Publications

2008

Hwang, Seungmin, Ting-Ting Wu, Leming M Tong, Kyeong Seon Kim, DeeAnn Martinez-Guzman, Arnaud D Colantonio, Christel H Uittenbogaart, and Ren Sun. (2008) 2008. “Persistent Gammaherpesvirus Replication and Dynamic Interaction With the Host in Vivo”. Journal of Virology 82 (24): 12498-509. https://doi.org/10.1128/JVI.01152-08.

Gammaherpesviruses establish life-long persistency inside the host and cause various diseases during their persistent infection. However, the systemic interaction between the virus and host in vivo has not been studied in individual hosts continuously, although such information can be crucial to control the persistent infection of the gammaherpesviruses. For the noninvasive and continuous monitoring of the interaction between gammaherpesvirus and the host, a recombinant murine gammaherpesvirus 68 (MHV-68, a gammaherpesvirus 68) was constructed to express a firefly luciferase gene driven by the viral M3 promoter (M3FL). Real-time monitoring of M3FL infection revealed novel sites of viral replication, such as salivary glands, as well as acute replication in the nose and the lung and progression to the spleen. Continuous monitoring of M3FL infection in individual mice demonstrated the various kinetics of transition to different organs and local clearance, rather than systemically synchronized clearance. Moreover, in vivo spontaneous reactivation of M3FL from latency was detected after the initial clearance of acute infection and can be induced upon treatment with either a proteasome inhibitor Velcade or an immunosuppressant cyclosporine A. Taken together, our results demonstrate that the in vivo replication and reactivation of gammaherpesvirus are dynamically controlled by the locally defined interaction between the virus and the host immune system and that bioluminescence imaging can be successfully used for the real-time monitoring of this dynamic interaction of MHV-68 with its host in vivo.

Sanchez, David Jesse, Daniel Miranda, Vaithilingaraja Arumugaswami, Seungmin Hwang, Adam E Singer, Ashkon Senaati, Arash Shahangian, Moon Jung Song, Ren Sun, and Genhong Cheng. (2008) 2008. “A Repetitive Region of Gammaherpesvirus Genomic DNA Is a Ligand for Induction of Type I Interferon”. Journal of Virology 82 (5): 2208-17.

Innate immune responses against viral infection, especially the induction of type I interferon, are critical for limiting the replication of the virus. Although it has been shown that DNA can induce type I interferon, to date no natural DNA ligand of a virus that induces type I interferon has been described. Here we screened the genome of murine gammaherpesvirus 68 with mutations at various genomic locations to map the region of DNA that induces type I interferon. A repetitive region termed the 100-base-pair repeat region is a ligand that is both necessary and sufficient for the viral genomic DNA to induce type I interferon. A region colinear with this ligand in the genome of Kaposi's sarcoma-associated herpesvirus also induces type I interferon. We have thus defined a repetitive region of the genomes of gammaherpesviruses as the first natural DNA virus ligand that induces type I interferon.

2007

Tarakanova, Vera L, Van Leung-Pineda, Seungmin Hwang, Chiao-Wen Yang, Katie Matatall, Mickael Basson, Ren Sun, Helen Piwnica-Worms, Barry P Sleckman, and Herbert W Virgin. (2007) 2007. “Gamma-Herpesvirus Kinase Actively Initiates a DNA Damage Response by Inducing Phosphorylation of H2AX to Foster Viral Replication”. Cell Host & Microbe 1 (4): 275-86.

DNA virus infection can elicit the DNA damage response in host cells, including ATM kinase activation and H2AX phosphorylation. This is considered to be the host cell response to replicating viral DNA. In contrast, we show that during infection of macrophages murine gamma-herpesvirus 68 (gammaHV68) actively induces H2AX phosphorylation by expressing a viral kinase (orf36). GammaHV68-encoded orf36 kinase and its EBV homolog, BGLF4, induce H2AX phosphorylation independently of other viral genes. The process requires the kinase domain of Orf36 and is enhanced by ATM. Orf36 is important for gammaHV68 replication in infected animals, and orf36, H2AX, and ATM are all critical for efficient gammaHV68 replication in primary macrophages. Thus, activation of proximal components of the DNA damage signaling response is an active viral kinase-driven strategy required for efficient gamma-herpesvirus replication.

Lee, Sangmi, Hye-Jeong Cho, Jung-Jin Park, Yong-Sun Kim, Seungmin Hwang, Ren Sun, and Moon Jung Song. (2007) 2007. “The ORF49 Protein of Murine Gammaherpesvirus 68 Cooperates With RTA in Regulating Virus Replication”. Journal of Virology 81 (18): 9870-7.

Our functional mapping study of murine gammaherpesvirus 68 (MHV-68, or gammaHV-68) revealed that a mutant harboring a transposon at the ORF49 locus (ORF49(null)) evidenced a highly attenuated in vitro growth. ORF49 resides adjacent to and in an opposite direction from RTA, the primary switch of the gammaherpesvirus life cycle. A FLAG-tagged ORF49 protein was able to transcomplement ORF49(null), and a revertant of ORF49(null) restored its attenuated growth to a level comparable to that of the wild type. The FLAG-tagged ORF49 protein promoted the ability of RTA to activate downstream target promoters and enhanced virus replication from the ORF50(null) virus in the presence of RTA. Furthermore, ORF49 enhanced wild-type virus replication by increasing the RTA transcript levels. Our data indicate that ORF49 may perform an important function in MHV-68 replication in cooperation with RTA.

2005

Rickabaugh, Tammy M, Helen J Brown, Ting-Ting Wu, Moon Jung Song, Seungmin Hwang, Hongyu Deng, Katherine Mitsouras, and Ren Sun. (2005) 2005. “Kaposi’s Sarcoma-Associated Herpesvirus/Human Herpesvirus 8 RTA Reactivates Murine Gammaherpesvirus 68 from Latency”. Journal of Virology 79 (5): 3217-22.

Murine gammaherpesvirus 68 (MHV-68), Kaposi's sarcoma-associated herpesvirus (HHV-8), and Epstein-Barr virus (EBV) are all members of the gammaherpesvirus family, characterized by their ability to establish latency in lymphocytes. The RTA protein, conserved in all gammaherpesviruses, is known to play a critical role in reactivation from latency. Here we report that HHV-8 RTA, not EBV RTA, was able to induce MHV-68 lytic viral proteins and DNA replication and processing and produce viable MHV-68 virions from latently infected cells at levels similar to those for MHV-68 RTA. HHV-8 RTA was also able to activate two MHV-68 lytic promoters, whereas EBV RTA was not. In order to define the domains of RTA responsible for their functional differences in viral promoter activation and initiation of the MHV-68 lytic cycle, chimeric RTA proteins were constructed by exchanging the N-terminal and C-terminal domains of the RTA proteins. Our data suggest that the species specificity of MHV-68 RTA resides in the N-terminal DNA binding domain.

Xie, Michael W, Fulai Jin, Heejun Hwang, Seungmin Hwang, Vikram Anand, Mara C Duncan, and Jing Huang. (2005) 2005. “Insights into TOR Function and Rapamycin Response: Chemical Genomic Profiling by Using a High-Density Cell Array Method”. Proceedings of the National Academy of Sciences of the United States of America 102 (20): 7215-20.

With the advent of complete genome sequences, large-scale functional analyses are generating new excitement in biology and medicine. To facilitate genomewide functional analyses, we developed a high-density cell array with quantitative and automated readout of cell fitness. Able to print at > x 10 higher density on a standard microtiter plate area than currently possible, our cell array allows single-plate screening of the complete set of Saccharomyces cerevisiae gene-deletion library and significantly reduces the amount of small molecules and other materials needed for the study. We used this method to map the relation between genes and cell fitness in response to rapamycin, a medically important natural product that targets the eukaryotic kinase Tor. We discuss the implications for pharmacogenomics and the uncharted complexity in genotype-dependent drug response in molecularly targeted therapies. Our analysis leads to several basic findings, including a class of gene deletions that confer better fitness in the presence of rapamycin. This result provides insights into possible therapeutic uses of rapamycin/CCI-779 in the treatment of neurodegenerative diseases (including Alzheimer's, Parkinson's, and Huntington's diseases), and cautions the possible existence of similar rapamycin-enhanceable mutations in cancer. It is well established in yeast that although TOR2 has a unique rapamycin-insensitive function, TOR1 and TOR2 are interchangeable in the rapamycin-sensitive functions. We show that even the rapamycin-sensitive functions are distinct between TOR1 and TOR2 and map the functional difference to a approximately 120-aa region at the N termini of the proteins. Finally, we discuss using cell-based genomic pattern recognition in designing electronic or optical biosensors.

Song, Moon Jung, Seungmin Hwang, Wendy H Wong, Ting-Ting Wu, Sangmi Lee, Hsiang-I Liao, and Ren Sun. (2005) 2005. “Identification of Viral Genes Essential for Replication of Murine Gamma-Herpesvirus 68 Using Signature-Tagged Mutagenesis”. Proceedings of the National Academy of Sciences of the United States of America 102 (10): 3805-10.

Gamma-herpesviruses, Epstein-Barr virus, and Kaposi's sarcoma-associated herpesvirus are important human pathogens, because they are involved in tumor development. Murine gamma-herpesvirus-68 (MHV-68 or gammaHV-68) has emerged as a small animal model system for the study of gamma-herpesvirus pathogenesis and host-virus interactions. To identify the genes required for viral replication in vitro and in vivo, we generated 1,152 mutants using signature-tagged transposon mutagenesis on an infectious bacterial artificial chromosome of MHV-68. Almost every ORF was mutated by random insertion. For each ORF, a mutant with an insertion proximal to the N terminus of each ORF was examined for the ability to grow in fibroblasts. Our results indicate that 41 genes are essential for in vitro growth, whereas 26 are nonessential and 6 attenuated. Replication-competent mutants were pooled to infect mice, which led to the discovery of ORF 54 being important for MHV-68 to replicate in the lung. This genetic analysis of a tumor-associated herpesvirus at the whole genome level validates signature-tagged transposon mutagenesis screening as an effective genetic system to identify important virulent genes in vivo and define interactions with the host immune system.

2004

Song, Moon Jung, Seungmin Hwang, Wendy Wong, June Round, DeeAnn Martinez-Guzman, Yaron Turpaz, Jie Liang, et al. (2004) 2004. “The DNA Architectural Protein HMGB1 Facilitates RTA-Mediated Viral Gene Expression in Gamma-2 Herpesviruses”. Journal of Virology 78 (23): 12940-50.

Replication and transcription activator (RTA), an immediate-early gene product of gamma-2 herpesviruses including Kaposi's sarcoma-associated herpesvirus (KSHV) and murine gamma herpesvirus 68 (MHV-68), plays a critical role in controlling the viral life cycle. RTA acts as a strong transcription activator for several downstream genes of KSHV and MHV-68 through direct DNA binding, as well as via indirect mechanisms. HMGB1 (also called HMG-1) protein is a highly conserved nonhistone chromatin protein with the ability to bind and bend DNA. HMGB1 protein promoted RTA binding to different RTA target sites in vitro, with greater enhancement to low-affinity sites than to high-affinity sites. Box A or box B and homologues of HMGB1 also enhanced RTA binding to DNA. Transient transfection of HMGB1 stimulated RTA transactivation of RTA-responsive promoters from KSHV and MHV-68. Furthermore, MHV-68 viral gene expression, as well as viral replication, was significantly reduced in HMGB1-deficient cells than in the wild type. This abated viral gene expression was partially restored by HMGB1 transfection into HMGB1(-/-) cells. These results suggest an important function of the DNA architectural protein, HMGB1, in RTA-mediated gene expression, as well as viral replication in gamma-2 herpesviruses.

2003

Hwang, Seungmin, Daeyoup Lee, Yousang Gwack, Hyesun Min, and Joonho Choe. (2003) 2003. “Kaposi’s Sarcoma-Associated Herpesvirus K8 Protein Interacts With HSNF5”. The Journal of General Virology 84 (Pt 3): 665-76. https://doi.org/10.1099/vir.0.18699-0.

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human gammaherpesvirus related to Epstein-Barr virus (EBV) and herpesvirus saimiri. KSHV open reading frame K8 encodes a basic region-leucine zipper protein of 237 aa that homodimerizes. K8 shows significant similarity to the EBV immediate-early protein Zta, a key regulator of EBV reactivation and replication. In this study, a carboxyl-terminal deletion mutant of K8, K8(1-115), that had strong transactivating properties was found. Screening using transcriptionally inactive K8(1-75) showed that K8 interacts and co-localizes with hSNF5, a cellular chromatin-remodelling factor, both in vivo and in vitro. This interaction requires aa 48-183 of hSNF5 and 1-75 of K8. In a yeast expression system, the ability of K8 and K8(1-115) to activate transcription requires the presence of SNF5, the yeast homologue of hSNF5. These data suggest a mechanism by which the SWI-SNF complex is recruited to specific genes. They also suggest that K8 functions as a transcriptional activator under specific conditions and that its transactivation activity requires its interaction with the cellular chromatin remodelling factor hSNF5.

2002

Byun, Hyewon, Yousang Gwack, Seungmin Hwang, and Joonho Choe. (2002) 2002. “Kaposi’s Sarcoma-Associated Herpesvirus Open Reading Frame (ORF) 50 Transactivates K8 and ORF57 Promoters via Heterogeneous Response Elements”. Molecules and Cells 14 (2): 185-91.

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) belongs to the human gammaherpesvirus family that undergoes both lytic and latent life cycles in host cells. Open reading frame (ORF) 50 is the most important protein in reactivation to lytic phase and functions as a strong transcriptional activator of the early and late genes of KSHV. Since transactivation of promoters by ORF50 is achieved via response elements, we have attempted to identify ORF50 response elements in K8 and ORF57 promoters of KSHV by transient transfection assays with deletion mutants. Our data reveal that specific regions within the K8 (74661-74760) and ORF57 (81851-81931) promoters contain ORF50 response elements, which are heterogeneous, unlike those of Epstein-Barr virus and Herpesvirus saimiri. We additionally identify an AP-1 binding site at the ORF57 promoter between 81882 and 81889, and show that AP-1 participates in ORF57 promoter activation by ORF50. Our findings collectively indicate that ORF50 activates various viral proteins through both direct binding and cellular transcriptional factor-mediated mechanisms.