Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Virol ; : e0054824, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864622

ABSTRACT

Most mature B-cell malignancies originate from the malignant transformation of germinal center (GC) B cells. The GC reaction appears to have a role in malignant transformation, in which a major player of the GC reaction is BCL6, a key regulator of this process. We now demonstrate that BCL6 protein levels were dramatically decreased in Epstein-Barr virus (EBV)-positive lymphoblastoid cell lines and Burkitt's lymphoma cell lines. Notably, BCL6 degradation was significantly enhanced in the presence of both EBNA3C and FBXO11. Furthermore, the amino-terminal domain of EBNA3C, which contains residues 50-100, interacts directly with FBXO11. The expression of EBNA3C and FBXO11 resulted in a significant induction of cell proliferation. Furthermore, BCL6 protein expression levels were regulated by EBNA3C via the Skp Cullin Fbox (SCF)FBXO11 complex, which mediated its ubiquitylation, and knockdown of FBXO11 suppressed the transformation of lymphoblastoid cell lines. These data provide new insights into the function of EBNA3C in B-cell transformation during GC reaction and raise the possibility of developing new targeted therapies against EBV-associated cancers. IMPORTANCE: The novel revelation in our study involves the suppression of BCL6 expression by the essential Epstein-Barr virus (EBV) antigen EBNA3C, shedding new light on our current comprehension of how EBV contributes to lymphomagenesis by impeding the germinal center reaction. It is crucial to note that while several EBV latent proteins are expressed in infected cells, the collaborative mechanisms among these proteins in regulating B-cell development or inducing B-cell lymphoma require additional investigation. Nonetheless, our findings carry significance for the development of emerging strategies aimed at addressing EBV-associated cancers.

2.
J Biomed Sci ; 30(1): 18, 2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36918845

ABSTRACT

BACKGROUND: Reactivation of Epstein Barr virus (EBV) leads to modulation of the viral and cellular epitranscriptome. N6-methyladenosine (m6A) modification is a type of RNA modification that regulates metabolism of mRNAs. Previous reports demonstrated that m6A modification affects the stability and metabolism of EBV encoded mRNAs. However, the effect of reactivation on reprograming of the cellular mRNAs, and how this contributes to successful induction of lytic reactivation is not known. METHODS: Methylated RNA immunoprecipitation sequencing (MeRIP-seq), transcriptomic RNA sequencing (RNA-seq) and RNA pull-down PCR were used to screen and validate differentially methylated targets. Western blotting, quantitative real-time PCR (RT-qPCR) and immunocytochemistry were used to investigate the expression and localization of different proteins. RNA stability and polysome analysis assays were used to detect the half-lives and translation efficiencies of downstream genes. Insertion of point mutation to disrupt the m6A methylation sites was used to verify the effect of m6A methylation on its stability and expression levels. RESULTS: We report that during EBV reactivation the m6A eraser ALKBH5 is significantly downregulated leading to enhanced methylation of the cellular transcripts DTX4 and TYK2, that results in degradation of TYK2 mRNAs and higher efficiency of translation of DTX4 mRNAs. This resulted in attenuation of IFN signaling that promoted progression of viral lytic replication. Furthermore, inhibition of m6A methylation of these transcripts led to increased production of IFN, and a substantial reduction in viral copy number, which suggests abrogation of lytic viral replication. CONCLUSION: Our findings illuminate the significance of m6A modification in overcoming the innate immune response during EBV reactivation. We now report that during lytic reactivation EBV targets the RNA methylation system of the host to attenuate the innate immune response by suppressing the interferon signaling which facilitates successful lytic replication of the virus.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Humans , Herpesvirus 4, Human/genetics , Epstein-Barr Virus Infections/genetics , Virus Activation/genetics , Virus Replication/genetics , RNA
3.
Cell Death Dis ; 13(10): 894, 2022 10 22.
Article in English | MEDLINE | ID: mdl-36272970

ABSTRACT

Epstein-Barr virus (EBV) is an opportunistic pathogen that can manifest itself as a potential contributor to human diseases years after primary infection, specifically in lymphoid and epithelial cell malignancies in immune-competent and immune-compromised hosts. The virus shuttles between B cells and epithelial cells during its infection cycle, facilitating its persistence and transmission in humans. While EBV efficiently infects and transforms B-lymphocytes, epithelial cells are not as susceptible to transformation in vitro. We utilized a 3D platform for culturing normal oral keratinocyte cells (NOKs) using Matrigel for greater insights into the molecular interactions between EBV and infected cells. We determined the transcriptome of EBV infected NOKs and peripheral blood mononuclear cells (PBMCs) for 7 and 15 days. LMPs (-1, -2A, and -2B) and EBNAs (-1, -2, -3A, -3B and -3C) were detected in all samples, and lytic gene expression was significantly higher in NOKs than PBMCs. We identified over 2000 cellular genes that were differentially expressed (P-value<0.05). Gene ontology (GO) and pathway analyses significantly identified pathways related to collagen-activation, chemokine signaling, immune response, metabolism, and antiviral responses. We also identified significant changes in metalloproteases and genes encoding chemotactic ligands and cell surface molecules. C-X-C chemokine receptor type 4 (CXCR4) was dramatically downregulated in PBMCs and upregulated in NOKs. However, MMP1 was significantly downregulated in NOKs and upregulated in PBMCs. Therefore, multiple pathways contribute to distinct pathologies associated with EBV infection in epithelial and B cells, and MMP1 and CXCR4 are critical molecules involved in regulation of latent and lytic states linked to viral associated diseases.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Humans , Herpesvirus 4, Human/metabolism , Epstein-Barr Virus Infections/metabolism , Transcriptome/genetics , Matrix Metalloproteinase 1/metabolism , Host Microbial Interactions , Leukocytes, Mononuclear/metabolism , Host-Pathogen Interactions/genetics , B-Lymphocytes/metabolism , Epithelial Cells/metabolism , Receptors, Chemokine/metabolism , Antiviral Agents , Chemokines/metabolism
4.
Cancers (Basel) ; 14(3)2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35158879

ABSTRACT

Deregulation of the ubiquitin-proteasome system (UPS) plays a critical role in the development of numerous human cancers. Epstein-Barr virus (EBV), the first known human tumor virus, has evolved distinct molecular mechanisms to manipulate the ubiquitin-proteasome system, facilitate its successful infection, and drive opportunistic cancers. The interactions of EBV antigens with the ubiquitin-proteasome system can lead to oncogenesis through the targeting of cellular factors involved in proliferation. Recent studies highlight the central role of the ubiquitin-proteasome system in EBV infection. This review will summarize the versatile strategies in EBV-mediated oncogenesis that contribute to the development of specific therapeutic approaches to treat EBV-associated malignancies.

5.
Elife ; 102021 07 19.
Article in English | MEDLINE | ID: mdl-34279223

ABSTRACT

The cellular adaptive response to hypoxia, mediated by high HIF1α levels includes metabolic reprogramming, restricted DNA replication and cell division. In contrast to healthy cells, the genome of cancer cells, and Kaposi's sarcoma associated herpesvirus (KSHV) infected cells maintains replication in hypoxia. We show that KSHV infection, despite promoting expression of HIF1α in normoxia, can also restrict transcriptional activity, and promoted its degradation in hypoxia. KSHV-encoded vCyclin, expressed in hypoxia, mediated HIF1α cytosolic translocation, and its degradation through a non-canonical lysosomal pathway. Attenuation of HIF1α levels by vCyclin allowed cells to bypass the block to DNA replication and cell proliferation in hypoxia. These results demonstrated that KSHV utilizes a unique strategy to balance HIF1α levels to overcome replication arrest and induction of the oncogenic phenotype, which are dependent on the levels of oxygen in the microenvironment.


Subject(s)
Cyclins/genetics , Cyclins/metabolism , DNA Replication , Herpesvirus 8, Human/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Cell Proliferation , Gene Expression Regulation, Viral , HEK293 Cells , Herpesviridae Infections/metabolism , Humans , Oxygen/metabolism , Transcriptome , Virus Replication/physiology
7.
Viruses ; 12(12)2020 12 01.
Article in English | MEDLINE | ID: mdl-33271926

ABSTRACT

Epigenetics is a versatile player in manipulating viral infection and a potential therapeutic target for the treatment of viral-induced diseases. Both epigenetics and metabolism are crucial in establishing a highly specific transcriptional network, which may promote or suppress virus infection. Human herpesvirus infection can induce a broad range of human malignancies and is largely dependent on the status of cellular epigenetics as well as its related metabolism. However, the crosstalk between epigenetics and metabolism during herpesvirus infection has not been fully explored. Here, we describe how epigenetic regulation of cellular metabolism affects herpesvirus infection and induces viral diseases. This further highlights the importance of epigenetics and metabolism during viral infection and provides novel insights into the development of targeted therapies.


Subject(s)
Energy Metabolism , Epigenesis, Genetic , Gene Expression Regulation, Viral , Herpesviridae Infections/genetics , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Herpesviridae/physiology , Host-Pathogen Interactions/genetics , Acetyl Coenzyme A/metabolism , DNA Methylation , Fatty Acids/metabolism , Flavin-Adenine Dinucleotide/metabolism , Glutamates/metabolism , Glycolysis , Histones/metabolism , Humans , NAD
8.
Cancers (Basel) ; 12(9)2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32916819

ABSTRACT

The Epstein-Barr virus (EBV) is the first human tumor virus identified that can transform quiescent B lymphocytes into lymphoblastoid cell lines (LCLs) in vitro. EBV can establish asymptomatic life-long persistence and is associated with multiple human malignancies, including non-Hodgkin lymphoma and Hodgkin lymphoma, as well as infectious mononucleosis. Although EBV-associated lymphomagenesis has been investigated for over 50 years, viral-mediated transformation is not completely understood, and the development of EBV-specific therapeutic strategies to treat the associated cancers is still a major challenge. However, the rapid development of several novel therapies offers exciting possibilities to target EBV-induced lymphomas. This review highlights targeted therapies with potential for treating EBV-associated lymphomas, including small molecule inhibitors, immunotherapy, cell therapy, preventative and therapeutic vaccines, and other potent approaches, which are novel strategies for controlling, preventing, and treating these viral-induced malignances.

9.
Bioorg Med Chem Lett ; 30(23): 127553, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32971261

ABSTRACT

Brusatol, a quassinoid natural product, is effective against multiple diseases including hematologic malignancies, as we reported recently by targeting the PI3Kγ isoform, but toxicity limits its further development. Herein, we report the synthesis of a series of conjugates of brusatol with amino acids and short peptides at its enolic hydroxyl at C-3. A number of conjugates with smaller amino acids and peptides demonstrated activities comparable to brusatol. Through in vitro and in vivo evaluations, we identified UPB-26, a conjugate of brusatol with a L- ß-homoalanine, which exhibits good chemical stability at physiological pH's (SGF and SIF), moderate rate of conversion to brusatol in both human and rat plasmas, improved mouse liver microsomal stability, and most encouragingly, enhanced safety compared to brusatol in mice upon IP administration.


Subject(s)
Aminobutyrates/pharmacology , Antineoplastic Agents/pharmacology , Quassins/pharmacology , Aminobutyrates/chemical synthesis , Aminobutyrates/metabolism , Aminobutyrates/toxicity , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Male , Mice, Inbred NOD , Mice, SCID , Microsomes, Liver/metabolism , Molecular Structure , Quassins/chemical synthesis , Quassins/metabolism , Quassins/toxicity , Rats , Structure-Activity Relationship
10.
Annu Rev Virol ; 7(1): 309-331, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32991266

ABSTRACT

Among all of the known biological carcinogens, Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) are two of the classical oncogenic herpesviruses known to induce the oncogenic phenotype. Many studies have revealed important functions related to epigenetic alterations of the EBV and KSHV genomes that mediate oncogenesis, but the detailed mechanisms are not fully understood. It is also challenging to fully describe the critical cellular events that drive oncogenesis as well as a comprehensive map of the molecular contributors. This review introduces the roles of epigenetic modifications of these viral genomes, including DNA methylation, histone modification, chromatin remodeling, and noncoding RNA expression, and elucidates potential strategies utilized for inducing oncogenesis by these human gammaherpesviruses.


Subject(s)
Carcinogenesis/genetics , Epigenesis, Genetic , Gammaherpesvirinae/genetics , Genome, Viral , Herpesviridae Infections/virology , Tumor Virus Infections/virology , Gammaherpesvirinae/classification , Gammaherpesvirinae/pathogenicity , Herpesviridae Infections/complications , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics , Humans , Tumor Virus Infections/complications , Virus Latency/genetics
11.
J Virol ; 94(22)2020 10 27.
Article in English | MEDLINE | ID: mdl-32847849

ABSTRACT

Epstein-Barr virus (EBV) was discovered as the first human tumor virus more than 50 years ago. EBV infects more than 90% of the human population worldwide and is associated with numerous hematologic malignancies and epithelial malignancies. EBV establishes latent infection in B cells, which is the typical program seen in lymphomagenesis. Understanding EBV-mediated transcription regulatory networks is one of the current challenges that will uncover new insights into the mechanism of viral-mediated lymphomagenesis. Here, we describe the regulatory profiles of several cellular factors (E2F6, E2F1, Rb, HDAC1, and HDAC2) together with EBV latent nuclear antigens using next-generation sequencing (NGS) analysis. Our results show that the E2F-Rb-HDAC complex exhibits similar distributions in genomic regions of EBV-positive cells and is associated with oncogenic super-enhancers involving long-range regulatory regions. Furthermore, EBV latent antigens cooperatively hijack this complex to bind at KLFs gene loci and facilitate KLF14 gene expression in lymphoblastoid cell lines (LCLs). These results demonstrate that EBV latent antigens can function as master regulators of this multisubunit repressor complex (E2F-Rb-HDAC) to reverse its suppressive activities and facilitate downstream gene expression that can contribute to viral-induced lymphomagenesis. These results provide novel insights into targets for the development of new therapeutic interventions for treating EBV-associated lymphomas.IMPORTANCE Epstein-Barr virus (EBV), as the first human tumor virus, infects more than 90% of the human population worldwide and is associated with numerous human cancers. Exploring EBV-mediated transcription regulatory networks is critical to understand viral-associated lymphomagenesis. However, the detailed mechanism is not fully explored. Now we describe the regulatory profiles of the E2F-Rb-HDAC complex together with EBV latent antigens, and we found that EBV latent antigens cooperatively facilitate KLF14 expression by antagonizing this multisubunit repressor complex in EBV-positive cells. This provides potential therapeutic targets for the treatment of EBV-associated cancers.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , B-Lymphocytes/virology , Cell Line , E2F1 Transcription Factor , E2F6 Transcription Factor , Epstein-Barr Virus Nuclear Antigens , Gene Expression Regulation, Viral , Herpesvirus 4, Human/pathogenicity , Histone Deacetylase 1 , Histone Deacetylase 2 , Humans , Latent Infection , Retinoblastoma Protein , Viral Proteins/metabolism , Virus Latency
12.
Commun Biol ; 3(1): 267, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32461675

ABSTRACT

Development of novel PI3K inhibitors is an important strategy to overcome their resistance and poor tolerability in clinical trials. The quassinoid family member Brusatol shows specific inhibitory activity against hematologic malignancies. However, the mechanism of its anti-cancer activity is unknown. We investigated the anti-cancer activity of Brusatol on multiple hematologic malignancies derived cell lines. The results demonstrated that the PI3Kγ isoform was identified as a direct target of Brusatol, and inhibition was dramatically reduced on cells with lower PI3Kγ levels. Novel synthetic analogs were also developed and tested in vitro and in vivo. They shared comparable or superior potency in their ability to inhibit malignant hematologic cell lines, and in a xenograft transplant mouse model. One unique analog had minimal toxicity to normal human cells and in a mouse model. These new analogs have enhanced potential for development as a new class of PI3K inhibitors for treatment of hematologic malignancies.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/genetics , Hematologic Neoplasms/drug therapy , Quassins/pharmacology , Animals , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Hematologic Neoplasms/genetics , Heterografts , Isoenzymes , Male , Mice , Mice, Inbred NOD , Transplantation, Heterologous
13.
PLoS Pathog ; 15(6): e1007796, 2019 06.
Article in English | MEDLINE | ID: mdl-31226160

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous oncogenic virus that induces many cancers. N6-Methyladenosine (m6A) modification regulates many cellular processes. We explored the role of m6A in EBV gene regulation and associated cancers. We have comprehensively defined m6A modification of EBV latent and lytic transcripts. Furthermore, m6A modification demonstrated a functional role in regulation of the stability of viral transcripts. The methyltransferase METTL14 was induced at the transcript and protein levels, and knock-down of METTL14 led to decreased expression of latent EBV transcripts. METTL14 was also significantly induced in EBV-positive tumors, promoted growth of EBV-transformed cells and tumors in Xenograft animal models. Mechanistically, the viral-encoded latent oncoprotein EBNA3C activated transcription of METTL14, and directly interacted with METTL14 to promote its stability. This demonstrated that EBV hijacks METTL14 to drive EBV-mediated tumorigenesis. METTL14 is now a new target for development of therapeutics for treatment of EBV-associated cancers.


Subject(s)
Cell Transformation, Viral , Epstein-Barr Virus Infections/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Herpesvirus 4, Human/metabolism , Methyltransferases/biosynthesis , Neoplasm Proteins/biosynthesis , Neoplasms/metabolism , Adenosine/analogs & derivatives , Adenosine/genetics , Adenosine/metabolism , Animals , Cell Line, Tumor , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , HEK293 Cells , Humans , Male , Methyltransferases/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/virology
14.
PLoS Pathog ; 15(4): e1007732, 2019 04.
Article in English | MEDLINE | ID: mdl-30964921

ABSTRACT

[This corrects the article DOI: 10.1371/journal.ppat.1007253.].

15.
PLoS Pathog ; 15(1): e1007514, 2019 01.
Article in English | MEDLINE | ID: mdl-30615685

ABSTRACT

EBV latent antigen 3C (EBNA3C) is essential for EBV-induced primary B-cell transformation. Infection by EBV induces hypermethylation of a number of tumor suppressor genes, which contributes to the development of human cancers. The Ras association domain family isoform 1A (RASSF1A) is a cellular tumor suppressor, which regulates a broad range of cellular functions, including apoptosis, cell-cycle arrest, mitotic arrest, and migration. However, the expression of RASSF1A is lost in many human cancers by epigenetic silencing. In the present study, we showed that EBNA3C promoted B-cell transformation by specifically suppressing the expression of RASSF1A. EBNA3C directly interacted with RASSF1A and induced RASSF1A degradation via the ubiquitin-proteasome-dependent pathway. SCFSkp2, an E3-ubiquitin ligase, was recruited by EBNA3C to enhance RASSF1A degradation. Moreover, EBNA3C decreased the transcriptional activity of RASSF1A promoter by enhancing its methylation through EBNA3C-mediated modulation of DNMTs expression. EBNA3C also inhibited RASSF1A-mediated cell apoptosis, disrupted RASSF1A-mediated microtubule and chromosomal stability, and promoted cell proliferation by upregulating Cyclin D1 and Cyclin E expression. Our data provides new details, which sheds light on additional mechanisms by which EBNA3C can induce B-cell transformation. This will also facilitate the development of novel therapeutic approaches through targeting of the RASSF1A pathway.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Tumor Suppressor Proteins/genetics , Antigens, Viral/genetics , Apoptosis , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Viral/genetics , DNA Methylation/genetics , Down-Regulation , Epigenesis, Genetic/genetics , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Humans , Lymphocyte Activation/genetics , Promoter Regions, Genetic/genetics , Tumor Suppressor Proteins/metabolism
16.
Cancer Treat Res ; 177: 81-103, 2019.
Article in English | MEDLINE | ID: mdl-30523622

ABSTRACT

Herpesvirus-induced disease is one of the most lethal factors which leads to high mortality in HIV/AIDS patients. EBV, also known as human herpesvirus 4, can transform naive B cells into immortalized cells in vitro through the regulation of cell cycle, cell proliferation, and apoptosis. EBV infection is associated with several lymphoma and epithelial cancers in humans, which occurs at a much higher rate in immune deficient individuals than in healthy people, demonstrating that the immune system plays a vital role in inhibiting EBV activities. EBV latency infection proteins can mimic suppression cytokines or upregulate PD-1 on B cells to repress the cytotoxic T cells response. Many malignancies, including Hodgkin Lymphoma and non-Hodgkin's lymphomas occur at a much higher frequency in EBV positive individuals than in EBV negative people during the development of HIV infection. Importantly, understanding EBV pathogenesis at the molecular level will aid the development of novel therapies for EBV-induced diseases in HIV/AIDS patients.


Subject(s)
Acquired Immunodeficiency Syndrome , Herpesvirus 4, Human , Neoplasms , Acquired Immunodeficiency Syndrome/physiopathology , Acquired Immunodeficiency Syndrome/virology , Carcinogenesis , Coinfection/virology , Epstein-Barr Virus Infections/physiopathology , Epstein-Barr Virus Infections/virology , HIV Infections/physiopathology , HIV Infections/virology , Herpesvirus 4, Human/physiology , Humans , Immunocompromised Host , Lymphoma, AIDS-Related/physiopathology , Lymphoma, AIDS-Related/virology , Neoplasms/physiopathology , Neoplasms/virology
17.
PLoS Pathog ; 14(9): e1007253, 2018 09.
Article in English | MEDLINE | ID: mdl-30212568

ABSTRACT

Shugoshin-1 (Sgo1) protects the integrity of the centromeres, and H2A phosphorylation is critical for this process. The mitotic checkpoint kinase Bub1, phosphorylates H2A and ensures fidelity of chromosome segregation and chromosome number. Oncogenic KSHV induces genetic alterations through chromosomal instability (CIN), and its essential antigen LANA regulates Bub1. We show that LANA inhibits Bub1 phosphorylation of H2A and Cdc20, important for chromosome segregation and mitotic signaling. Inhibition of H2A phosphorylation at residue T120 by LANA resulted in dislocation of Sgo1, and cohesin from the centromeres. Arrest of Cdc20 phosphorylation also rescued degradation of Securin and Cyclin B1 at mitotic exit, and interaction of H2A, and Cdc20 with Bub1 was inhibited by LANA. The N-terminal nuclear localization sequence domain of LANA was essential for LANA and Bub1 interaction, reversed LANA inhibited phosphorylation of H2A and Cdc20, and attenuated LANA-induced aneuploidy and cell proliferation. This molecular mechanism whereby KSHV-induced CIN, demonstrated that the NNLS of LANA is a promising target for development of anti-viral therapies targeting KSHV associated cancers.


Subject(s)
Aneuploidy , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cell Cycle Proteins/metabolism , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/pathogenicity , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Antigens, Viral/chemistry , Cdc20 Proteins/metabolism , Cell Cycle Checkpoints , Cell Line , Centromere/metabolism , Chromosomal Instability , Cyclin B1/metabolism , Herpesvirus 8, Human/genetics , Histones/metabolism , Humans , Mitosis , Models, Biological , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Nuclear Proteins/chemistry , Phosphorylation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Securin/metabolism
18.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29997218

ABSTRACT

Cell cycle regulation is one of the hallmarks of virus-mediated oncogenesis. Epstein-Barr virus (EBV)-induced lymphomas express a repertoire of essential viral latent proteins that regulate expression of cell cycle-related proteins to dysregulate this process, thereby facilitating the proliferation of infected cells. We now demonstrate that the essential EBV latent protein 3C (EBNA3C) stabilizes cyclin D2 to regulate cell cycle progression. More specifically, EBNA3C directly binds to cyclin D2 and they colocalize together in nuclear compartments. We show that EBNA3C regulates the promoter of cyclin D2 through cooperation with master transcription factor Bcl6 and enhances its stability by inhibiting its ubiquitin-dependent degradation. EBNA3C also promoted cell proliferation in the presence of cyclin D2, suggesting that cyclin D2 contributes to EBNA3C-mediated cell cycle progression. These results provide new clues as to the role of this essential viral latent protein and its ability to regulate expression of cellular factors, which drives the oncogenic process.IMPORTANCE Epstein-Barr virus (EBV) is the first identified human tumor virus and is associated with a range of human cancers. During EBV-induced lymphomas, the essential viral latent proteins modify the expression of cell cycle-related proteins to disturb the cell cycle process, thereby facilitating the proliferative process. The essential EBV nuclear antigen 3C (EBNA3C) plays an important role in EBV-mediated B-cell transformation. Here we show that EBNA3C stabilizes cyclin D2 to regulate cell cycle progression. More specifically, EBNA3C directly binds to cyclin D2, and they colocalize together in nuclear compartments. EBNA3C enhances cyclin D2 stability by inhibiting its ubiquitin-dependent degradation and significantly promotes cell proliferation in the presence of cyclin D2. Our results provide novel insights into the function of EBNA3C on cell progression by regulating the cyclin D2 protein and raise the possibility of the development of new anticancer therapies against EBV-associated cancers.


Subject(s)
Cell Proliferation/genetics , Cyclin D2/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Expression Regulation , B-Lymphocytes/virology , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell Transformation, Viral , Herpesvirus 4, Human/physiology , Humans
19.
PLoS Pathog ; 14(5): e1007062, 2018 05.
Article in English | MEDLINE | ID: mdl-29746587

ABSTRACT

Kaposi's sarcoma associated herpesvirus (KSHV) infection stabilizes hypoxia inducible factors (HIFs). The interaction between KSHV encoded factors and HIFs plays a critical role in KSHV latency, reactivation and associated disease phenotypes. Besides modulation of large-scale signaling, KSHV infection also reprograms the metabolic activity of infected cells. However, the mechanism and cellular pathways modulated during these changes are poorly understood. We performed comparative RNA sequencing analysis on cells with stabilized hypoxia inducible factor 1 alpha (HIF1α) of KSHV negative or positive background to identify changes in global and metabolic gene expression. Our results show that hypoxia induces glucose dependency of KSHV positive cells with high glucose uptake and high lactate release. We identified the KSHV-encoded vGPCR, as a novel target of HIF1α and one of the main viral antigens of this metabolic reprogramming. Bioinformatics analysis of vGPCR promoter identified 9 distinct hypoxia responsive elements which were activated by HIF1α in-vitro. Expression of vGPCR alone was sufficient for induction of changes in the metabolic phenotype similar to those induced by KSHV under hypoxic conditions. Silencing of HIF1α rescued the hypoxia associated phenotype of KSHV positive cells. Analysis of the host transcriptome identified several common targets of hypoxia as well as KSHV encoded factors and other synergistically activated genes belonging to cellular pathways. These include those involved in carbohydrate, lipid and amino acids metabolism. Further DNA methyltranferases, DNMT3A and DNMT3B were found to be regulated by either KSHV, hypoxia, or both synergistically at the transcript and protein levels. This study showed distinct and common, as well as synergistic effects of HIF1α and KSHV-encoded proteins on metabolic reprogramming of KSHV-infected cells in the hypoxia.


Subject(s)
B-Lymphocytes/virology , Herpesvirus 8, Human/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , B-Lymphocytes/metabolism , Blotting, Western , Gene Expression Regulation, Viral , Glucose/metabolism , Herpesvirus 8, Human/genetics , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Lactic Acid/metabolism , Leukocytes, Mononuclear/virology , Metabolome , Microscopy, Confocal , Phenotype , Promoter Regions, Genetic , RNA, Viral/chemistry , Reactive Oxygen Species/analysis , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sequence Analysis, RNA , Transcriptional Activation
20.
Adv Exp Med Biol ; 1018: 57-74, 2017.
Article in English | MEDLINE | ID: mdl-29052132

ABSTRACT

Epstein-Barr virus (EBV) was the first human tumor virus discovered more than 50 years ago. EBV-associated lymphomagenesis is still a significant viral-associated disease as it involves a diverse range of pathologies, especially B-cell lymphomas. Recent development of high-throughput next-generation sequencing technologies and in vivo mouse models have significantly promoted our understanding of the fundamental molecular mechanisms which drive these cancers and allowed for the development of therapeutic intervention strategies. This review will highlight the current advances in EBV-associated B-cell lymphomas, focusing on transcriptional regulation, chromosome aberrations, in vivo studies of EBV-mediated lymphomagenesis, as well as the treatment strategies to target viral-associated lymphomas.


Subject(s)
Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , Lymphoma, B-Cell/genetics , Animals , Chromosome Aberrations , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/therapy , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Humans , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/virology , Mice , Transcriptional Activation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...