Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Divers ; 26(4): 2175-2188, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34668104

ABSTRACT

IFN-ß is a cytokine that plays a significant role in the immune system. Inhibition of IFN-ß might be used as a therapeutic approach to treat septic shock. A peptidomimetic previously developed by our research team, 1-benzyl-5-methyl-4-(n-octylamino)pyrimidin-2(1H)-one (LT87), was used as an cardioprotective agent in a myocardial ischemia (MI) mouse model. We have developed new LT87 derivatives by synthetizing its dimers in an attempt to extend its structural variety and enhance its biological activity. A dimeric derivative, LT127, exhibited a dose-dependent inhibition of LPS-mediated IFN-ß and subsequent CXCL10 mRNA transcription. The effect was selective and transduced through TLR4- and TRAM/TRIF-mediated signaling, with no significant effect on MyD88-dependent signaling. However, this effect was not specific to TLR4, since a similar effect was observed both on TLR8- and MDA5/RIG-I-stimulated IFN-ß expression. Nevertheless, LT127 might serve as a drug candidate, specifically as an inhibitor for IFN-ß production in order to develop a novel therapeutic approach to prevent septic shock.


Subject(s)
Interferon-beta , Peptidomimetics , Shock, Septic , Animals , Cytokines/metabolism , Interferon-beta/metabolism , Mice , Peptidomimetics/pharmacology , Shock, Septic/drug therapy , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
2.
Syst Rev ; 7(1): 191, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30428932

ABSTRACT

BACKGROUND: Originator trastuzumab (Herceptin®; H) is an antibody-targeted therapy to treat patients with human epidermal growth factor receptor 2-positive (HER2+) early breast cancer (EBC). We investigated the overall survival (OS) advantage conferred by the addition of H to chemotherapy for HER2+ EBC patients and how the OS advantage changed over time. METHODS: A systematic literature review (SLR) identified randomized controlled trials (RCTs) and non-randomized studies (NRSs) published from January 1, 1990 to January 19, 2017, comparing systemic therapies used in the neoadjuvant/adjuvant settings to treat HER2+ EBC patients. Bayesian cumulative network meta-analyses (cNMAs) of OS were conducted to assess the published literature over time. Heterogeneity was assessed through sensitivity and subgroup analyses. RESULTS: The SLR identified 31 unique studies (28 RCTs, 3 NRSs) included in the OS analyses from 2008 to 2016. In the reference case cNMA (RCTs alone), initial evidence demonstrated an OS advantage for H/chemotherapy compared with chemotherapy alone in HER2+ EBC patients. As additional OS data were published, the precision around this survival benefit strengthened over time. Both H/anthracycline-containing chemotherapy and H/non-anthracycline-containing chemotherapy regimens provided similar OS advantages for HER2+ EBC patients. CONCLUSION: This analysis represents the most comprehensive SLR/cNMA to date of published OS data in HER2+ EBC studies. These findings demonstrate why H/chemotherapy is now the established standard of care in HER2+ EBC. In the case of H, the benefits of early patient access far outweighed the risk of waiting for more precise information. SYSTEMATIC REVIEW REGISTRATION: PROSPERO CRD42017055763.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Anthracyclines/administration & dosage , Breast Neoplasms/metabolism , Female , Humans , Network Meta-Analysis , Receptor, ErbB-2/metabolism , Survival Rate , Trastuzumab/administration & dosage
3.
Syst Rev ; 6(1): 196, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29017563

ABSTRACT

BACKGROUND: Human epidermal growth factor receptor 2-positive (HER2+) breast cancer is an aggressive disease that makes up about 20% of all invasive breast cancers. HER2+ breast cancer is associated with poor prognosis and high mortality rates, but the development of HER2-targeted therapies, such as originator trastuzumab (Herceptin®), has substantially improved patient survival. Numerous clinical trials and reviews have investigated the efficacy of HER2-targeted therapies over the past few decades; however, no study has specifically investigated the vast body of evidence on trastuzumab in comparison to chemotherapy regimens, endocrine therapies, and other targeted therapies. This systematic review and cumulative network meta-analysis (NMA) will synthesize available evidence to evaluate the survival benefit conferred by the addition of originator trastuzumab to standard chemotherapy and to compare the most widely used trastuzumab regimens in patients with HER2+ early breast cancer, based on results from randomized controlled trials (RCTs) and comparative observational studies. METHODS/DESIGN: A systematic search of Embase, MEDLINE®, and the Cochrane Library has been designed by an experienced medical information specialist and peer reviewed by another senior information specialist. RCTs and comparative observational studies of patients with HER2+ early breast cancer indexed from 1990 onwards will be eligible for inclusion. Two investigators will independently assess studies for inclusion and use standardized data extraction templates to collect data on study and patient characteristics. The primary outcome of interest is overall survival. Bayesian cumulative NMA methods will be used to quantify the evolution of publicly available evidence using both fixed and random effects models. DISCUSSION: This study will evaluate survival trends associated with originator trastuzumab in patients with HER2+ early breast cancer. As originator trastuzumab has been researched in both clinical and real-world settings for close to 20 years, a cumulative NMA is likely to show improved precision around the parameter estimates for trastuzumab now compared with when the drug was initially launched in the USA in 1998. A better understanding of the evolution of publicly available comparative evidence for originator trastuzumab will further inform treatment for patients with HER2+ early breast cancer, providing benefit to patients, health professionals, and researchers. SYSTEMATIC REVIEW REGISTRATION: PROSPERO CRD42017055763 https://www.crd.york.ac.uk/PROSPERO.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Disease-Free Survival , Trastuzumab/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Humans , Network Meta-Analysis , Randomized Controlled Trials as Topic , Systematic Reviews as Topic
4.
PLoS One ; 10(2): e0118302, 2015.
Article in English | MEDLINE | ID: mdl-25710480

ABSTRACT

CD150 (IPO3/SLAM) belongs to the SLAM family of receptors and serves as a major entry receptor for measles virus. CD150 is expressed on normal and malignant cells of the immune system. However, little is known about its expression outside the hematopoietic system, especially tumors of the central nervous system (CNS). Although CD150 was not found in different regions of normal brain tissues, our immunohistochemical study revealed its expression in 77.6% of human CNS tumors, including glioblastoma, anaplastic astrocytoma, diffuse astrocytoma, ependymoma, and others. CD150 was detected in the cytoplasm, but not on the cell surface of glioma cell lines, and it was colocalized with the endoplasmic reticulum and Golgi complex markers. In addition to the full length mRNA of the mCD150 splice isoform, in glioma cells we found a highly expressed novel CD150 transcript (nCD150), containing an 83 bp insert. The insert is derived from a previously unrecognized exon designated Cyt-new, which is located 510 bp downstream of the transmembrane region exon, and is a specific feature of primate SLAMF1. Both mCD150 and nCD150 cDNA variants did not contain any mutations and had the leader sequence. The nCD150 transcript was also detected in normal and malignant B lymphocytes, primary T cells, dendritic cells and macrophages; however, in glioma cells nCD150 was found to be the predominant CD150 isoform. Similarly to mCD150, cell surface expression of nCD150 allows wild type measles virus entry to the cell. Our data indicate that CD150 expression in CNS tumors can be considered a new diagnostic marker and potential target for novel therapeutic approaches.


Subject(s)
Antigens, CD/genetics , Gene Expression Regulation , Receptors, Cell Surface/genetics , Amino Acid Sequence , Antigens, CD/chemistry , Antigens, CD/metabolism , B-Lymphocytes/metabolism , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Measles virus/physiology , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing , RNA, Messenger/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
5.
PLoS One ; 7(7): e42072, 2012.
Article in English | MEDLINE | ID: mdl-22848707

ABSTRACT

BACKGROUND: Epstein-Barr virus (EBV) encodes six nuclear transformation-associated proteins that induce extensive changes in cellular gene expression and signaling and induce B-cell transformation. The role of HIF1A in EBV-induced B-cell immortalization has not been previously studied. METHODS AND FINDINGS: Using Western blotting and Q-PCR, we found that HIF1A protein is stabilized in EBV-transformed lymphoblastoid cells. Western blotting, GST pulldown assays, and immunoprecipitation showed that EBV-encoded nuclear antigens EBNA-5 and EBNA-3 bind to prolylhydroxylases 1 and 2, respectively, thus inhibiting HIF1A hydroxylation and degradation. Immunostaining and Q-PCR showed that the stabilized HIF1A translocates to the nucleus, forms a heterodimer with ARNT, and transactivates several genes involved in aerobic glycolysis. Using biochemical assays and Q-PCR, we also found that lymphoblastoid cells produce high levels of lactate, lactate dehydrogenase and pyruvate. CONCLUSIONS: Our data suggest that activation of the aerobic glycolytic pathway, corresponding to the Warburg effect, occurs in EBV-transformed lymphoblastoid cells, in contrast to mitogen-activated B-cells.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Transformation, Viral , Glycolysis , Herpesvirus 4, Human/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Active Transport, Cell Nucleus , Aerobiosis , B-Lymphocytes/pathology , Cell Nucleus/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MCF-7 Cells , Procollagen-Proline Dioxygenase/metabolism , Protein Stability , Transcription, Genetic
6.
Int J Cancer ; 128(4): 817-25, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20473904

ABSTRACT

We report that MDM2, a negative regulator of p53, can bind to EBNA-5. Using GST pull-down assay, immunoprecipitation, surface plasmon resonance and immunostaining of lymphoblastoid cells, we found that trimolecular complexes are formed between EBNA-5, MDM2 and p53, where MDM2 serves as a bridge. The EBNA-5 binding to MDM2 counteracted destabilizing effect of the latter on the p53. In ubiquitination and degradation assays in vitro, EBNA-5 inhibited p53 polyubiquitination (but not monoubiquitination) in a concentration-dependent manner. This resembles the effect of p14ARF on p53. Moreover, EBNA-5 was found to inhibit the degradation of p53 in vitro. High levels of p53 expression were maintained in LCLs. The binding of EBNA-5 to MDM2 also could impair the functional activity of p53. The p53-dependent genes P21 and VDR were not induced in EBV-infected, in contrast to mitogen-activated cells. This may explain the tolerance of established LCLs to high levels of p53 without undergoing apoptosis.


Subject(s)
B-Lymphocytes/metabolism , Breast Neoplasms/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Trans-Activators , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , B-Lymphocytes/pathology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cells, Cultured , Chromatin Immunoprecipitation , Epstein-Barr Virus Nuclear Antigens/genetics , Female , Humans , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Ubiquitination
7.
Immunol Lett ; 134(2): 174-82, 2011 Jan 30.
Article in English | MEDLINE | ID: mdl-20933011

ABSTRACT

FCRL6 receptor is a more recently identified representative of the FCRL family. We generated a panel of mouse mAbs to baculovirus-derived recombinant FCRL6 protein. The clone 7B2 was found to specifically recognize a 63kDa protein expressed preferentially on the surface of CD8 T and CD56 NK cells in human peripheral blood and spleen. The clone 7B2 reacts with FCRL6 in Western blotting, FACS, and immunohistochemistry. In the T cell lineage, FCRL6 functions in antigen-experienced cells. Mitogenic stimulation of PB leukocytes in vitro resulted in an abrogation of the FCRL6 gene expression. We found a significant decrease in the FCRL6 gene expression in peripheral T cells of patients with certain autoimmune and blood diseases, and its upregulation at the late stages of HIV infection. Study of the FCRL6 association with signaling molecules showed its ability to recruit SHP-1, SHP-2, SHIP-1, and SHIP-2 phosphatases, and also adaptor protein Grb2 through phosphorylated cytoplasmic tyrosines. The current results demonstrate inhibitory potential of FCRL6 and suggest its possible involvement in modulation of CTL effector functions in various immune disorders.


Subject(s)
Carrier Proteins/immunology , Gene Expression Regulation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Alternative Splicing , Amino Acid Sequence , Autoimmune Diseases/immunology , Blood Cells/cytology , CD8-Positive T-Lymphocytes/immunology , Hematologic Diseases/immunology , Humans , Intracellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/immunology , Molecular Sequence Data , RNA, Messenger/immunology , Sequence Alignment , Spleen/cytology
8.
Immunol Cell Biol ; 88(5): 565-74, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20231852

ABSTRACT

The CD150 receptor is expressed on thymocytes, activated and memory T cells, B cells, platelets, natural killer T cells, and mature dendritic cells, and is also detected on tumor cells of Hodgkin's lymphoma (HL) and diffuse large B-cell lymphoma with an activated B cell phenotype. Here, we report that the level of CD150 expression is elevated during B cell differentiation toward plasma cells. In primary tonsillar B cells and HL cell lines, CD150 signaling regulates the phosphorylation of three types of mitogen-activated protein kinases (MAPKs): extracellular signal-regulated kinase 1/2 (ERK1/2), p38 MAPK, and Jun N-terminal kinase 1/2 (JNK1/2). CD150 induced ERK1/2 activation in primary tonsillar B cells and in two HL cell lines. CD150 mediated activation of JNK1/2 p54 and JNK2-gamma kinase isoforms in all CD150(+) B cell lines we tested. CD150 associated with the serine/threonine kinase hematopoetic progenitor kinase 1 (HPK1) regardless of CD150 tyrosine phosphorylation or binding of the SH2D1A adaptor protein to CD150, and HPK1 overexpression enhanced CD150-mediated JNK1/2 phosphorylation. CD150 ligation inhibited cell proliferation of all studied HL cell lines and induced apoptosis in L1236 HL cells that did not depend on JNK activity. As signaling through CD150 modulates MAPK activity in HL tumor cells, CD150 may contribute to regulation of tumor cell maintenance in low-rate proliferating HLs.


Subject(s)
Antigens, CD/immunology , B-Lymphocytes/immunology , Enzyme Activation/immunology , Hodgkin Disease/immunology , Mitogen-Activated Protein Kinases/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Blotting, Western , Cell Differentiation/immunology , Cell Line, Tumor , Cell Separation , Flow Cytometry , Hodgkin Disease/metabolism , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinase 8/immunology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/immunology , Mitogen-Activated Protein Kinase 9/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Transfection
9.
Proc Natl Acad Sci U S A ; 106(47): 19866-71, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19903879

ABSTRACT

We report that the overexpression of mitochondrial ribosomal protein MRPS18-2 (S18-2) can immortalize primary rat embryonic fibroblasts (REFs). The immortalized cells (18IM) lose contact inhibition, form foci, and are capable of anchorage-independent growth. Concurrently, mesodermal markers, such as vimentin, smooth muscle actin, and Fut4, disappear completely. 18IM cells express embryonic stem cell markers, such as SSEA-1, Sox2, and Oct3/4. In confluent cultures, a portion of cells also express ectoderm- and endoderm-specific pan-keratin, ectoderm-specific beta-III-tubulin, mesoderm-specific MHC class II, and become stainable for fat with Oil red O. None of these changes was detected in c-myc+Ha-ras (MR)-transformed cells. In immunodeficient mice, 18IM cells formed small transiently growing tumors that have down-regulated SSEA-1 and showed pan-keratin staining. We conclude that S18-2 can immortalize REFs and induces them to express stem cell traits.


Subject(s)
Cell Transformation, Neoplastic , Embryonic Stem Cells/physiology , Fibroblasts/physiology , Ribosomal Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Transplantation , Cells, Cultured , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Mice , Mice, SCID , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/genetics
10.
Mol Biotechnol ; 41(3): 270-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18989794

ABSTRACT

We have developed a surface plasmon resonance (SPR)-based immunocapture approach to study multimeric protein-protein complexes. A composition and spatial architecture of protein complexes that contained GST-tagged p53, p14ARF, and MDM2 was examined by the developed approach. Obtained results verified that the p53 protein possesses two binding sites for MDM2. Ternary complexes containing p14ARF, MDM2, and p53 proteins could only be formed when MDM2 protein functions as a bridging molecule. That was confirmed by immunoprecipitation and immunostaining.


Subject(s)
Multiprotein Complexes/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Tumor Suppressor Protein p14ARF/chemistry , Tumor Suppressor Protein p53/chemistry , HCT116 Cells , Humans , Immunohistochemistry , Microscopy, Fluorescence , Multiprotein Complexes/metabolism , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Reproducibility of Results , Sensitivity and Specificity , Staphylococcal Protein A/metabolism , Surface Plasmon Resonance , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism
11.
Proc Natl Acad Sci U S A ; 105(14): 5489-94, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18391203

ABSTRACT

Epstein-Barr virus (EBV), like other DNA tumor viruses, induces an S-phase in the natural host cell, the human B lymphocyte. This is linked with blast transformation. It is believed that the EBV-encoded nuclear antigen 6 (EBNA-6) is involved in the regulation of cell cycle entry. However, the possible mechanism of this regulation is not approached. In our current study, we found that EBNA-6 binds to a MRPS18-2 protein, and targets it to the nucleus. We found that MRPS18-2 binds to both hypo- and hyperphosphorylated forms of Rb protein specifically. This binding targets the small pocket of pRb, which is a site of interaction with E2F1. The MRPS18-2 competes with the binding of E2F1 to pRb, thereby raising the level of free E2F1. Our experimental data suggest that EBNA-6 may play a major role in the entry of EBV infected B cells into the S phase by binding to and raising the level of nuclear MRPS18-2, protein. This would inhibit pRb binding to E2F1 competitively and lift the block preventing S-phase entry.


Subject(s)
Active Transport, Cell Nucleus , Antigens, Viral/metabolism , E2F1 Transcription Factor/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Mitochondrial Proteins/metabolism , Retinoblastoma Protein/metabolism , Ribosomal Proteins/metabolism , Antigens, Viral/physiology , Cell Line , DNA, Complementary , Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/chemistry , Humans , Multiprotein Complexes/metabolism , Transfection
12.
Anal Bioanal Chem ; 386(7-8): 2063-73, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17086389

ABSTRACT

Biosensor technologies based on optical readout are widely used in protein-protein interaction studies. Here we describe a fast and simple approach to the creation of oriented interfacial architectures for surface plasmon resonance (SPR) transducers, based on conventional biochemical procedures and custom reagents. The proposed protocol permits the oriented affinity-capture of GST fusion proteins by a specific antibody which is bound to protein A, which in turn has been immobilized on the transducer surface (after the surface has been modified by guanidine thiocyanate). The applicability of the method was demonstrated by studying the interaction between retinoblastoma tumor suppressor protein (pRb) and MRS18-2 proteins. The formation of the pRb-MRS18-2 protein complex was examined and the pRb binding site (A-box-spacer-B-box) was mapped. We have also shown that MRS18-2, which was detected as the Epstein-Barr virus-encoded EBNA-6 binding partner using the yeast two-hybrid system, binds to pRb in GST pull-down assays.


Subject(s)
Immunoassay/methods , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Surface Plasmon Resonance/methods , Viral Proteins/metabolism , Antibodies/immunology , Binding Sites/immunology , Cell Line, Tumor , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Herpesvirus 4, Human , Humans , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/immunology
13.
Exp Cell Res ; 303(1): 47-55, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15572026

ABSTRACT

Epstein-Barr virus (EBV) transforms resting human B cells into immortalized immunoblasts. EBV-encoded nuclear antigens EBNA-5 (also called EBNA-LP) is one of the earliest viral proteins expressed in freshly infected B cells. We have recently shown that EBNA-5 binds p14ARF, a nucleolar protein that regulates the p53 pathway. Here, we report the identification of another protein with partially nucleolar localization, the v-fos transformation effector Fte-1 (Fte-1/S3a), as an EBNA-5 binding partner. In transfected cells, Fte-1/S3a and EBNA-5 proteins showed high levels of colocalization in extranucleolar inclusions. Fte-1/S3a has multiple biological functions. It enhances v-fos-mediated cellular transformation and is part of the small ribosomal subunit. It also interacts with the transcriptional factor CHOP and apoptosis regulator poly(ADP-ribose) polymerase (PARP). Fte-1/S3a is regularly expressed at high levels in both tumors and cancer cell lines. Its high expression favors the maintenance of malignant phenotype and undifferentiated state, whereas its down-regulation is associated with cellular differentiation and growth arrest. Here, we show that EBV-induced B cell transformation leads to the up-regulation of Fte-1/S3a. We suggest that EBNA-5 through binding may influence the growth promoting, differentiation inhibiting, or apoptosis regulating functions of Fte-1/S3a.


Subject(s)
B-Lymphocytes/metabolism , Cell Differentiation/physiology , Epstein-Barr Virus Nuclear Antigens/metabolism , Ribosomal Proteins/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/virology , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Herpesvirus 4, Human/genetics , Humans , Immunohistochemistry , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Transcription Factor CHOP , Transcription Factors/metabolism , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...