaccharide (LPS), R837 or CpG ODN 1826 was reduced in IRF-5 (P68) expressing cells as compared to the control cells. CONCLUSION: IRF-5P68 acts as a dominant negative regulator that interferes with IRF-5-mediated production of pro-inflammatory cytokines. The functional characterization of the novel IRF-5 mutant in transformed B and T cell lines and in ATL and CLL patients may lead to a better understanding of the role of these transcriptional regulators in hematopoietic malignancies.


Subject(s)
Interferon Regulatory Factors/genetics , Mutant Proteins/genetics , Transcriptional Activation , Animals , Cell Line , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Genes, Dominant , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/physiology , Interleukin-12/genetics , Interleukin-12/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Jurkat Cells , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia-Lymphoma, Adult T-Cell/blood , Leukemia-Lymphoma, Adult T-Cell/pathology , Lipopolysaccharides/pharmacology , Lymphocytes/metabolism , Lymphocytes/pathology , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Mutant Proteins/metabolism , Mutant Proteins/physiology , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
J Virol ; 82(12): 5735-49, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18417567

ABSTRACT

Vesicular stomatitis virus (VSV) is a candidate oncolytic virus that replicates and induces cell death in cancer cells while sparing normal cells. Although defects in the interferon antiviral response facilitate VSV oncolysis, other host factors, including translational and growth regulatory mechanisms, also appear to influence oncolytic virus activity. We previously demonstrated that VSV infection induces apoptosis in proliferating CD4(+) T lymphocytes from adult T-cell leukemia samples but not in resting T lymphocytes or primary chronic lymphocytic leukemia cells that remain arrested in G(0). Activation of primary CD4(+) T lymphocytes with anti-CD3/CD28 is sufficient to induce VSV replication and cell death in a manner dependent on activation of the MEK1/2, c-Jun NH(2)-terminal kinase, or phosphatidylinositol 3-kinase pathway but not p38. VSV replication is specifically impaired by the cell cycle inhibitor olomoucine or rapamycin, which induces early G(1) arrest, but not by aphidicolin or Taxol, which blocks at the G(1)1S or G(2)1M phase, respectively; this result suggests a requirement for cell cycle entry for efficient VSV replication. The relationship between increased protein translation following G(0)/G(1) transition and VSV permissiveness is highlighted by the absence of mTOR and/or eIF4E phosphorylation whenever VSV replication is impaired. Furthermore, VSV protein production in activated T cells is diminished by small interfering RNA-mediated eIF4E knockdown. These results demonstrate that VSV replication in primary T lymphocytes relies on cell cycle transition from the G(0) phase to the G(1) phase, which is characterized by a sharp increase in ribogenesis and protein synthesis.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Cycle , Protein Biosynthesis , Vesicular stomatitis Indiana virus/physiology , B-Lymphocytes/virology , Cell Death , Cell Line , Flow Cytometry , Humans , Lymphocyte Activation , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Transfection , Viral Plaque Assay , Virus Replication
10.
Virology ; 365(1): 20-33, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17451770

ABSTRACT

An effective type I interferon (IFN-alpha/beta) response is critical for the control of many viral infections. Using an oncolytic strain of vesicular stomatitis virus, we have examined the cross-talk between virus-induced apoptosis and initiation of innate immune response. The intrinsic apoptotic cascade, specifically the Bax-Bcl-2-Caspase-9 cascade, was revealed as the primary pathway of VSV-induced apoptosis. Cell death was significantly reduced in BaxBak(-/-) murine embryonic fibroblasts (MEFs) and in human A549 epithelial cells treated with siRNA against Bax. Although inhibition of apoptosis resulted in enhanced virus replication in the BaxBak(-/-) MEFs as compared to wild-type cells, induction of the IFN antiviral response and expression of cytokine genes were attenuated in virus-infected cells. Moreover, Bax but not Bak pro-apoptotic protein was required for IRF-3 phosphorylation and activation, further substantiating a role for the intrinsic mitochondrial pathway in the innate immune response. Therefore, virus-induced apoptosis through a Bax-dependent mitochondrial pathway appears to enhance the full development of the IRF-3 mediated IFN antiviral response.


Subject(s)
Immunity, Innate , Interferon Regulatory Factor-3/physiology , Rhabdoviridae Infections/immunology , Vesicular stomatitis Indiana virus/immunology , bcl-2-Associated X Protein/physiology , Animals , Cell Culture Techniques , Fibroblasts/drug effects , Fibroblasts/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Vesicular stomatitis Indiana virus/physiology
11.
J Virol ; 80(12): 6072-83, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731946

ABSTRACT

Intracellular RNA virus infection is detected by the cytoplasmic RNA helicase RIG-I that plays an essential role in signaling to the host antiviral response. Recently, the adapter molecule that links RIG-I sensing of incoming viral RNA to downstream signaling and gene activation events was characterized by four different groups; MAVS/IPS-1-1/VISA/Cardif contains an amino-terminal CARD domain and a carboxyl-terminal mitochondrial transmembrane sequence that localizes to the mitochondrial membrane. Furthermore, the hepatitis C virus NS3-4A protease complex specifically targets MAVS/IPS-1/VISA/Cardif for cleavage as part of its immune evasion strategy. With a novel search program written in python, we also identified an uncharacterized protein, KIAA1271 (K1271), containing a single CARD-like domain at the N terminus and a Leu-Val-rich C terminus that is identical to that of MAVS/IPS-1/VISA/Cardif. Using a combination of biochemical analysis, subcellular fractionation, and confocal microscopy, we now demonstrate that NS3-4A cleavage of MAVS/IPS-1/VISA/Cardif/K1271 results in its dissociation from the mitochondrial membrane and disrupts signaling to the antiviral immune response. Furthermore, virus-induced IKKepsilon kinase, but not TBK1, colocalized strongly with MAVS at the mitochondrial membrane, and the localization of both molecules was disrupted by NS3-4A expression. Mutation of the critical cysteine 508 to alanine was sufficient to maintain mitochondrial localization of MAVS/IPS-1/VISA/Cardif and IKKepsilon in the presence of NS3-4A. These observations provide an outline of the mechanism by which hepatitis C virus evades the interferon antiviral response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hepacivirus , I-kappa B Kinase/metabolism , Mitochondrial Proteins/metabolism , Multiprotein Complexes/metabolism , Viral Nonstructural Proteins/metabolism , Hepacivirus/chemistry , Humans , Intracellular Membranes/metabolism , Mitochondria/ultrastructure , Peptide Hydrolases/metabolism , Signal Transduction/immunology
12.
Trends Mol Med ; 12(2): 53-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16406812

ABSTRACT

Intracellular viral infection is detected by the cytoplasmic RNA helicase RIG-I, which has an essential role in initiating the host antiviral response. The adaptor molecule that connects RIG-I sensing of incoming viral RNA to downstream signaling and gene activation has recently been elucidated by four independent research groups, and has been ascribed four different names: MAVS, IPS-1, VISA and Cardif. The fact that MAVS/IPS-1/VISA/Cardif localizes to the mitochondrial membrane suggests a link between viral infection, mitochondrial function and development of innate immunity. Furthermore, the hepatitis C virus NS3/4A protease specifically cleaves MAVS/IPS-1/VISA/Cardif as part of its immune-evasion strategy. These studies highlight a novel role for the mitochondria and for caspase activation and recruitment domain (CARD)-containing proteins in coordinating immune and apoptotic responses.


Subject(s)
Immunity, Innate/immunology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NF-kappa B/metabolism , Animals , Apoptosis , Caspases/metabolism , Humans
13.
J Biol Chem ; 281(4): 2095-103, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16306043

ABSTRACT

Activation of the interferon regulatory factors (IRFs) 3 and 7 transcription factors is essential for the induction of type I interferon (IFN) and development of the innate antiviral response. Retinoic acid-inducible gene I has been shown to contribute to virus-induced IFN production independent of the Toll-like receptor pathways in response to a variety of RNA viruses and double-stranded RNA. In the present study, we demonstrate that the NF-kappaB-inducible, anti-apoptotic protein A20 efficiently blocks RIG-I-mediated activation of NF-kappaB-, IRF-3-, and IRF-7-dependent promoters but only weakly interferes with TRIF-TLR-3-mediated IFN activation. Expression of A20 completely blocked CARD domain containing DeltaRIG-I-induced IRF-3 Ser-396 phosphorylation, homodimerization, and DNA binding. The level of A20 inhibition was upstream of the TBK1/IKKepsilon kinases that phosphorylate IRF3 and IRF7 and paradoxically, A20 selectively degraded the TRIF protein but not RIG-I. A20 possesses two ubiquitin-editing domains, an N-terminal deubiquitination domain and a C-terminal ubiquitin ligase domain consisting of seven zinc finger domains. Deletion of the N-terminal de-ubiquitination domain had no significant effect on the inhibitory effect of A20, whereas deletion or mutation of zinc finger motif 7 ablated the inhibitory function of A20 on IRF- or NF-kappaB-mediated gene expression. Furthermore, cells stably expressing the active form of RIG-I induced an antiviral state that interfered with replication of vesicular stomatitis virus, an effect that was reversed by stable co-expression of A20. These results suggest that the virus-inducible, NF-kappaB-dependent activation of A20 functions as a negative regulator of RIG-I-mediated induction of the antiviral state.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Proteins/physiology , RNA Helicases/physiology , Ubiquitin/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Antiviral Agents/chemistry , Blotting, Western , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases , DNA-Binding Proteins , Dimerization , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Humans , Immunoprecipitation , Interferons/metabolism , Intracellular Signaling Peptides and Proteins , Luciferases/metabolism , Mutagenesis , Nuclear Proteins , Open Reading Frames , Phosphorylation , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , RNA Viruses/metabolism , RNA, Double-Stranded/chemistry , Receptors, Immunologic , Signal Transduction , Toll-Like Receptors/metabolism , Transcription Factors/chemistry , Transcriptional Activation , Transfection , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Protein Ligases/chemistry , Vesicular stomatitis Indiana virus/metabolism , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...