92 results on '"Kenneth M. Kaye"'
Search Results
2. Macrophages drive KSHV B cell latency
- Author
-
Agnieszka Szymula, Gabriela Samayoa-Reyes, Sidney Ogolla, Bing Liu, Shijun Li, Athira George, Nicholas Van Sciver, Rosemary Rochford, J. Pedro Simas, and Kenneth M. Kaye
- Subjects
CP: Immunology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Kaposi’s sarcoma herpesvirus (KSHV) establishes lifelong infection and persists in latently infected B cells. Paradoxically, in vitro B cell infection is inefficient, and cells rapidly die, suggesting the absence of necessary factor(s). KSHV epidemiology unexpectedly mirrors that of malaria and certain helminthic infections, while other herpesviruses are ubiquitous. Elevated circulating monocytes are common in these parasitic infections. Here, we show that KSHV infection of monocytes or M-CSF-differentiated (M2) macrophages is highly efficient. Proteomic analyses demonstrate that infection induces macrophage production of B cell chemoattractants and activating factor. We find that KSHV acts with monocytes or M2 macrophages to stimulate B cell survival, proliferation, and plasmablast differentiation. Further, macrophages drive infected plasma cell differentiation and long-term viral latency. In Kenya, where KSHV is endemic, we find elevated monocyte levels in children with malaria. These findings demonstrate a role for mononuclear phagocytes in KSHV B cell latency and suggest that mononuclear phagocyte abundance may underlie KSHV’s geographic disparity.
- Published
- 2023
- Full Text
- View/download PDF
3. Severe COVID-19 pneumonia leads to post-COVID-19 lung abnormalities on follow-up CT scans
- Author
-
Takuya Hino, Mizuki Nishino, Vladimir I. Valtchinov, Staci Gagne, Elizabeth Gay, Noriaki Wada, Shu Chi Tseng, Bruno Madore, Charles R.G. Guttmann, Kousei Ishigami, Yi Li, David C. Christiani, Gary M. Hunninghake, Bruce D. Levy, Kenneth M. Kaye, and Hiroto Hatabu
- Subjects
COVID-19 ,COVID-19 pneumonia ,COVID-19 related lung abnormalities ,Lung ,Abnormalities ,Chest CT ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Purpose: To investigate the association of the maximal severity of pneumonia on CT scans obtained within 6-week of diagnosis with the subsequent development of post-COVID-19 lung abnormalities (Co-LA). Methods: COVID-19 patients diagnosed at our hospital between March 2020 and September 2021 were studied retrospectively. The patients were included if they had (1) at least one chest CT scan available within 6-week of diagnosis; and (2) at least one follow-up chest CT scan available ≥ 6 months after diagnosis, which were evaluated by two independent radiologists. Pneumonia Severity Categories were assigned on CT at diagnosis according to the CT patterns of pneumonia and extent as: 1) no pneumonia (Estimated Extent, 0%); 2) non-extensive pneumonia (GGO and OP, 40%). Co-LA on follow-up CT scans, categorized using a 3-point Co-LA Score (0, No Co-LA; 1, Indeterminate Co-LA; and 2, Co-LA). Results: Out of 132 patients, 42 patients (32%) developed Co-LA on their follow-up CT scans 6–24 months post diagnosis. The severity of COVID-19 pneumonia was associated with Co-LA: In 47 patients with extensive pneumonia, 33 patients (70%) developed Co-LA, of whom 18 (55%) developed fibrotic Co-LA. In 52 with non-extensive pneumonia, 9 (17%) developed Co-LA: In 33 with no pneumonia, none (0%) developed Co-LA. Conclusions: Higher severity of pneumonia at diagnosis was associated with the increased risk of development of Co-LA after 6–24 months of SARS-CoV-2 infection.
- Published
- 2023
- Full Text
- View/download PDF
4. Primary effusion lymphoma enhancer connectome links super-enhancers to dependency factors
- Author
-
Chong Wang, Luyao Zhang, Liangru Ke, Weiyue Ding, Sizun Jiang, Difei Li, Yohei Narita, Isabella Hou, Jun Liang, Shijun Li, Haipeng Xiao, Eva Gottwein, Kenneth M. Kaye, Mingxiang Teng, and Bo Zhao
- Subjects
Science - Abstract
Primary effusion lymphoma (PEL) has a very poor prognosis. Here, the authors perform H3K27ac HiChIP in PEL cells and generate the PEL enhancer connectome, linking enhancers and promoters in PEL, as well as super-enhancers to dependency factors.
- Published
- 2020
- Full Text
- View/download PDF
5. COVID-19 Vaccination reduced pneumonia severity
- Author
-
Noriaki Wada, Yi Li, Takuya Hino, Staci Gagne, Vladimir I. Valtchinov, Elizabeth Gay, Mizuki Nishino, Bruno Madore, Charles R.G. Guttmann, Sheila Bond, Kousei Ishigami, Gary M. Hunninghake, Bruce D. Levy, Kenneth M. Kaye, David C. Christiani, and Hiroto Hatabu
- Subjects
COVID-19 ,Pneumonia ,CT ,Vaccination ,Lung ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Purpose: To investigate the effect of vaccinations and boosters on the severity of COVID-19 pneumonia on CT scans during the period of Delta and Omicron variants. Methods: Retrospectively studied were 303 patients diagnosed with COVID-19 between July 2021 and February 2022, who had obtained at least one CT scan within 6 weeks around the COVID-19 diagnosis (−2 to +4 weeks). The severity of pneumonia was evaluated with a 6-point scale Pneumonia Score. The association between demographic and clinical data and vaccination status (booster/additional vaccination, complete vaccination and un-vaccination) and the difference between Pneumonia Scores by vaccination status were investigated. Results: Of 303 patients (59.4 ± 16.3 years; 178 females), 62 (20 %) were in the booster/additional vaccination group, 117 (39 %) in the complete vaccination group, and 124 (41 %) in the unvaccinated group. Interobserver agreement of the Pneumonia Score was high (weighted kappa score = 0.875). Patients in the booster/additionally vaccinated group tended to be older (P = 0.0085) and have more underlying comorbidities (P
- Published
- 2022
- Full Text
- View/download PDF
6. Kaposi’s Sarcoma Herpesvirus Genome Persistence
- Author
-
Franceline Juillard, Min Tan, Shijun Li, and Kenneth M. Kaye
- Subjects
KSHV ,DNA binding ,latency-associated nuclear antigen (LANA) ,chromosome ,viral persistence ,Microbiology ,QR1-502 - Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) has an etiologic role in Kaposi’s sarcoma, primary effusion lymphoma and multicentric Castleman’s disease. These diseases are most common in immunocompromised individuals, especially those with AIDS. Similar to all herpesviruses, KSHV infection is lifelong. KSHV infection in tumor cells is primarily latent, with only a small subset of cells undergoing lytic infection. During latency, the KSHV genome persists as a multiple copy, extrachromosomal episome in the nucleus. In order to persist in proliferating tumor cells, the viral genome replicates once per cell cycle and then segregates to daughter cell nuclei. KSHV only expresses several genes during latent infection. Prominent among these genes, is the latency-associated nuclear antigen (LANA). LANA is responsible for KSHV genome persistence and also exerts transcriptional regulatory effects. LANA mediates KSHV DNA replication and in addition, is responsible for segregation of replicated genomes to daughter nuclei. LANA serves as a molecular tether, bridging the viral genome to mitotic chromosomes to ensure that KSHV DNA reaches progeny nuclei. N-terminal LANA attaches to mitotic chromosomes by binding histones H2A/H2B at the surface of the nucleosome. C-terminal LANA binds specific KSHV DNA sequence and also has a role in chromosome attachment. In addition to the essential roles of N- and C-terminal LANA in genome persistence, internal LANA sequence is also critical for efficient episome maintenance. LANA’s role as an essential mediator of virus persistence makes it an attractive target for inhibition in order to prevent or treat KSHV infection and disease.
- Published
- 2016
- Full Text
- View/download PDF
7. Viral Infection, Pulmonary Fibrosis, and Long COVID
- Author
-
Hiroto Hatabu, Kenneth M. Kaye, and David C. Christiani
- Subjects
Pulmonary and Respiratory Medicine ,Critical Care and Intensive Care Medicine - Published
- 2023
- Full Text
- View/download PDF
8. MLL1 is regulated by KSHV LANA and is important for virus latency
- Author
-
Bing Liu, Ángel L Álvarez, Qiming Sun, J. Pedro Simas, Min Tan, Franceline Juillard, Rute Chitas, Han Xue, Agnieszka Szymula, Colin E. McVey, Shijun Li, Tânia F Custódio, Kenneth M. Kaye, Jing Huang, She Chen, and Veritati - Repositório Institucional da Universidade Católica Portuguesa
- Subjects
Gene Expression Regulation, Viral ,AcademicSubjects/SCI00010 ,Protein Conformation ,viruses ,Biology ,Crystallography, X-Ray ,Virus ,Antigen ,Cell Line, Tumor ,Virus latency ,Genetics ,medicine ,WDR5 ,Humans ,Epigenetics ,Molecular Biology ,Antigens, Viral ,Sarcoma, Kaposi ,Intracellular Signaling Peptides and Proteins ,virus diseases ,Nuclear Proteins ,MLL1 complex ,Histone-Lysine N-Methyltransferase ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Cell biology ,Virus Latency ,Histone methyltransferase ,Gene Knockdown Techniques ,DNA, Viral ,Herpesvirus 8, Human ,Host-Pathogen Interactions ,H3K4me3 ,Myeloid-Lymphoid Leukemia Protein ,Protein Binding - Abstract
Mixed lineage leukemia 1 (MLL1) is a histone methyltransferase. Kaposi's sarcoma-associated herpesvirus (KSHV) is a leading cause of malignancy in AIDS. KSHV latently infects tumor cells and its genome is decorated with epigenetic marks. Here, we show that KSHV latency-associated nuclear antigen (LANA) recruits MLL1 to viral DNA where it establishes H3K4me3 modifications at the extensive KSHV terminal repeat elements during primary infection. LANA interacts with MLL1 complex members, including WDR5, integrates into the MLL1 complex, and regulates MLL1 activity. We describe the 1.5-Å crystal structure of N-terminal LANA peptide complexed with MLL1 complex member WDR5, which reveals a potential regulatory mechanism. Disruption of MLL1 expression rendered KSHV latency establishment highly deficient. This deficiency was rescued by MLL1 but not by catalytically inactive MLL1. Therefore, MLL1 is LANA regulable and exerts a central role in virus infection. These results suggest broad potential for MLL1 regulation, including by non-host factors.
- Published
- 2021
9. Reply to Kamp et al.: Novel Insight into Pulmonary Fibrosis and Long Covid
- Author
-
Hiroto Hatabu, Kenneth M. Kaye, and David C. Christiani
- Subjects
Pulmonary and Respiratory Medicine ,Critical Care and Intensive Care Medicine - Published
- 2023
- Full Text
- View/download PDF
10. ViroPanel
- Author
-
Matthew D. Ducar, Glenn J. Hanna, Anwesha Nag, Paul Van Hummelen, Winslow Powers, Kenneth M. Kaye, Jingwei Cheng, Gabriel J. Starrett, Aaron R. Thorner, Michael K. Slevin, Bruce M. Wollison, Danielle K. Manning, Elizabeth P. Garcia, Laura E. MacConaill, James A. DeCaprio, Neil A. Patel, and Robert Burns
- Subjects
0301 basic medicine ,Massive parallel sequencing ,Computational biology ,Biology ,medicine.disease_cause ,Genome ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Real-time polymerase chain reaction ,030220 oncology & carcinogenesis ,Genotype ,medicine ,Molecular Medicine ,Immunohistochemistry ,Carcinogenesis ,Virus Integration ,Oncovirus - Abstract
Precision cancer medicine aims to classify tumors by site, histology, and molecular testing to determine an individualized profile of cancer alterations. Viruses are a major contributor to oncogenesis, causing 12% to 20% of all human cancers. Several viruses are causal of specific types of cancer, promoting dysregulation of signaling pathways and resulting in carcinogenesis. In addition, integration of viral DNA into the host (human) genome is a hallmark of some viral species. Tests for the presence of viral infection used in the clinical setting most often use quantitative PCR or immunohistochemical staining. Both approaches have limitations and need to be interpreted/scored appropriately. In some cases, results are not binary (virus present/absent), and it is unclear what to do with a weakly or partially positive result. In addition, viral testing of cancers is performed separately from tests to detect human genomic alterations and can thus be time-consuming and use limited valuable specimen. We present a hybrid-capture and massively parallel sequencing approach to detect viral infection that is integrated with targeted genomic analysis to provide a more complete tumor profile from a single sample.
- Published
- 2020
- Full Text
- View/download PDF
11. Cellular microRNA-127-3p suppresses oncogenic herpesvirus-induced transformation and tumorigenesis via down-regulation of SKP2
- Author
-
Soo Mi Lee, Frank J. Slack, and Kenneth M. Kaye
- Subjects
Cyclin E ,Carcinogenesis ,Cyclin A ,Mice, Nude ,medicine.disease_cause ,Cyclin-dependent kinase ,medicine ,Animals ,Humans ,E2F ,Transcription factor ,S-Phase Kinase-Associated Proteins ,Sarcoma, Kaposi ,Multidisciplinary ,biology ,Oncogene ,Cyclin-dependent kinase 2 ,Biological Sciences ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Cell Transformation, Neoplastic ,Herpesvirus 8, Human ,biology.protein ,Cancer research ,Female - Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) causes the endothelial tumor KS, a leading cause of morbidity and mortality in sub-Saharan Africa. KSHV-encoded microRNAs (miRNAs) are known to play an important role in viral oncogenesis; however, the role of host miRNAs in KS tumorigenesis remains largely unknown. Here, high-throughput small-RNA sequencing of the cellular transcriptome in a KS xenograft model revealed miR-127-3p as one of the most significantly down-regulated miRNAs, which we validated in KS patient tissues. We show that restoration of miR-127-3p suppresses KSHV-driven cellular transformation and proliferation and induces G(1) cell cycle arrest by directly targeting the oncogene SKP2. This miR-127-3p–induced G(1) arrest is rescued by disrupting the miR-127-3p target site in SKP2 messenger RNA (mRNA) using gene editing. Mechanistically, miR-127-3p–mediated SKP2 repression elevates cyclin-dependent kinase (CDK) inhibitor p21(Cip1) and down-regulates cyclin E, cyclin A, and CDK2, leading to activation of the RB protein tumor suppressor pathway and suppression of the transcriptional activities of E2F and Myc, key oncoprotein transcription factors crucial for KSHV tumorigenesis. Consequently, metabolomics analysis during miR-127-3p–induced cell cycle arrest revealed significant depletion of dNTP pools, consistent with RB-mediated repression of key dNTP biosynthesis enzymes. Furthermore, miR-127-3p reconstitution in a KS xenograft mouse model suppresses KSHV-positive tumor growth by targeting SKP2 in vivo. These findings identify a previously unrecognized tumor suppressor function for miR-127-3p in KS and demonstrate that the miR-127-3p/SKP2 axis is a viable therapeutic strategy for KS.
- Published
- 2021
12. KSHV LANA acetylation-selective acidic domain reader sequence mediates virus persistence
- Author
-
André F. Seixas, Aura Franco, Ángel L. Álvarez, Marta Pires de Miranda, Bing Liu, Agnieszka Szymula, Franceline Juillard, J. Pedro Simas, Min Tan, Shijun Li, Kenneth M. Kaye, and Veritati - Repositório Institucional da Universidade Católica Portuguesa
- Subjects
viruses ,Mutant ,Acidic domain reader ,Biology ,Microbiology ,Virus ,Protein–protein interaction ,Chimera (genetics) ,Mice ,Acetylation-regulated interaction ,medicine ,Animals ,Humans ,Gene ,Antigens, Viral ,Latency-associated nuclear antigen ,Multidisciplinary ,Germinal center ,Nuclear Proteins ,virus diseases ,Kaposi's sarcoma herpesvirus ,Acetylation ,Biological Sciences ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Virus persistence ,Bromodomain ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,Kaposi´s sarcoma herpesvirus ,DNA, Viral ,Herpesvirus 8, Human ,Female ,Primary effusion lymphoma ,Plasmids - Abstract
Significance Understanding how viruses modulate host cell function is central to prospects for potential therapies. A recently described mechanism mediates selective protein interactions between acidic domain readers and unacetylated, lysine-rich regions, opposite of bromodomain function. Here we show that the latency-associated nuclear antigen (LANA) encoded by Kaposi´s sarcoma-associated herpesvirus (KSHV) contains a SET oncoprotein homologous acidic domain reader that interacts with p53 in an acetylation-dependent manner. Significantly, the LANA acidic domain reader is critical for viral latency and persistent infection. The finding that KSHV has evolved a mechanism for acetylation-dependent protein interaction underscores the physiological importance of this cellular regulatory posttranslational modification. Furthermore, opportunity may exist to develop inhibitors that disrupt LANA acidic domain reader function for therapy of KSHV-associated tumors., Viruses modulate biochemical cellular pathways to permit infection. A recently described mechanism mediates selective protein interactions between acidic domain readers and unacetylated, lysine-rich regions, opposite of bromodomain function. Kaposi´s sarcoma (KS)-associated herpesvirus (KSHV) is tightly linked with KS, primary effusion lymphoma, and multicentric Castleman’s disease. KSHV latently infects cells, and its genome persists as a multicopy, extrachromosomal episome. During latency, KSHV expresses a small subset of genes, including the latency-associated nuclear antigen (LANA), which mediates viral episome persistence. Here we show that LANA contains two tandem, partially overlapping, acidic domain sequences homologous to the SET oncoprotein acidic domain reader. This domain selectively interacts with unacetylated p53, as evidenced by reduced LANA interaction after overexpression of CBP, which acetylates p53, or with an acetylation mimicking carboxyl-terminal domain p53 mutant. Conversely, the interaction of LANA with an acetylation-deficient p53 mutant is enhanced. Significantly, KSHV LANA mutants lacking the acidic domain reader sequence are deficient for establishment of latency and persistent infection. This deficiency was confirmed under physiological conditions, on infection of mice with a murine gammaherpesvirus 68 chimera expressing LANA, where the virus was highly deficient in establishing latent infection in germinal center B cells. Therefore, LANA’s acidic domain reader is critical for viral latency. These results implicate an acetylation-dependent mechanism mediating KSHV persistence and expand the role of acidic domain readers.
- Published
- 2020
13. Identification of a nucleoside analog active against adenosine kinase–expressing plasma cell malignancies
- Author
-
Denise Hernandez-Hopkins, Kenneth M. Kaye, Yifang Liu, J. David Warren, Jennifer Totonchy, Ethel Cesarman, Ruben Niesvizky, Ilaria Guasparri, Peter Barelli, David Jayabalan, Hufeng Zhou, Olivier Elemento, Duane C. Hassane, Michelle A. Sahai, Robert H. Shoemaker, Gunkut Akar, Utthara Nayar, Shizuko Sei, Jonathan Reichel, and Jouliana Sadek
- Subjects
Male ,0301 basic medicine ,Cell Survival ,T cell ,Antineoplastic Agents ,Mice, SCID ,Adenosine kinase ,Biology ,Plasma cell ,Inhibitory Concentration 50 ,Mice ,03 medical and health sciences ,Mice, Inbred NOD ,Cell Line, Tumor ,Lymphoma, Primary Effusion ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Adenosine Kinase ,Purine Nucleosides ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,ADK ,Lymphoma ,030104 developmental biology ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Primary effusion lymphoma ,Nucleoside ,Plasmablastic lymphoma ,Research Article - Abstract
Primary effusion lymphoma (PEL) is a largely incurable malignancy of B cell origin with plasmacytic differentiation. Here, we report the identification of a highly effective inhibitor of PEL. This compound, 6-ethylthioinosine (6-ETI), is a nucleoside analog with toxicity to PEL in vitro and in vivo, but not to other lymphoma cell lines tested. We developed and performed resistome analysis, an unbiased approach based on RNA sequencing of resistant subclones, to discover the molecular mechanisms of sensitivity. We found different adenosine kinase-inactivating (ADK-inactivating) alterations in all resistant clones and determined that ADK is required to phosphorylate and activate 6-ETI. Further, we observed that 6-ETI induces ATP depletion and cell death accompanied by S phase arrest and DNA damage only in ADK-expressing cells. Immunohistochemistry for ADK served as a biomarker approach to identify 6-ETI-sensitive tumors, which we documented for other lymphoid malignancies with plasmacytic features. Notably, multiple myeloma (MM) expresses high levels of ADK, and 6-ETI was toxic to MM cell lines and primary specimens and had a robust antitumor effect in a disseminated MM mouse model. Several nucleoside analogs are effective in treating leukemias and T cell lymphomas, and 6-ETI may fill this niche for the treatment of PEL, plasmablastic lymphoma, MM, and other ADK-expressing cancers.
- Published
- 2017
- Full Text
- View/download PDF
14. Primary cytomegalovirus infection with invasive disease in a patient with inflammatory bowel disease
- Author
-
Jingyi Gong, Raymond A. Isidro, Kenneth M. Kaye, and Eric A. Meyerowitz
- Subjects
Adult ,medicine.medical_specialty ,Fever ,Gastrointestinal Diseases ,Congenital cytomegalovirus infection ,Opportunistic Infections ,Inflammatory bowel disease ,Gastroenterology ,Antiviral Agents ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Ganciclovir ,Gastrointestinal tract ,Unusual Presentation of More Common Disease/Injury ,business.industry ,Headache ,Valganciclovir ,General Medicine ,Myalgia ,Viral Load ,medicine.disease ,Inflammatory Bowel Diseases ,Mercaptopurine ,Cytomegalovirus infection ,Treatment Outcome ,030220 oncology & carcinogenesis ,Cytomegalovirus Infections ,030211 gastroenterology & hepatology ,Female ,medicine.symptom ,business ,Odynophagia ,Viral load ,medicine.drug - Abstract
A 37-year-old woman with a history of inflammatory bowel disease on mercaptopurine presented with a week of recurrent fever, headache, myalgias and mildly elevated serum transaminases and leucopenia. Her workup revealed primary cytomegalovirus (CMV) infection with atypical lymphocytosis, elevated viral load, positive IgM and negative IgG. Two weeks after her initial presentation, she developed odynophagia and diarrhoea prompting endoscopic evaluation with biopsies, which demonstrated CMV disease of the gastrointestinal tract. Her fever and systemic symptoms improved rapidly with initiation of intravenous ganciclovir. She was transitioned to and maintained on oral valganciclovir until two and half months after discharge when her symptoms and lab abnormalities had fully subsided.
- Published
- 2019
15. ViroPanel: Hybrid Capture and Massively Parallel Sequencing for Simultaneous Detection and Profiling of Oncogenic Virus Infection and Tumor Genome
- Author
-
Michael K, Slevin, Bruce M, Wollison, Winslow, Powers, Robert T, Burns, Neil, Patel, Matthew D, Ducar, Gabriel J, Starrett, Elizabeth P, Garcia, Danielle K, Manning, Jingwei, Cheng, Glenn J, Hanna, Kenneth M, Kaye, Paul, Van Hummelen, Anwesha, Nag, Aaron R, Thorner, James A, DeCaprio, and Laura E, MacConaill
- Subjects
Genotype ,Genome, Human ,Virus Integration ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Genome, Viral ,Genomics ,Cell Transformation, Viral ,Polymorphism, Single Nucleotide ,Sensitivity and Specificity ,Article ,Tumor Virus Infections ,Neoplasms ,Humans ,Precision Medicine ,Phylogeny - Abstract
Precision cancer medicine aims to classify tumors by site, histology, and molecular testing to determine an individualized profile of cancer alterations. Viruses are a major contributor to oncogenesis, causing 12% to 20% of all human cancers. Several viruses are causal of specific types of cancer, promoting dysregulation of signaling pathways and resulting in carcinogenesis. In addition, integration of viral DNA into the host (human) genome is a hallmark of some viral species. Tests for the presence of viral infection used in the clinical setting most often use quantitative PCR or immunohistochemical staining. Both approaches have limitations and need to be interpreted/scored appropriately. In some cases, results are not binary (virus present/absent), and it is unclear what to do with a weakly or partially positive result. In addition, viral testing of cancers is performed separately from tests to detect human genomic alterations and can thus be time-consuming and use limited valuable specimen. We present a hybrid-capture and massively parallel sequencing approach to detect viral infection that is integrated with targeted genomic analysis to provide a more complete tumor profile from a single sample.
- Published
- 2019
16. NDRG1 facilitates the replication and persistence of Kaposi’s sarcoma-associated herpesvirus by interacting with the DNA polymerase clamp PCNA
- Author
-
Xiaozhen Liang, Kenneth M. Kaye, Deguang Liang, Ke Lan, Zhe Zou, Xing Wang, Rui Sun, Xiaoxi Lin, and Fang Zhang
- Subjects
Male ,Molecular biology ,viruses ,Cell Cycle Proteins ,DNA-Directed DNA Polymerase ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Virus Replication ,Biochemistry ,Virions ,Virus latency ,Medicine and Health Sciences ,Biology (General) ,Antigens, Viral ,0303 health sciences ,Viral Genomics ,biology ,030302 biochemistry & molecular biology ,Intracellular Signaling Peptides and Proteins ,virus diseases ,Nuclear Proteins ,Kaposi's Sarcoma-Associated Herpesvirus ,Genomics ,Herpesviridae Infections ,Cell cycle ,3. Good health ,Cell biology ,Viral Persistence and Latency ,Up-Regulation ,Virus Latency ,Nucleic acids ,Medical Microbiology ,Viral Pathogens ,Viruses ,Herpesvirus 8, Human ,Viral Genome ,Pathogens ,Viral genome replication ,Research Article ,Plasmids ,Adult ,DNA Replication ,Herpesviruses ,QH301-705.5 ,Immunology ,Microbial Genomics ,Genome, Viral ,DNA construction ,Viral Structure ,Transfection ,Microbiology ,Cell Line ,03 medical and health sciences ,Host chromosome ,Virology ,Proliferating Cell Nuclear Antigen ,medicine ,Genetics ,Humans ,Kaposi's sarcoma-associated herpesvirus ,Microbial Pathogens ,Sarcoma, Kaposi ,030304 developmental biology ,Cell Nucleus ,Biology and life sciences ,DNA replication ,Organisms ,DNA ,RC581-607 ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Viral Replication ,Proliferating cell nuclear antigen ,Research and analysis methods ,Molecular biology techniques ,HEK293 Cells ,Viral replication ,Plasmid Construction ,DNA, Viral ,biology.protein ,Parasitology ,Immunologic diseases. Allergy ,DNA viruses - Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) latently infects host cells and establishes lifelong persistence as an extra-chromosomal episome in the nucleus. To persist in proliferating cells, the viral genome typically replicates once per cell cycle and is distributed into daughter cells. This process involves host machinery utilized by KSHV, however the underlying mechanisms are not fully elucidated. In present study, we found that N-Myc downstream regulated gene 1 (NDRG1), a cellular gene known to be non-detectable in primary B cells and endothelial cells which are the major cell types for KSHV infection in vivo, was highly upregulated by KSHV in these cells. We further demonstrated that the high expression of NDRG1 was regulated by latency-associated nuclear antigen (LANA), the major viral latent protein which tethers the viral genome to host chromosome and plays an essential role in viral genome maintenance. Surprisingly, knockdown of NDRG1 in KSHV latently infected cells resulted in a significant decrease of viral genome copy number in these cells. Interestingly, NDRG1 can directly interact with proliferating cell nuclear antigen (PCNA), a cellular protein which functions as a DNA polymerase clamp during DNA replication. Intriguingly, we found that NDRG1 forms a complex with LANA and PCNA and serves as a scaffold protein bridging these two proteins. We further demonstrated that NDRG1 is critical for mediating LANA to recruit PCNA onto terminal repeat (TR) of KSHV genome, and facilitates viral DNA replication and episome persistence. Taken together, our findings suggest that NDRG1 plays an important role in KSHV viral genome replication, and provide new clues for understanding of KSHV persistence., Author summary KSHV latently infects cells and establishes lifelong persistence, but the underlying mechanisms of this process has not been fully elucidated. Here, we find a novel host protein NDRG1 is highly up-regulated by KSHV infection and the viral protein LANA is essential in this process. NDRG1 is a multiple functional protein, but the role in KSHV infection remains unknown. Our findings show that NDRG1 functions as a scaffold protein that forms a complex with PCNA and LANA, thereby helping LANA load PCNA onto the viral genome and facilitating the replication and persistence of KSHV. Since NDRG1 is non-detectable in primary B cells and endothelial cells, which are the major cell types susceptible to KSHV infection in vivo, NDRG1 might be a candidate of therapeutic target for inhibition of KSHV persistence in malignant cells.
- Published
- 2019
17. Electrical response of a B lymphoma cell line latently infected with Kaposi’s sarcoma herpesvirus
- Author
-
Mohammadali Safavieh, Hadi Shafiee, Franceline Juillard, Sultan Khetani, Kenneth M. Kaye, Vivasvat Kaul, and Manoj Kumar Kanakasabapathy
- Subjects
Extramural ,010401 analytical chemistry ,Cell ,Biomedical Engineering ,Biophysics ,02 engineering and technology ,General Medicine ,Biology ,021001 nanoscience & nanotechnology ,medicine.disease ,01 natural sciences ,Virology ,0104 chemical sciences ,Lymphoma ,Signal frequency ,medicine.anatomical_structure ,Cell culture ,Electrochemistry ,medicine ,0210 nano-technology ,Kaposi's sarcoma ,Gene ,Biotechnology - Abstract
Certain viruses, such as herpesviruses, are capable of persistent and latent infection of host cells. Distinguishing and separating live, latently infected cells from uninfected cells is not easily attainable using current approaches. The ability to perform such separation would greatly enhance the ability to study primary, infected cells and potentially enable elimination of latently infected cells from the host. Here, the dielectrophoretic response of B cells infected with Kaposi's sarcoma-associated herpesvirus (KSHV) were investigated and compared to uninfected B cells. We evaluated the effect of applied voltage, signal frequency, and flow rate of the sample on the cell capture efficiency. We achieved 37.1% ± 8.5% difference in capture efficiencies between latently KSHV-infected and uninfected BJAB B lymphoma cells at the chip operational conditions of 1V, 50 kHz and 0.02 μl/min sample flow rate. Our results show that latently infected B lymphoma cells demonstrated significantly different electrical response compared to uninfected B cells and DEP-based microchips can be potentially used for sorting latently infected cells based on their electrical properties.
- Published
- 2016
- Full Text
- View/download PDF
18. Kaposi's Sarcoma-Associated Herpesvirus LANA-Adjacent Regions with Distinct Functions in Episome Segregation or Maintenance
- Author
-
Franceline Juillard, Shijun Li, Kenneth M. Kaye, Min Tan, and Erika De León Vázquez
- Subjects
DNA Replication ,Cell division ,viruses ,Immunology ,Mutant ,Mitosis ,Genome, Viral ,medicine.disease_cause ,Virus Replication ,Microbiology ,Chromosomes ,Cell Line ,chemistry.chemical_compound ,Viral Proteins ,Plasmid ,Virology ,Cell Line, Tumor ,medicine ,Humans ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,Sarcoma, Kaposi ,Cell Nucleus ,biology ,DNA replication ,Terminal Repeat Sequences ,virus diseases ,Nuclear Proteins ,Antigens, Nuclear ,biochemical phenomena, metabolism, and nutrition ,Cell biology ,Virus Latency ,Virus-Cell Interactions ,Histone ,HEK293 Cells ,chemistry ,Insect Science ,DNA, Viral ,Herpesvirus 8, Human ,biology.protein ,DNA ,Plasmids - Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA) is a 1,162-amino-acid protein that mediates episome persistence of viral genomes. LANA binds the KSHV terminal-repeat (TR) sequence through its carboxy-terminal domain to mediate DNA replication. LANA simultaneously binds mitotic chromosomes and TR DNA to segregate virus genomes to daughter cell nuclei. Amino-terminal LANA attaches to chromosomes by binding histones H2A/H2B, and carboxy-terminal LANA contributes to mitotic-chromosome binding. Although amino- and carboxy-terminal LANA are essential for episome persistence, they are not sufficient, since deletion of all internal LANA sequence renders LANA highly deficient for episome maintenance. Internal LANA sequence upstream of the internal repeat elements contributes to episome segregation and persistence. Here, we investigate this region with a panel of LANA deletion mutants. Mutants retained the ability to associate with mitotic chromosomes and bind TR DNA. In contrast to prior results, deletion of most of this sequence did not reduce LANA’s ability to mediate DNA replication. Deletions of upstream sequence within the region compromised segregation of TR DNA to daughter cells, as assessed by retention of green fluorescent protein (GFP) expression from a replication-deficient TR plasmid. However, deletion of this upstream sequence did not reduce episome maintenance. In contrast, deletions that included an 80-amino-acid sequence immediately downstream resulted in highly deficient episome persistence. LANA with this downstream sequence deleted maintained the ability to replicate and segregate TR DNA, suggesting a unique role for the residues. Therefore, this work identifies adjacent LANA regions with distinct roles in episome segregation and persistence. IMPORTANCE KSHV LANA mediates episomal persistence of viral genomes. LANA binds the KSHV terminal-repeat (TR) sequence to mediate DNA replication and tethers KSHV DNA to mitotic chromosomes to segregate genomes to daughter cell nuclei. Here, we investigate LANA sequence upstream of the internal repeat elements that contributes to episome segregation and persistence. Mutants with deletions within this sequence maintained the ability to bind mitotic chromosomes or bind and replicate TR DNA. Deletion of upstream sequence within the region reduced segregation of TR DNA to daughter cells, but not episome maintenance. In contrast, mutants with deletions of 80 amino acids immediately downstream were highly deficient for episome persistence yet maintained the ability to replicate and segregate TR DNA, the two principal components of episome persistence, suggesting another role for the residues. In summary, this work identifies adjacent LANA sequence with distinct roles in episome segregation and persistence.
- Published
- 2018
19. In Vivo Persistence of Chimeric Virus after Substitution of the Kaposi's Sarcoma-Associated Herpesvirus LANA DNA Binding Domain with That of Murid Herpesvirus 4
- Author
-
J. Pedro Simas, Marta Pires de Miranda, Colin E. McVey, Kenneth M. Kaye, and Ana Patrícia Quendera
- Subjects
0301 basic medicine ,LANA ,Murid herpesvirus 4 ,viruses ,Immunology ,DNA-binding domain ,Biology ,medicine.disease_cause ,biology.organism_classification ,Microbiology ,Virology ,Fusion protein ,Kaposi's sarcoma-associated herpesvirus ,03 medical and health sciences ,030104 developmental biology ,Lytic cycle ,Insect Science ,Extrachromosomal DNA ,medicine ,Pathogenesis and Immunity ,MLANA ,murid herpesvirus 4 ,Oncovirus - Abstract
KSHV is a human oncogenic virus for which there is no tractable, immunocompetent animal model of infection. MuHV-4, a related rodent gammaherpesvirus, enables pathogenesis studies in mice. In latency, both viruses persist as extrachromosomal, circular genomes (episomes). LANA proteins encoded by KSHV (kLANA) and MuHV-4 (mLANA) contain a C-terminal DNA binding domain (DBD) that acts on the virus terminal repeats to enable episome persistence. mLANA is a smaller protein than kLANA. Their DBDs are structurally conserved but differ strikingly in the conformation of DNA binding. We report a recombinant, chimeric MuHV-4 which contains kLANA in place of mLANA, but in which the DBD is replaced with that of mLANA. Results showed that kLANA functionally accommodated mLANA's mode of DNA binding. In fact, the new chimeric virus established latency in vivo more efficiently than MuHV-4 expressing full-length kLANA., The latency-associated nuclear antigen from Kaposi's sarcoma-associated herpesvirus (KSHV), kLANA, and its homolog from the murid herpesvirus 4 (MuHV-4), mLANA, are essential for viral latency. kLANA is nearly four times the size of mLANA, mainly due to an extensive central repeat region that is absent in mLANA. Both proteins harbor a C-terminal DNA binding domain (DBD). The DBD binds the terminal repeat (TR) DNA sequences of the viral genome to mediate persistence. Despite structural conservation, the kLANA and mLANA DBDs differ in sequence and mode of oligomerization. kLANA DBD oligomers are flexible and bent, while mLANA DBD oligomers bind DNA in a rigid, linear conformation. We previously reported that kLANA and mLANA acted reciprocally on TR sequences. Furthermore, a MuHV-4 expressing kLANA instead of mLANA (v-kLANA) established latency in mice, albeit at a lower magnitude than the wild-type (WT) virus. Here, we asked if kLANA can accommodate the mLANA DBD and generated a fusion protein which contains kLANA but with the mLANA C-terminal region in place of that of kLANA. We report a recombinant MuHV-4 (v-KM) encoding this LANA fusion protein instead of mLANA. The fusion protein was expressed in lytic infection in vitro and assembled nuclear LANA dots in infected splenocytes. Results demonstrated that kLANA functionally accommodated mLANA's mode of DNA binding, allowing MuHV-4 chimeric virus to establish latency in vivo. Notably, v-KM established latency in germinal center B cells more efficiently than did v-kLANA, although levels were reduced compared to WT MuHV-4. IMPORTANCE KSHV is a human oncogenic virus for which there is no tractable, immunocompetent animal model of infection. MuHV-4, a related rodent gammaherpesvirus, enables pathogenesis studies in mice. In latency, both viruses persist as extrachromosomal, circular genomes (episomes). LANA proteins encoded by KSHV (kLANA) and MuHV-4 (mLANA) contain a C-terminal DNA binding domain (DBD) that acts on the virus terminal repeats to enable episome persistence. mLANA is a smaller protein than kLANA. Their DBDs are structurally conserved but differ strikingly in the conformation of DNA binding. We report a recombinant, chimeric MuHV-4 which contains kLANA in place of mLANA, but in which the DBD is replaced with that of mLANA. Results showed that kLANA functionally accommodated mLANA's mode of DNA binding. In fact, the new chimeric virus established latency in vivo more efficiently than MuHV-4 expressing full-length kLANA.
- Published
- 2018
20. KSHV but not MHV-68 LANA induces a strong bend upon binding to terminal repeat viral DNA
- Author
-
Maria Arménia Carrondo, Tania F. Custodio, J. Pedro Simas, Bruno Correia, Colin E. McVey, Maxim V. Petoukhov, Kenneth M. Kaye, Min Tan, Franceline Juillard, Rajesh Ponnusamy, Dmitri I. Svergun, Marta Pires de Miranda, and Repositório da Universidade de Lisboa
- Subjects
Models, Molecular ,Rhadinovirus ,viruses ,Plasma protein binding ,Biology ,medicine.disease_cause ,DNA-binding protein ,03 medical and health sciences ,chemistry.chemical_compound ,Tetramer ,Structural Biology ,Genetics ,medicine ,MLANA ,Binding site ,Nuclear protein ,Antigens, Viral ,030304 developmental biology ,0303 health sciences ,Mutation ,Binding Sites ,030302 biochemistry & molecular biology ,Terminal Repeat Sequences ,virus diseases ,Nuclear Proteins ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,Cell biology ,Protein Structure, Tertiary ,chemistry ,ddc:540 ,DNA, Viral ,Herpesvirus 8, Human ,Nucleic Acid Conformation ,Thermodynamics ,Protein Multimerization ,DNA ,Protein Binding - Abstract
© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research. This is an Open Access article distributed under the terms of the Creative Commons Attribution License http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited., Latency-associated nuclear antigen (LANA) is central to episomal tethering, replication and transcriptional regulation of γ2-herpesviruses. LANA binds cooperatively to the terminal repeat (TR) region of the viral episome via adjacent LANA binding sites (LBS), but the molecular mechanism by which LANA assembles on the TR remains elusive. We show that KSHV LANA and MHV-68 LANA proteins bind LBS DNA using strikingly different modes. Solution structure of LANA complexes revealed that while kLANA tetramer is intrinsically bent both in the free and bound state to LBS1-2 DNA, mLANA oligomers instead adopt a rigid linear conformation. In addition, we report a novel non-ring kLANA structure that displays more flexibility at its assembly interface than previously demonstrated. We identified a hydrophobic pivot point located at the dimer-dimer assembly interface, which gives rotational freedom for kLANA to adopt variable conformations to accommodate both LBS1-2 and LBS2-1-3 DNA. Alterations in the arrangement of LBS within TR or at the tetramer assembly interface have a drastic effect on the ability of kLANA binding. We also show kLANA and mLANA DNA binding functions can be reciprocated. Although KSHV and MHV-68 are closely related, the findings provide new insights into how the structure, oligomerization, and DNA binding of LANA have evolved differently to assemble on the TR DNA., Fundação para a Ciência e a Tecnologia (FCT) Investigator Grant [IF/01023/2013 to C.E.M.]; Harvard Medical School Portugal Program in Translational Research and Information [HMSP-ICT/0021/2010 to J.P.S., C.E.M., M.A.C. and K.M.K.]; National Cancer Institute/National Institutes of Health (NCI/NIH) [CA082036 to KMK]; National Institutes of Dental and Craniofacial Research/National Institutes of Health (NIDCR/NIH) [DE025208 to K.M.K.]; European Community's Seventh Framework Programme (FP7/2007–2013) under BioStruct-X [283570]. This work was also supported by Instruct, part of the European Strategy Forum on Research Infrastructures (ESFRI) and through national member agreements. Funding for open access charges: FCT.
- Published
- 2015
- Full Text
- View/download PDF
21. Functional Heterogeneity in the CD4+ T Cell Response to Murine γ-Herpesvirus 68
- Author
-
Marcia A. Blackman, Zhuting Hu, Edward J. Usherwood, and Kenneth M. Kaye
- Subjects
CD4-Positive T-Lymphocytes ,Rhadinovirus ,Cell Survival ,T cell ,Immunology ,Apoptosis ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,T-Lymphocytes, Regulatory ,Article ,Interferon-gamma ,Interleukin 21 ,medicine ,Animals ,Antigens, Ly ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Interleukin 3 ,Mice, Knockout ,B-Lymphocytes ,Tumor Necrosis Factor-alpha ,ZAP70 ,Cell Differentiation ,Herpesviridae Infections ,Th1 Cells ,Flow Cytometry ,Natural killer T cell ,Cell biology ,Mice, Inbred C57BL ,Tumor Virus Infections ,medicine.anatomical_structure ,Host-Pathogen Interactions ,Spleen - Abstract
CD4+ T cells are critical for the control of virus infections, T cell memory, and immune surveillance. We studied the differentiation and function of murine γ-herpesvirus 68 (MHV-68)–specific CD4+ T cells using gp150-specific TCR-transgenic mice. This allowed a more detailed study of the characteristics of the CD4+ T cell response than did previously available approaches for this virus. Most gp150-specific CD4+ T cells expressed T-bet and produced IFN-γ, indicating that MHV-68 infection triggered differentiation of CD4+ T cells largely into the Th1 subset, whereas some became follicular Th cells and Foxp3+ regulatory T cells. These CD4+ T cells were protective against MHV-68 infection in the absence of CD8+ T cells and B cells, and protection depended on IFN-γ secretion. Marked heterogeneity was observed in the CD4+ T cells, based on lymphocyte Ag 6C (Ly6C) expression. Ly6C expression positively correlated with IFN-γ, TNF-α, and granzyme B production; T-bet and KLRG1 expression; proliferation; and CD4+ T cell–mediated cytotoxicity. Ly6C expression inversely correlated with survival, CCR7 expression, and secondary expansion potential. Ly6C+ and Ly6C− gp150-specific CD4+ T cells were able to interconvert in a bidirectional manner upon secondary Ag exposure in vivo. These results indicate that Ly6C expression is closely associated with antiviral activity in effector CD4+ T cells but is inversely correlated with memory potential. Interconversion between Ly6C+ and Ly6C− cells may maintain a balance between the two Ag-specific CD4+ T cell populations during MHV-68 infection. These findings have significant implications for Ly6C as a surface marker to distinguish functionally distinct CD4+ T cells during persistent virus infection.
- Published
- 2015
- Full Text
- View/download PDF
22. Cross-species conservation of episome maintenance provides a basis for in vivo investigation of Kaposi's sarcoma herpesvirus LANA
- Author
-
Colin E. McVey, Sofia A. Cerqueira, Bruno Correia, Rajesh Ponnusamy, Chantal Beauchemin, Edward J. Usherwood, J. Pedro Simas, Min Tan, Aline C. Habison, Marta Pires de Miranda, Kenneth M. Kaye, and Repositório da Universidade de Lisboa
- Subjects
0301 basic medicine ,B Cells ,Physiology ,viruses ,Artificial Gene Amplification and Extension ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Polymerase Chain Reaction ,Biochemistry ,White Blood Cells ,Mice ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,Antigens, Viral ,lcsh:QH301-705.5 ,B-Lymphocytes ,Nuclear Proteins ,Transfection ,Kaposi's Sarcoma-Associated Herpesvirus ,Viral Persistence and Latency ,Virus Latency ,Nucleic acids ,Medical Microbiology ,Viral Pathogens ,Viruses ,Herpesvirus 8, Human ,Cellular Types ,Pathogens ,Research Article ,Plasmids ,lcsh:Immunologic diseases. Allergy ,Herpesviruses ,Immune Cells ,Immunology ,Genome, Viral ,Biology ,DNA replication ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Virology ,medicine ,Genetics ,Animals ,MLANA ,Kaposi's sarcoma-associated herpesvirus ,Antibody-Producing Cells ,Molecular Biology Techniques ,Gene ,Kaposi's sarcoma ,Microbial Pathogens ,Molecular Biology ,Sarcoma, Kaposi ,Blood Cells ,Organisms ,Germinal center ,Biology and Life Sciences ,Cell Biology ,DNA ,medicine.disease ,Germinal Center ,Viral Replication ,030104 developmental biology ,Viral replication ,lcsh:Biology (General) ,DNA, Viral ,Parasitology ,DNA viruses ,lcsh:RC581-607 ,Spleen - Abstract
Copyright: © 2017 Habison et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited., Many pathogens, including Kaposi's sarcoma herpesvirus (KSHV), lack tractable small animal models. KSHV persists as a multi-copy, nuclear episome in latently infected cells. KSHV latency-associated nuclear antigen (kLANA) binds viral terminal repeat (kTR) DNA to mediate episome persistence. Model pathogen murine gammaherpesvirus 68 (MHV68) mLANA acts analogously on mTR DNA. kLANA and mLANA differ substantially in size and kTR and mTR show little sequence conservation. Here, we find kLANA and mLANA act reciprocally to mediate episome persistence of TR DNA. Further, kLANA rescued mLANA deficient MHV68, enabling a chimeric virus to establish latent infection in vivo in germinal center B cells. The level of chimeric virus in vivo latency was moderately reduced compared to WT infection, but WT or chimeric MHV68 infected cells had similar viral genome copy numbers as assessed by immunofluorescence of LANA intranuclear dots or qPCR. Thus, despite more than 60 Ma of evolutionary divergence, mLANA and kLANA act reciprocally on TR DNA, and kLANA functionally substitutes for mLANA, allowing kLANA investigation in vivo. Analogous chimeras may allow in vivo investigation of genes of other human pathogens., This work was supported in part by National Institutes of Health grants CA082036 (NCI), DE025208, and DE024971 (both NIDCR), to KMK, FCT PTDC/IMI-MIC/0980/2014 to JPS, FCT Harvard Medical School Portugal Program in Translational Research (HMSP-ICT/0021/2010) to JPS, KMK, CEM, Instituto de Medicina Molecular Directors Fund to JPS, and iNOVA4Health Research Unit (LISBOA-01-0145-FEDER-007344) FCT/FEDER (PT2020 Partnership Agreement) to CEM. M.P.M is supported by a fellowship from Fundação para a Ciência e Tecnologia (FCT), Portugal.
- Published
- 2017
23. Assay Development and High-Throughput Screening for Inhibitors of Kaposi’s Sarcoma–Associated Herpesvirus N-Terminal Latency-Associated Nuclear Antigen Binding to Nucleosomes
- Author
-
Kenneth M. Kaye, Patrick W. Faloon, Chantal Beauchemin, and Nathan J. Moerke
- Subjects
Erythrocytes ,Cell Survival ,viruses ,Mitosis ,Enzyme-Linked Immunosorbent Assay ,Fluorescence Polarization ,Peptide binding ,medicine.disease_cause ,Antiviral Agents ,Biochemistry ,Article ,Analytical Chemistry ,Histones ,chemistry.chemical_compound ,Plasmid ,Protein Domains ,Extrachromosomal DNA ,medicine ,Animals ,Humans ,Antigens ,Nuclear protein ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,Fluorescent Dyes ,Glutathione Transferase ,biology ,Nuclear Proteins ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,High-Throughput Screening Assays ,Nucleosomes ,Spectrometry, Fluorescence ,Histone ,chemistry ,Drug Design ,Herpesvirus 8, Human ,biology.protein ,Molecular Medicine ,Chickens ,DNA ,HeLa Cells ,Plasmids ,Biotechnology - Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) has a causative role in several human malignancies, especially in immunocompromised hosts. KSHV latently infects tumor cells and persists as an extrachromosomal episome (plasmid). KSHV latency-associated nuclear antigen (LANA) mediates KSHV episome persistence. LANA binds specific KSHV sequence to replicate viral DNA. In addition, LANA tethers KSHV genomes to mitotic chromosomes to efficiently segregate episomes to daughter nuclei after mitosis. N-terminal LANA (N-LANA) binds histones H2A and H2B to attach to chromosomes. Currently, there are no specific inhibitors of KSHV latent infection. To enable high-throughput screening (HTS) of inhibitors of N-LANA binding to nucleosomes, here we develop, miniaturize, and validate a fluorescence polarization (FP) assay that detects fluorophore-labeled N-LANA peptide binding to nucleosomes. We also miniaturize a counterscreen to identify DNA intercalators that nonspecifically inhibit N-LANA binding to nucleosomes, and also develop an enzyme-linked immunosorbent assay to assess N-LANA binding to nucleosomes in the absence of fluorescence. HTS of libraries containing more than 350,000 compounds identified multiple compounds that inhibited N-LANA binding to nucleosomes. No compounds survived all counterscreens, however. More complex small-molecule libraries will likely be necessary to identify specific inhibitors of N-LANA binding to histones H2A and H2B; these assays should prove useful for future screens.
- Published
- 2014
- Full Text
- View/download PDF
24. Timeless-Dependent DNA Replication-Coupled Recombination Promotes Kaposi's Sarcoma-Associated Herpesvirus Episome Maintenance and Terminal Repeat Stability
- Author
-
Kenneth M. Kaye, Paul M. Lieberman, Jayaraju Dheekollu, and Horng-Shen Chen
- Subjects
DNA Replication ,Chromatin Immunoprecipitation ,viruses ,Immunology ,Cell Cycle Proteins ,Biology ,medicine.disease_cause ,Microbiology ,Genomic Instability ,Replication fork protection ,chemistry.chemical_compound ,Minichromosome maintenance ,Virology ,In Situ Nick-End Labeling ,medicine ,Humans ,Electrophoresis, Gel, Two-Dimensional ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,DNA Primers ,Recombination, Genetic ,Intracellular Signaling Peptides and Proteins ,Terminal Repeat Sequences ,DNA replication ,Nuclear Proteins ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Cell cycle ,Flow Cytometry ,Molecular biology ,Electrophoresis, Gel, Pulsed-Field ,Genome Replication and Regulation of Viral Gene Expression ,DNA-Binding Proteins ,Bromodeoxyuridine ,chemistry ,Lytic cycle ,Insect Science ,Herpesvirus 8, Human ,Carrier Proteins ,Chromatin immunoprecipitation ,DNA ,Plasmids - Abstract
Kaposi's Sarcoma-associated herpesvirus (KSHV) is maintained as a stable episome in latently infected pleural effusion lymphoma (PEL) cells. Episome maintenance is conferred by the binding of the KSHV-encoded LANA protein to the viral terminal repeats (TR). Here, we show that DNA replication in the KSHV TR is coupled with DNA recombination and mediated in part through the cellular replication fork protection factors Timeless (Tim) and Tipin. We show by two-dimensional (2D) agarose gel electrophoresis that replication forks naturally stall and form recombination-like structures at the TR during an unperturbed cell cycle. Chromatin immunoprecipitation (ChIP) assays revealed that Tim and Tipin are selectively enriched at the KSHV TR during S phase and in a LANA-dependent manner. Tim depletion inhibited LANA-dependent TR DNA replication and caused the loss of KSHV episomes from latently infected PEL cells. Tim depletion resulted in the aberrant accumulation of recombination structures and arrested MCM helicase at TR. Tim depletion did not induce the KSHV lytic cycle or apoptotic cell death. We propose that KSHV episome maintenance requires Tim-assisted replication fork protection at the viral terminal repeats and that Tim-dependent recombination-like structures form at TR to promote DNA repeat stability and viral genome maintenance.
- Published
- 2013
- Full Text
- View/download PDF
25. 3-D Microwell Array System for Culturing Virus Infected Tumor Cells
- Author
-
Alessandro Tocchio, Utkan Demirci, Alyssa Holman, Thiruppathiraja Chinnasamy, Sebnem Unluisler, Srikar Srivatsa, Pu Chen, Franceline Juillard, Chantal Beauchemin, Umut A. Gurkan, Serli Canikyan, Rami El Assal, and Kenneth M. Kaye
- Subjects
Gene Expression Regulation, Viral ,0301 basic medicine ,Cell ,Cell Culture Techniques ,02 engineering and technology ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Antigen ,Cell Line, Tumor ,Virus latency ,medicine ,Humans ,Antigens, Viral ,B-Lymphocytes ,Tumor microenvironment ,Multidisciplinary ,Nuclear Proteins ,Viral Load ,021001 nanoscience & nanotechnology ,medicine.disease ,Virus Latency ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Lytic cycle ,Cell culture ,Herpesvirus 8, Human ,Cancer cell ,Virus Activation ,0210 nano-technology ,Carcinogenesis - Abstract
Cancer cells have been increasingly grown in pharmaceutical research to understand tumorigenesis and develop new therapeutic drugs. Currently, cells are typically grown using two-dimensional (2-D) cell culture approaches, where the native tumor microenvironment is difficult to recapitulate. Thus, one of the main obstacles in oncology is the lack of proper infection models that recount main features present in tumors. In recent years, microtechnology-based platforms have been employed to generate three-dimensional (3-D) models that better mimic the native microenvironment in cell culture. Here, we present an innovative approach to culture Kaposi’s sarcoma-associated herpesvirus (KSHV) infected human B cells in 3-D using a microwell array system. The results demonstrate that the KSHV-infected B cells can be grown up to 15 days in a 3-D culture. Compared with 2-D, cells grown in 3-D had increased numbers of KSHV latency-associated nuclear antigen (LANA) dots, as detected by immunofluorescence microscopy, indicating a higher viral genome copy number. Cells in 3-D also demonstrated a higher rate of lytic reactivation. The 3-D microwell array system has the potential to improve 3-D cell oncology models and allow for better-controlled studies for drug discovery.
- Published
- 2016
- Full Text
- View/download PDF
26. Latency-Associated Nuclear Antigen E3 Ubiquitin Ligase Activity Impacts Gammaherpesvirus-Driven Germinal Center B Cell Proliferation
- Author
-
Shijun Li, Kenneth M. Kaye, Min Tan, J. Pedro Simas, Colin E. McVey, Sofia A. Cerqueira, and Franceline Juillard
- Subjects
0301 basic medicine ,Rhadinovirus ,viruses ,Viral pathogenesis ,Ubiquitin-Protein Ligases ,Immunology ,DNA Mutational Analysis ,Mutation, Missense ,Microbiology ,03 medical and health sciences ,Mice ,Ubiquitin ,Virology ,Animals ,MLANA ,Antigens, Viral ,Cell Proliferation ,B-Lymphocytes ,030102 biochemistry & molecular biology ,biology ,Murid herpesvirus 4 ,Germinal center ,Nuclear Proteins ,biology.organism_classification ,Germinal Center ,Ubiquitin ligase ,Virus-Cell Interactions ,030104 developmental biology ,Insect Science ,DNA, Viral ,Host-Pathogen Interactions ,biology.protein ,Mutant Proteins ,Cullin ,Protein Binding - Abstract
Viruses have evolved mechanisms to hijack components of cellular E3 ubiquitin ligases, thus modulating the ubiquitination pathway. However, the biological relevance of such mechanisms for viral pathogenesis in vivo remains largely unknown. Here, we utilized murid herpesvirus 4 (MuHV-4) infection of mice as a model system to address the role of MuHV-4 latency-associated nuclear antigen (mLANA) E3 ligase activity in gammaherpesvirus latent infection. We show that specific mutations in the mLANA SOCS box (V199A, V199A/L202A, or P203A/P206A) disrupted mLANA's ability to recruit Elongin C and Cullin 5, thereby impairing the formation of the Elongin BC/Cullin 5/SOCS (EC 5 S mLANA ) complex and mLANA's E3 ligase activity on host NF-κB and Myc. Although these mutations resulted in considerably reduced mLANA binding to viral terminal repeat DNA as assessed by electrophoretic mobility shift assay (EMSA), the mutations did not disrupt mLANA's ability to mediate episome persistence. In vivo , MuHV-4 recombinant viruses bearing these mLANA SOCS box mutations exhibited a deficit in latency amplification in germinal center (GC) B cells. These findings demonstrate that the E3 ligase activity of mLANA contributes to gammaherpesvirus-driven GC B cell proliferation. Hence, pharmacological inhibition of viral E3 ligase activity through targeting SOCS box motifs is a putative strategy to control gammaherpesvirus-driven lymphoproliferation and associated disease. IMPORTANCE The gammaherpesviruses Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) cause lifelong persistent infection and play causative roles in several human malignancies. Colonization of B cells is crucial for virus persistence, and access to the B cell compartment is gained by virus-driven proliferation in germinal center (GC) B cells. Infection of B cells is predominantly latent, with the viral genome persisting as a multicopy episome and expressing only a small subset of viral genes. Here, we focused on latency-associated nuclear antigen (mLANA) encoded by murid herpesvirus-4 (MuHV-4), which exhibits homology in sequence, structure, and function to KSHV LANA (kLANA), thereby allowing the study of LANA-mediated pathogenesis in mice. Our experiments show that mLANA's E3 ubiquitin ligase activity is necessary for efficient expansion of latency in GC B cells, suggesting that the development of pharmacological inhibitors of LANA E3 ubiquitin ligase activity may allow strategies to interfere with gammaherpesvirus-driven lymphoproliferation and associated disease.
- Published
- 2016
27. Kaposi’s Sarcoma Herpesvirus Genome Persistence
- Author
-
Min Tan, Shijun Li, Franceline Juillard, and Kenneth M. Kaye
- Subjects
0301 basic medicine ,Microbiology (medical) ,viruses ,lcsh:QR1-502 ,Review ,KSHV ,latency-associated nuclear antigen (LANA) ,Genome ,Microbiology ,lcsh:Microbiology ,viral persistence ,03 medical and health sciences ,Extrachromosomal DNA ,medicine ,chromosome ,DNA binding ,Kaposi's sarcoma ,Gene ,Genetics ,biology ,virus diseases ,Cell cycle ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Virology ,3. Good health ,030104 developmental biology ,Histone ,Lytic cycle ,latency-associated nuclear antigen ,biology.protein ,Primary effusion lymphoma - Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) has an etiologic role in Kaposi’s sarcoma, primary effusion lymphoma and multicentric Castleman’s disease. These diseases are most common in immunocompromised individuals, especially those with AIDS. Similar to all herpesviruses, KSHV infection is lifelong. KSHV infection in tumor cells is primarily latent, with only a small subset of cells undergoing lytic infection. During latency, the KSHV genome persists as a multiple copy, extrachromosomal episome in the nucleus. In order to persist in proliferating tumor cells, the viral genome replicates once per cell cycle and then segregates to daughter cell nuclei. KSHV only expresses several genes during latent infection. Prominent among these genes, is the latency-associated nuclear antigen (LANA). LANA is responsible for KSHV genome persistence and also exerts transcriptional regulatory effects. LANA mediates KSHV DNA replication and in addition, is responsible for segregation of replicated genomes to daughter nuclei. LANA serves as a molecular tether, bridging the viral genome to mitotic chromosomes to ensure that KSHV DNA reaches progeny nuclei. N-terminal LANA attaches to mitotic chromosomes by binding histones H2A/H2B at the surface of the nucleosome. C-terminal LANA binds specific KSHV DNA sequence and also has a role in chromosome attachment. In addition to the essential roles of N- and C-terminal LANA in genome persistence, internal LANA sequence is also critical for efficient episome maintenance. LANA’s role as an essential mediator of virus persistence makes it an attractive target for inhibition in order to prevent or treat KSHV infection and disease.
- Published
- 2016
28. Murine Gammaherpesvirus 68 LANA Acts on Terminal Repeat DNA To Mediate Episome Persistence
- Author
-
A. C. Habison, Chantal Beauchemin, J. P. Simas, Kenneth M. Kaye, and Edward J. Usherwood
- Subjects
Rhadinovirus ,Immunology ,Biology ,Virus Replication ,Microbiology ,Cell Line ,Mice ,03 medical and health sciences ,Plasmid ,Virology ,Virus latency ,medicine ,Animals ,MLANA ,Nuclear protein ,Antigens, Viral ,Mitosis ,030304 developmental biology ,0303 health sciences ,030306 microbiology ,Terminal Repeat Sequences ,Nuclear Proteins ,Promoter ,biology.organism_classification ,medicine.disease ,Virus Latency ,Genome Replication and Regulation of Viral Gene Expression ,Viral replication ,Insect Science ,DNA, Viral ,Plasmids ,Protein Binding - Abstract
Murine gammaherpesvirus 68 (MHV68) ORF73 (mLANA) has sequence homology to Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA). LANA acts on the KSHV terminal repeat (TR) elements to mediate KSHV episome maintenance. Disruption of mLANA expression severely reduces the ability of MHV68 to establish latent infection in mice, consistent with the possibility that mLANA mediates episome persistence. Here we assess the roles of mLANA and MHV68 TR (mTR) elements in episome persistence. mTR-associated DNA persisted as an episome in latently MHV68-infected tumor cells, demonstrating that the mTR elements can serve as a cis- acting element for MHV68 episome maintenance. In some cases, both control vector and mTR-associated DNAs integrated into MHV68 episomal genomes. Therefore, we also assessed the roles of mTRs as well as mLANA in the absence of infection. DNA containing both mLANA and mTRs in cis persisted as an episome in murine A20 or MEF cells. In contrast, mTR DNA never persisted as an episome in the absence of mLANA. mLANA levels were increased when mLANA was expressed from its native promoters, and episome maintenance was more efficient with higher mLANA levels. Increased numbers of mTRs conferred more efficient episome maintenance, since DNA containing mLANA and eight mTR elements persisted more efficiently in A20 cells than did DNA with mLANA and two or four mTRs. Similar to KSHV LANA, mLANA broadly associated with mitotic chromosomes but relocalized to concentrated dots in the presence of episomes. Therefore, mLANA acts on mTR elements to mediate MHV68 episome persistence.
- Published
- 2012
- Full Text
- View/download PDF
29. Newer Beta-lactam Antibiotics: Doripenem, Ceftobiprole, Ceftaroline, and Cefepime
- Author
-
Jose A. Bazan, Kenneth M. Kaye, and Stanley I. Martin
- Subjects
Microbiology (medical) ,medicine.medical_specialty ,medicine.drug_class ,Cefepime ,Antibiotics ,Ceftobiprole ,Phases of clinical research ,beta-Lactams ,Community-acquired pneumonia ,Internal medicine ,medicine ,Humans ,Dosing ,Intensive care medicine ,Bacteria ,business.industry ,Bacterial Infections ,General Medicine ,medicine.disease ,Anti-Bacterial Agents ,Clinical trial ,Infectious Diseases ,Pharmacodynamics ,Doripenem ,business ,medicine.drug - Abstract
This article reviews the new beta-lactam (beta-lactam) antibiotics doripenem, ceftobiprole, and ceftaroline. It covers pharmacokinetic and pharmacodynamic properties, dosing, in vitro activities, safety, and clinical trial results. Doripenem (Doribax) has been approved by the US Food and Drug Administration (FDA) for the treatment of complicated intra-abdominal and urinary tract infections. At this writing, ceftobiprole is under review by the FDA for approval based on results of phase 3 clinical trials, whereas at least one phase 3 clinical trial of ceftaroline has been completed. The article also reviews recent data suggesting increased overall mortality with Cefepime (Maxipime) use compared with other beta-lactam antibiotics and the potential risk for neurotoxicity in the setting of renal failure.
- Published
- 2011
- Full Text
- View/download PDF
30. Rapid and quantitative assessment of KSHV LANA-mediated DNA replication
- Author
-
Kenneth M. Kaye and Erika De León Vázquez
- Subjects
Gene Expression Regulation, Viral ,viruses ,Mutant ,Biology ,Virus Replication ,Polymerase Chain Reaction ,Article ,law.invention ,chemistry.chemical_compound ,law ,Cell Line, Tumor ,Virology ,Humans ,Antigens, Viral ,Polymerase chain reaction ,Southern blot ,Terminal Repeat Sequences ,DNA replication ,Nuclear Proteins ,virus diseases ,General Medicine ,Transfection ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,Real-time polymerase chain reaction ,Viral replication ,chemistry ,DNA, Viral ,Herpesvirus 8, Human ,DNA - Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA) mediates DNA replication of terminal repeat (TR) DNA to enable viral episome persistence in latently infected cells. Southern blotting is routinely used to detect LANA-replicated DNA. We developed and validated a real-time PCR assay for TR-associated DNA and compared it with Southern blot analysis. Both PCR and Southern blot detected LANA-replicated DNA, but the PCR assay was more rapid and did not require radioisotope. PCR detection at 24 and 72 hours post-transfection demonstrated rapid loss of transfected TR DNA. LANA, and to a lesser extent a moderately deficient LANA mutant, reduced the rate of DNA loss through addition of replicated TR DNA and reduction in the loss of non-replicated DNA, the latter of which is consistent with LANA's nuclear segregation function. Therefore, this work develops a rapid, sensitive, and quantitative PCR (qPCR) assay to detect KSHV LANA-replicated DNA and demonstrates that LANA reduces TR DNA loss after transfection through replication and nuclear partitioning of TR DNA.
- Published
- 2011
- Full Text
- View/download PDF
31. Lytic and Latent Antigens of the Human Gammaherpesviruses Kaposi's Sarcoma-Associated Herpesvirus and Epstein-Barr Virus Induce T-Cell Responses with Similar Functional Properties and Memory Phenotypes
- Author
-
Christian Brander, David T. Scadden, Todd J. Suscovich, Sheila C. Dollard, Tania M. Welzel, Daniel Kaufmann, Kenneth M. Kaye, Elizabeth E. Brown, Jeffrey N. Martin, Fred Wang, John V. Chisholm, Leah M. Henry, Murli Narayan, Florian Bihl, and Tauheed Zaman
- Subjects
Herpesvirus 4, Human ,Cellular immunity ,T-Lymphocytes ,viruses ,T cell ,Immunology ,medicine.disease_cause ,Microbiology ,Virus ,Herpesviridae ,Immunophenotyping ,Epitopes ,Viral Proteins ,Virology ,medicine ,Humans ,Gammaherpesvirinae ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,Immunity, Cellular ,biology ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Epstein–Barr virus ,medicine.anatomical_structure ,Lytic cycle ,Insect Science ,Herpesvirus 8, Human ,Pathogenesis and Immunity ,Immunologic Memory - Abstract
The cellular immunity against Kaposi's sarcoma-associated herpesvirus (KSHV) is poorly characterized and has not been compared to T-cell responses against other human herpesviruses. Here, novel and dominant targets of KSHV-specific cellular immunity are identified and compared to T cells specific for lytic and latent antigens in a second human gammaherpesvirus, Epstein-Barr virus. The data identify a novel HLA-B57- and HLA-B58-restricted epitope in the Orf57 protein and show consistently close parallels in immune phenotypes and functional response patterns between cells targeting lytic or latent KSHV- and EBV-encoded antigens, suggesting common mechanisms in the induction of these responses.
- Published
- 2007
- Full Text
- View/download PDF
32. Determination of Kaposi's Sarcoma-Associated Herpesvirus C-Terminal Latency-Associated Nuclear Antigen Residues Mediating Chromosome Association and DNA Binding
- Author
-
Viswanathan Srinivasan, Mia Kazanjian, Brenna Kelley-Clarke, Takashi Komatsu, Kenneth M. Kaye, Te-Ana Harris, Mary E. Ballestas, and Andrew J. Barbera
- Subjects
DNA Replication ,HMG-box ,viruses ,Molecular Sequence Data ,Immunology ,Mitosis ,Electrophoretic Mobility Shift Assay ,Biology ,Virus Replication ,medicine.disease_cause ,Microbiology ,chemistry.chemical_compound ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Chromosomes, Human ,Humans ,Immunoprecipitation ,Electrophoretic mobility shift assay ,Amino Acid Sequence ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,Mutagenesis ,DNA replication ,Nuclear Proteins ,virus diseases ,Chromosome ,DNA ,DNA-binding domain ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,Virus-Cell Interactions ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Amino Acid Substitution ,chemistry ,Insect Science ,COS Cells ,DNA, Viral ,Herpesvirus 8, Human ,Mutagenesis, Site-Directed ,Protein Binding - Abstract
Kaposi's sarcoma-associated herpesvirus latency-associated nuclear antigen (LANA) tethers viral terminal repeat (TR) DNA to mitotic chromosomes to mediate episome persistence. The 1,162-amino-acid LANA protein contains both N- and C-terminal chromosome attachment regions. The LANA C-terminal domain self-associates to specifically bind TR DNA and mitotic chromosomes. Here, we used alanine scanning substitutions spanning residues 1023 to 1145 to investigate LANA self-association, DNA binding, and C-terminal chromosome association. No residues were essential for LANA oligomerization, as assayed by coimmunoprecipitation experiments, consistent with redundant roles for amino acids in self-association. Different subsets of amino acids were important for DNA binding, as assayed by electrophoretic mobility shift assay, and mitotic chromosome association, indicating that distinct C-terminal LANA subdomains effect DNA and chromosome binding. The DNA binding domains of LANA and EBNA1 are predicted to be structurally homologous; certain LANA residues important for DNA binding correspond to those with roles in EBNA1 DNA binding, providing genetic support for at least partial structural homology. In contrast to the essential role of N-terminal LANA chromosome targeting residues in DNA replication, deficient C-terminal chromosome association did not reduce LANA-mediated DNA replication.
- Published
- 2007
- Full Text
- View/download PDF
33. Multitarget, quantitative nanoplasmonic electrical field-enhanced resonating device (NE2RD) for diagnostics
- Author
-
Hidemi S. Yamamoto, Chiara Filippini, Duane R. Wesemann, Priscilla L. Yang, Yuko Takagi, Michael Vetter, Franceline Juillard, Ping Ping Kuang, Margot Carocci, Daniel R. Kuritzkes, Murat Baday, Mehmet Ozgun Ozen, Emily Hanhauser, Demir Akin, Sanjiv S. Gambhir, Ronald W. Davis, Kristen S. Hobbs, Raina N. Fichorova, Umit Hakan Yildiz, Fatih Inci, Timothy J. Henrich, Lars M. Steinmetz, Naside Gozde Durmus, Amit Singhal, Steven C. Schachter, Utkan Demirci, ShuQi Wang, Kenneth M. Kaye, Semih Calamak, Max L. Nibert, and Daryl T.-Y. Lau
- Subjects
Computer science ,Microfluidics ,Nanotechnology ,Bioengineering ,Enzyme-Linked Immunosorbent Assay ,Biosensing Techniques ,Environment ,label-free ,Cell Line ,Software portability ,Electricity ,multiple biotargets ,Clinical Research ,Limit of Detection ,Cell Line, Tumor ,Wide dynamic range ,Humans ,Fluidics ,Diagnostic Techniques and Procedures ,screening and diagnosis ,Multidisciplinary ,Tumor ,Dynamic range ,Coinfection ,nanoparticle ,biodetection ,Osmolar Concentration ,Temperature ,Reproducibility of Results ,Equipment Design ,Hydrogen-Ion Concentration ,Chip ,4.1 Discovery and preclinical testing of markers and technologies ,Nanostructures ,Detection ,Good Health and Well Being ,PNAS Plus ,Generic health relevance ,point-of-need ,Sensitivity (electronics) ,Biosensor - Abstract
Recent advances in biosensing technologies present great potential for medical diagnostics, thus improving clinical decisions. However, creating a label-free general sensing platform capable of detecting multiple biotargets in various clinical specimens over a wide dynamic range, without lengthy sample-processing steps, remains a considerable challenge. In practice, these barriers prevent broad applications in clinics and at patients' homes. Here, we demonstrate the nanoplasmonic electrical field-enhanced resonating device (NE(2)RD), which addresses all these impediments on a single platform. The NE(2)RD employs an immunodetection assay to capture biotargets, and precisely measures spectral color changes by their wavelength and extinction intensity shifts in nanoparticles without prior sample labeling or preprocessing. We present through multiple examples, a label-free, quantitative, portable, multitarget platform by rapidly detecting various protein biomarkers, drugs, protein allergens, bacteria, eukaryotic cells, and distinct viruses. The linear dynamic range of NE(2)RD is five orders of magnitude broader than ELISA, with a sensitivity down to 400 fg/mL This range and sensitivity are achieved by self-assembling gold nanoparticles to generate hot spots on a 3D-oriented substrate for ultrasensitive measurements. We demonstrate that this precise platform handles multiple clinical samples such as whole blood, serum, and saliva without sample preprocessing under diverse conditions of temperature, pH, and ionic strength. The NE(2)RD's broad dynamic range, detection limit, and portability integrated with a disposable fluidic chip have broad applications, potentially enabling the transition toward precision medicine at the point-of-care or primary care settings and at patients' homes.
- Published
- 2015
34. Printed Flexible Plastic Microchip for Viral Load Measurement through Quantitative Detection of Viruses in Plasma and Saliva
- Author
-
Utkan Demirci, Daniel R. Kuritzkes, Hadi Shafiee, Mert Keser, Magesh Sadasivam, Kenneth M. Kaye, Mehmet Yuksekkaya, Emily Hanhauser, Franceline Juillard, Timothy J. Henrich, and Manoj Kumar Kanakasabapathy
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Saliva ,Point-of-Care Systems ,Human immunodeficiency virus (HIV) ,HIV Infections ,Biology ,medicine.disease_cause ,Article ,Virus ,Herpes virus ,Lab-On-A-Chip Devices ,medicine ,Humans ,Viral load measurement ,Multidisciplinary ,Herpesviridae Infections ,Reference Standards ,Viral Load ,Virology ,3. Good health ,Herpesvirus 8, Human ,HIV-1 ,Viral load ,Biosensor ,Treatment monitoring ,Biomedical engineering - Abstract
We report a biosensing platform for viral load measurement through electrical sensing of viruses on a flexible plastic microchip with printed electrodes. Point-of-care (POC) viral load measurement is of paramount importance with significant impact on a broad range of applications, including infectious disease diagnostics and treatment monitoring specifically in resource-constrained settings. Here, we present a broadly applicable and inexpensive biosensing technology for accurate quantification of bioagents, including viruses in biological samples, such as plasma and artificial saliva, at clinically relevant concentrations. Our microchip fabrication is simple and mass-producible as we print microelectrodes on flexible plastic substrates using conductive inks. We evaluated the microchip technology by detecting and quantifying multiple Human Immunodeficiency Virus (HIV) subtypes (A, B, C, D, E, G and panel), Epstein-Barr Virus (EBV) and Kaposi’s Sarcoma-associated Herpes Virus (KSHV) in a fingerprick volume (50 µL) of PBS, plasma, and artificial saliva samples for a broad range of virus concentrations between 102 copies/mL and 107 copies/mL. We have also evaluated the microchip platform with discarded, de-identified HIV-infected patient samples by comparing our microchip viral load measurement results with reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) as the gold standard method using Bland-Altman Analysis.
- Published
- 2015
- Full Text
- View/download PDF
35. Contributors
- Author
-
Kjersti Aagaard, Fredrick M. Abrahamian, Ban Mishu Allos, David R. Andes, Fred Y. Aoki, Michael A. Apicella, Kevin L. Ard, Cesar A. Arias, David M. Aronoff, Michael H. Augenbraun, Francisco Averhoff, Dimitri T. Azar, Larry M. Baddour, Lindsey R. Baden, Carol J. Baker, Ronald C. Ballard, Gerard R. Barber, Scott D. Barnes, Dan H. Barouch, Alan D. Barrett, Miriam Baron Barshak, Sridhar V. Basavaraju, Byron E. Batteiger, Stephen G. Baum, Arnold S. Bayer, J. David Beckham, Susan E. Beekmann, Beth P. Bell, John E. Bennett, Dennis A. Bente, Elie F. Berbari, Jonathan Berman, Joseph S. Bertino, Adarsh Bhimraj, Holly H. Birdsall, Alan L. Bisno, Brian G. Blackburn, Lucas S. Blanton, Martin J. Blaser, Thomas P. Bleck, Nicole M.A. Blijlevens, David A. Bobak, William Bonnez, John C. Boothroyd, Luciana L. Borio, Patrick J. Bosque, John Bower, Robert W. Bradsher, Itzhak Brook, Kevin E. Brown, Patricia D. Brown, Barbara A. Brown-Elliott, Roberta L. Bruhn, Amy E. Bryant, Eileen M. Burd, Jane C. Burns, Larry M. Bush, Stephen B. Calderwood, Luz Elena Cano, Charles C.J. Carpenter, Mary T. Caserta, Elio Castagnola, Richard E. Chaisson, Henry F. Chambers, Stephen J. Chapman, James D. Chappell, Lea Ann Chen, Sharon C-A Chen, Anthony W. Chow, Rebecca A. Clark, Jeffrey I. Cohen, Myron S. Cohen, Ronit Cohen-Poradosu, Susan E. Cohn, Mark Connors, Lawrence Corey, Mackenzie L. Cottrell, Timothy L. Cover, Heather L. Cox, William A. Craig, Kent B. Crossley, Clyde S. Crumpacker, James W. Curran, Bart J. Currie, Erika D'Agata, Inger K. Damon, Rabih O. Darouiche, Roberta L. DeBiasi, George S. Deepe, Carlos del Rio, Andrew S. Delemos, Frank R. DeLeo, Gregory P. DeMuri, Peter Densen, Terence S. Dermody, Robin Dewar, James H. Diaz, Carl W. Dieffenbach, Jules L. Dienstag, Yohei Doi, Raphael Dolin, J. Peter Donnelly, Michael S. Donnenberg, Gerald R. Donowitz, Philip R. Dormitzer, James M. Drake, J. Stephen Dumler, J. Stephen Dummer, Herbert L. DuPont, David T. Durack, Marlene L. Durand, Paul H. Edelstein, Michael B. Edmond, John E. Edwards, Morven S. Edwards, George M. Eliopoulos, Richard T. Ellison, Timothy P. Endy, N. Cary Engleberg, Hakan Erdem, Joel D. Ernst, Peter B. Ernst, Rick M. Fairhurst, Jessica K. Fairley, Stanley Falkow, Ann R. Falsey, Anthony S. Fauci, Thomas Fekete, Paul D. Fey, Steven M. Fine, Daniel W. Fitzgerald, Anthony R. Flores, Derek Forster, Vance G. Fowler, David O. Freedman, Arthur M. Friedlander, John N. Galgiani, John I. Gallin, Robert C. Gallo, Tejal N. Gandhi, Wendy S. Garrett, Charlotte A. Gaydos, Thomas W. Geisbert, Jeffrey A. Gelfand, Steven P. Gelone, Dale N. Gerding, Anne A. Gershon, Janet R. Gilsdorf, Ellie J.C. Goldstein, Fred M. Gordin, Paul S. Graman, M. Lindsay Grayson, Jeffrey Bruce Greene, Patricia M. Griffin, David E. Griffith, Richard L. Guerrant, H. Cem Gul, David A. Haake, David W. Haas, Charles Haines, Caroline Breese Hall, Joelle Hallak, Scott A. Halperin, Margaret R. Hammerschlag, Rashidul Haque, Jason B. Harris, Claudia Hawkins, Roderick J. Hay, Craig W. Hedberg, David K. Henderson, Donald A. Henderson, Kevin P. High, Adrian V.S. Hill, David R. Hill, Alan R. Hinman, Martin S. Hirsch, Aimee Hodowanec, Tobias M. Hohl, Steven M. Holland, Robert S. Holzman, Edward W. Hook, David C. Hooper, Thomas M. Hooton, Harold W. Horowitz, C. Robert Horsburgh, James M. Horton, Duane R. Hospenthal, Kevin Hsueh, James M. Hughes, Noreen A. Hynes, Nicole M. Iovine, Jonathan R. Iredell, Michael G. Ison, J. Michael Janda, Edward N. Janoff, Eric C. Johannsen, Angela D.M. Kashuba, Dennis L. Kasper, Donald Kaye, Keith S. Kaye, Kenneth M. Kaye, James W. Kazura, Jay S. Keystone, Rima Khabbaz, David A. Khan, Yury Khudyakov, Rose Kim, Charles H. King, Louis V. Kirchhoff, Jerome O. Klein, Michael Klompas, Bettina M. Knoll, Kirk U. Knowlton, Jane E. Koehler, Stephan A. Kohlhoff, Eija Könönen, Dimitrios P. Kontoyiannis, Igor J. Koralnik, Poonum S. Korpe, Anita A. Koshy, Joseph A. Kovacs, Phyllis Kozarsky, John Krieger, Andrew T. Kroger, Matthew J. Kuehnert, Nalin M. Kumar, Merin Elizabeth Kuruvilla, Regina C. LaRocque, James E. Leggett, Helena Legido-Quigley, Paul N. Levett, Donald P. Levine, Matthew E. Levison, Alexandra Levitt, Russell E. Lewis, W. Conrad Liles, Aldo A.M. Lima, Ajit P. Limaye, W. Ian Lipkin, Nathan Litman, Bennett Lorber, Ruth Ann Luna, Conan MacDougall, Rob Roy, Philip A. Mackowiak, Lawrence C. Madoff, Alan J. Magill, James H. Maguire, Frank Maldarelli, Lewis Markoff, Jeanne M. Marrazzo, Thomas J. Marrie, Thomas Marth, David H. Martin, Gregory J. Martin, Francisco M. Marty, Melanie Jane Maslow, Henry Masur, Alison Mawle, Kenneth H. Mayer, John T. McBride, James S. McCarthy, William M. McCormack, Catherine C. McGowan, Kenneth McIntosh, Paul S. Mead, Malgorzata Mikulska, Robert F. Miller, Samuel I. Miller, David H. Mitchell, John F. Modlin, Rajal K. Mody, Robert C. Moellering, Matthew Moffa, Susan Moir, José G. Montoya, Thomas A. Moore, Philippe Moreillon, J. Glenn Morris, Caryn Gee Morse, Robin Moseley, Robert S. Munford, Edward L. Murphy, Timothy F. Murphy, Barbara E. Murray, Clinton K. Murray, Patrick R. Murray, Daniel M. Musher, Jerod L. Nagel, Esteban C. Nannini, Anna Narezkina, Theodore E. Nash, William M. Nauseef, Jennifer L. Nayak, Marguerite A. Neill, Judith A. O'Donnell, Christopher A. Ohl, Pablo C. Okhuysen, Andrew B. Onderdonk, Steven M. Opal, Walter A. Orenstein, Douglas R. Osmon, Michael T. Osterholm, Stephen M. Ostroff, Michael N. Oxman, Slobodan Paessler, Andrea V. Page, Manjunath P. Pai, Tara N. Palmore, Raj Palraj, Peter G. Pappas, Mark S. Pasternack, Thomas F. Patterson, Deborah Pavan-Langston, David A. Pegues, Robert L. Penn, John R. Perfect, Stanley Perlman, Brett W. Petersen, Phillip K. Peterson, William A. Petri, Cathy A. Petti, Jennifer A. Philips, Julie V. Philley, Michael Phillips, Larry K. Pickering, Peter Piot, Jason M. Pogue, Aurora Pop-Vicas, Cynthia Portal-Celhay, John H. Powers, Richard N. Price, Yok-Ai Que, Justin D. Radolf, Sanjay Ram, Didier Raoult, Jonathan I. Ravdin, Stuart C. Ray, Annette C. Reboli, Marvin S. Reitz, David A. Relman, Cybèle A. Renault, Angela Restrepo, John H. Rex, Elizabeth G. Rhee, Norbert J. Roberts, José R. Romero, Alan L. Rothman, Craig R. Roy, Kathryn L. Ruoff, Mark E. Rupp, Charles E. Rupprecht, Thomas A. Russo, William A. Rutala, Edward T. Ryan, Amar Safdar, Mohammad M. Sajadi, Juan C. Salazar, Juan Carlos Sarria, Maria C. Savoia, Paul E. Sax, W. Michael Scheld, Joshua T. Schiffer, David Schlossberg, Thomas Schneider, Anne Schuchat, Jane R. Schwebke, Cynthia L. Sears, Leopoldo N. Segal, Parham Sendi, Kent A. Sepkowitz, Edward J. Septimus, Alexey Seregin, Stanford T. Shulman, George K. Siberry, Omar K. Siddiqi, Costi D. Sifri, Michael S. Simberkoff, Francesco R. Simonetti, Kamaljit Singh, Nina Singh, Upinder Singh, Scott W. Sinner, Sumathi Sivapalasingam, Leonard N. Slater, A. George Smulian, Jack D. Sobel, M. Rizwan Sohail, David E. Soper, Tania C. Sorrell, James M. Steckelberg, Allen C. Steere, Neal H. Steigbigel, James P. Steinberg, David S. Stephens, Timothy R. Sterling, David A. Stevens, Dennis L. Stevens, Jacob Strahilevitz, Charles W. Stratton, Anthony F. Suffredini, Kathryn N. Suh, Mark S. Sulkowski, Morton N. Swartz, Thomas R. Talbot, C. Sabrina Tan, Ming Tan, Chloe Lynne Thio, David L. Thomas, Lora D. Thomas, Stephen J. Thomas, Anna R. Thorner, Angela María Tobón, Edmund C. Tramont, John J. Treanor, Jason Trubiano, Athe M.N. Tsibris, Allan R. Tunkel, Ronald B. Turner, Kenneth L. Tyler, Ahmet Uluer, Diederik van de Beek, Walter J.F.M. van der Velden, Edouard G. Vannier, Trevor C. Van, James Versalovic, Claudio Viscoli, Ellen R. Wald, Matthew K. Waldor, David H. Walker, Richard J. Wallace, Edward E. Walsh, Stephen R. Walsh, Peter D. Walzer, Christine A. Wanke, Cirle A. Warren, Ronald G. Washburn, Valerie Waters, David J. Weber, Michael D. Weiden, Geoffrey A. Weinberg, Daniel J. Weisdorf, Louis M. Weiss, David F. Welch, Thomas E. Wellems, Richard P. Wenzel, Melinda Wharton, A. Clinton White, Richard J. Whitley, Walter R. Wilson, Glenn W. Wortmann, William F. Wright, Jo-Anne H. Young, Vincent B. Young, Nadezhda Yun, Werner Zimmerli, Stephen H. Zinner, and John J. Zurlo
- Published
- 2015
- Full Text
- View/download PDF
36. Kaposi's Sarcoma–Associated Herpesvirus (Human Herpesvirus 8)
- Author
-
Kenneth M. Kaye
- Subjects
business.industry ,Multicentric Castleman's disease ,medicine ,Kaposi's sarcoma-associated herpesvirus ,medicine.disease_cause ,business ,Virology ,Human herpesvirus - Published
- 2015
- Full Text
- View/download PDF
37. The Kaposi Sarcoma Herpesvirus latency-associated nuclear antigen DNA binding domain dorsal positive electrostatic patch facilitates DNA replication and episome persistence
- Author
-
Rajesh Ponnusamy, Franceline Juillard, Kenneth M. Kaye, Bruno Correia, Maria Arménia Carrondo, J. Pedro Simas, Colin E. McVey, Min Tan, Shijun Li, and Repositório da Universidade de Lisboa
- Subjects
DNA Replication ,Viral protein ,viruses ,Static Electricity ,Biology ,medicine.disease_cause ,Microbiology ,Biochemistry ,chemistry.chemical_compound ,Plasmid ,Cell Line, Tumor ,medicine ,Kaposi sarcoma-associated herpesvirus (KSHV) ,Humans ,Binding site ,Nuclear protein ,Antigens, Viral ,Molecular Biology ,Cancer ,Mutation ,Latency-associated nuclear antigen (LANA) ,Site-directed mutagenesis ,Binding Sites ,Structure-function ,Terminal Repeat Sequences ,DNA replication ,Nuclear Proteins ,virus diseases ,Herpesvirus ,Cell Biology ,DNA-binding domain ,Tumor virus ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,Virus Latency ,chemistry ,Episome ,Herpesvirus 8, Human ,DNA viruses ,DNA ,Plasmids - Abstract
© 2015 ASBMB. Currently published by Elsevier Inc; originally published by American Society for Biochemistry and Molecular Biology., Kaposi sarcoma-associated herpesvirus (KSHV) has a causative role in several human malignancies. KSHV latency-associated nuclear antigen (LANA) mediates persistence of viral episomes in latently infected cells. LANA mediates KSHV DNA replication and segregates episomes to progeny nuclei. The structure of the LANA DNA binding domain was recently solved, revealing a positive electrostatic patch opposite the DNA binding surface, which is the site of BET protein binding. Here we investigate the functional role of the positive patch in LANA-mediated episome persistence. As expected, LANA mutants with alanine or glutamate substitutions in the central, peripheral, or lateral portions of the positive patch maintained the ability to bind DNA by EMSA. However, all of the substitution mutants were deficient for LANA DNA replication and episome maintenance. Mutation of the peripheral region generated the largest deficiencies. Despite these deficiencies, all positive patch mutants concentrated to dots along mitotic chromosomes in cells containing episomes, similar to LANA. The central and peripheral mutants, but not the lateral mutants, were reduced for BET protein interaction as assessed by co-immunoprecipitation. However, defects in BET protein binding were independent of episome maintenance function. Overall, the reductions in episome maintenance closely correlated with DNA replication deficiencies, suggesting that the replication defects account for the reduced episome persistence. Therefore, the electrostatic patch exerts a key role in LANA-mediated DNA replication and episome persistence and may act through a host cell partner(s) other than a BET protein or by inducing specific structures or complexes.
- Published
- 2015
38. Epstein-Barr Virus (Infectious Mononucleosis, Epstein-Barr Virus–Associated Malignant Diseases, and Other Diseases)
- Author
-
Kenneth M. Kaye and Eric Johannsen
- Subjects
Mononucleosis ,business.industry ,medicine.disease ,medicine.disease_cause ,Hodgkin's lymphoma ,Epstein–Barr virus ,Virology ,Nasopharyngeal carcinoma ,Immunology ,Medicine ,Hemophagocytosis ,Lymphoproliferative disease ,business ,Burkitt's lymphoma - Published
- 2015
- Full Text
- View/download PDF
39. Kaposi's Sarcoma-Associated Herpesvirus Latency-Associated Nuclear Antigen Interacts with Bromodomain Protein Brd4 on Host Mitotic Chromosomes
- Author
-
William J. Harrington, Jianxin You, Mary E. Ballestas, Gerald V. Denis, Viswanathan Srinivasan, Peter M. Howley, and Kenneth M. Kaye
- Subjects
Oncogene Proteins, Fusion ,viruses ,Immunology ,Mitosis ,Cell Cycle Proteins ,HIV Infections ,Genome, Viral ,Plasma protein binding ,Biology ,medicine.disease_cause ,Microbiology ,Chromosomes ,Cell Line ,Virology ,medicine ,Humans ,Gammaherpesvirinae ,Nuclear protein ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,Transcription factor ,Binding Sites ,Genome ,Nuclear Proteins ,virus diseases ,Chromosome ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Molecular biology ,Virus-Cell Interactions ,Protein Structure, Tertiary ,Bromodomain ,Insect Science ,Herpesvirus 8, Human ,Protein Binding ,Transcription Factors - Abstract
The latency-associated nuclear antigen (LANA) of Kaposi's sarcoma-associated herpesvirus (KSHV) is required for viral episome maintenance in host cells during latent infection. Two regions of the protein have been implicated in tethering LANA/viral episomes to the host mitotic chromosomes, and LANA chromosome-binding sites are subjects of high interest. Because previous studies had identified bromodomain protein Brd4 as the mitotic chromosome anchor for the bovine papillomavirus E2 protein, which tethers the viral episomes to host mitotic chromosomes (J. You, J. L. Croyle, A. Nishimura, K. Ozato, and P. M. Howley, Cell 117: 349-360, 2004, and J. You, M. R. Schweiger, and P. M. Howley, J. Virol. 79: 14956-14961, 2005), we examined whether KSHV LANA interacts with Brd4. We found that LANA binds Brd4 in vivo and in vitro and that the binding is mediated by a direct protein-protein interaction between the ET (extraterminal) domain of Brd4 and a carboxyl-terminal region of LANA previously implicated in chromosome binding. Brd4 associates with mitotic chromosomes throughout mitosis and demonstrates a strong colocalization with LANA and the KSHV episomes on host mitotic chromosomes. Although another bromodomain protein, RING3/Brd2, binds to LANA in a similar fashion in vitro, it is largely excluded from the mitotic chromosomes in KSHV-uninfected cells and is partially recruited to the chromosomes in KSHV-infected cells. These data identify Brd4 as an interacting protein for the carboxyl terminus of LANA on mitotic chromosomes and suggest distinct functional roles for the two bromodomain proteins RING3/Brd2 and Brd4 in LANA binding. Additionally, because Brd4 has recently been shown to have a role in transcription, we examined whether Brd4 can regulate the CDK2 promoter, which can be transactivated by LANA.
- Published
- 2006
- Full Text
- View/download PDF
40. Kaposi’s Sarcoma-Associated Herpesvirus LANA Hitches a Ride on the Chromosome
- Author
-
Kenneth M. Kaye, Karolin Luger, Jayanth V. Chodaparambil, Brenna Kelley-Clarke, and Andrew J. Barbera
- Subjects
Models, Molecular ,Protein Conformation ,viruses ,Crystallography, X-Ray ,medicine.disease_cause ,Chromosomes ,Histones ,medicine ,Nucleosome ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,Ku Autoantigen ,Sarcoma, Kaposi ,Molecular Biology ,Mitosis ,biology ,DNA replication ,Nuclear Proteins ,virus diseases ,Chromosome ,Antigens, Nuclear ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Virology ,Nucleosomes ,Chromatin ,DNA-Binding Proteins ,Histone ,DNA, Viral ,Herpesvirus 8, Human ,biology.protein ,Primary effusion lymphoma ,Poly(ADP-ribose) Polymerases ,Dimerization ,Developmental Biology - Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) latently infects tumor cells and has an etiologic role in Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. Survival in rapidly dividing cells depends on a carefully orchestrated chain of events. The viral genome, or episome, must replicate in concert with cellular genetic material, and then efficiently segregate to progeny nuclei. KSHV achieves this through its latency associated nuclear antigen (LANA), which simultaneously binds to viral DNA and mitotic chromosomes to efficiently partition episomes. LANA's N-terminal region has been shown to be essential for efficient KSHV DNA replication and tethering to mitotic chromosomes. The precise mechanism by which LANA attaches to host chromosomes has been an area of active investigation. We recently reported that this association is mediated by the chromatin components histones H2A and H2B. Binding between LANA and these proteins was demonstrated in vivo and in vitro, and use of an H2A-H2B depleted system demonstrated their central role in LANA's chromosome binding. Further, we provided a structural description of the interaction of LANA's N-terminal chromosome association region with the nucleosome using x-ray crystallography. Our data offer further insight into the mechanism of KSHV latency, and also reveal a new concept for a role of the nucleosome as a docking site for other proteins.
- Published
- 2006
- Full Text
- View/download PDF
41. KSHV LANA1 binds DNA as an oligomer and residues N-terminal to the oligomerization domain are essential for DNA binding, replication, and episome persistence
- Author
-
Kenneth M. Kaye, Andrew J. Barbera, Mary E. Ballestas, Brenna Kelley-Clarke, and Takashi Komatsu
- Subjects
DNA Replication ,HMG-box ,DNA polymerase ,DNA polymerase II ,Virus Replication ,03 medical and health sciences ,Virology ,Animals ,Replication protein A ,Antigens, Viral ,030304 developmental biology ,0303 health sciences ,DNA clamp ,biology ,030302 biochemistry & molecular biology ,DNA replication ,Terminal Repeat Sequences ,Nuclear Proteins ,KSHV LANA1 ,Molecular biology ,DNA binding site ,DNA, Viral ,Herpesvirus 8, Human ,biology.protein ,Episome persistence ,Binding domain ,Plasmids - Abstract
Latency-associated nuclear antigen 1 (LANA1) binds to Kaposi's sarcoma-associated herpesvirus (KSHV) terminal repeat (TR) DNA to mediate episome replication and persistence. LANA1 concentrates at sites of TR DNA along mitotic chromosomes, consistent with tethering KSHV DNA to chromosomes for efficient segregation of episomes to progeny nuclei. We now investigate LANA1 C-terminus self-association and DNA binding. The TR DNA binding domain was localized to LANA1 residues 996–1139. Scanning deletions within this region ablated both LANA1 oligomerization and DNA binding, consistent with a requirement for oligomerization to bind DNA. Furthermore, LANA1 bound TR DNA as an oligomer. Deletion of amino acids 1007–1021, N-terminal to the LANA1 oligomerization domain, ablated DNA binding, DNA replication, and episome persistence, implicating these residues in contacting DNA. Indeed, LANA1 residues 1007–1021 correspond to EBNA1 residues that contact the cognate sequence. Like EBNA1, the LANA1 DNA-binding domain has oligomerization activity and critical residues essential for recognizing DNA.
- Published
- 2004
- Full Text
- View/download PDF
42. The Kaposi's Sarcoma-Associated Herpesvirus Latency-Associated Nuclear Antigen 1 N Terminus Is Essential for Chromosome Association, DNA Replication, and Episome Persistence
- Author
-
Mary E. Ballestas, Andrew J. Barbera, and Kenneth M. Kaye
- Subjects
DNA Replication ,Immunology ,Biology ,medicine.disease_cause ,Microbiology ,Structure-Activity Relationship ,chemistry.chemical_compound ,Plasmid ,Control of chromosome duplication ,Cell Line, Tumor ,Virology ,medicine ,Chromosomes, Human ,Humans ,Gammaherpesvirinae ,Kaposi's sarcoma-associated herpesvirus ,Antigens, Viral ,Mutation ,Terminal Repeat Sequences ,DNA replication ,Nuclear Proteins ,Chromosome ,biology.organism_classification ,Molecular biology ,Virus-Cell Interactions ,chemistry ,Insect Science ,Herpesvirus 8, Human ,DNA ,Plasmids - Abstract
To persist in latently infected, proliferating cells, Kaposi's sarcoma-associated herpesvirus (KSHV) episomes must replicate and efficiently segregate to progeny nuclei. Episome persistence in uninfected cells requires latency-associated nuclear antigen 1 (LANA1) in trans and cis -acting KSHV terminal repeat (TR) DNA. The LANA1 C terminus binds TR DNA, and LANA1 mediates TR-associated DNA replication in transient assays. LANA1 also concentrates at sites of KSHV TR DNA episomes along mitotic chromosomes, consistent with a tethering role to efficiently segregate episomes to progeny nuclei. LANA1 amino acids 5 to 22 constitute a chromosome association region (Piolot et al., J. Virol. 75: 3948-3959, 2001). We now investigate LANA1 residues 5 to 22 with scanning alanine substitutions. Mutations targeting LANA1 5 GMR 7 , 8 LRS 10 , and 11 GRS 13 eliminated chromosome association, DNA replication, and episome persistence. LANA1 mutated at 14 TG 15 retained the ability to associate with chromosomes but was partially deficient in DNA replication and episome persistence. These results provide genetic support for a key role of the LANA1 N terminus in chromosome association, LANA1-mediated DNA replication, and episome persistence.
- Published
- 2004
- Full Text
- View/download PDF
43. The KSHV latency-associated nuclear antigen a multifunctional protein
- Author
-
Kenneth M Kaye
- Published
- 2002
- Full Text
- View/download PDF
44. Abstract 5110: A novel nucleoside analog therapeutically active against plasma cell malignancies and other ADK-expressing cancers including colon and pancreatic adenocarcinomas
- Author
-
Shizuko Sei, Robert H. Shoemaker, Jonathan Reichel, Utthara Nayar, Jouliana Sadek, Jennifer Totonchy, Kenneth M. Kaye, Olivier Elemento, David Warren, and Ethel Cesarman
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Cancer ,Purine analogue ,Plasma cell ,medicine.disease ,ADK ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine ,Adenocarcinoma ,Primary effusion lymphoma ,business ,Nucleoside ,Multiple myeloma - Abstract
A number of nucleoside analogues are used successfully for the treatment of several cancers, and in particular leukemias and lymphomas, but these have distinct efficacies for different tumor types, and many malignancies do not respond to currently available nucleoside analogues or other forms of chemotherapy. A high throughput screen conducted in our lab to search for inhibitors of primary effusion lymphoma (PEL) identified the nucleoside analog 6-ethylthioinosine (6-ETI) as a potent and selective inhibitor of PEL, a largely incurable malignancy of B cell origin with plasmacytic differentiation. 6-ETI induced necrosis and ATP-depletion accompanied by S-phase arrest, DNA damage and inhibition of DNA synthesis. To understand 6-ETI mechanism of selectivity, RNA-seq analysis of in vitro generated drug-resistant PEL clones revealed inactivating mutations and loss of expression of adenosine kinase (ADK) as the mechanism of resistance. In vitro assays showed that 6-ETI is a pro-drug that gets phosphorylated and activated by adenosine kinase (ADK) into its active form. We found high ADK expression in PEL cell lines and primary specimens of PEL, multiple myeloma (MM) and plasmablastic lymphoma (PBL) patient samples. 6-ETI was effective at killing multiple myeloma cell lines, primary MM specimens, and had a remarkable anti-tumor response in a disseminated multiple myeloma and PEL xenograft mouse models. Thus, ADK expression can serve as a predictive biomarker to help identify patients that are most likely to respond to 6-ETI treatment. To further assess the spectrum of activity and sensitivity of 6-ETI, we examined ADK expression in other cancer subtypes and found that colorectal and pancreatic adenocarcinomas also overexpress ADK and are highly sensitive to killing by 6-ETI at the low nanomolar concentration. We also found high ADK expression in primary colon and pancreatic adenocarcinoma patient specimens. We compared 6-ETI to other FDA-approved purine analogs and failed to find other compounds with similar potency or selectivity profile. Herein, we report the identification of a novel purine analog, 6-ethylthioinosine, as an effective therapeutic with exquisite sensitivity to plasma cell malignancies and other ADK-expressing cancers. We have successfully used RNASeq-based “resistome” analysis to identify its mechanism of specificity and discovered a new biomarker that can potentially impact patient care and the treatment of some of the most aggressive tumors. Citation Format: Jouliana Sadek, Utthara Nayar, Jonathan Reichel, Jennifer Totonchy, Shizuko Sei, Robert Shoemaker, David Warren, Olivier Elemento, Kenneth Kaye, Ethel Cesarman. A novel nucleoside analog therapeutically active against plasma cell malignancies and other ADK-expressing cancers including colon and pancreatic adenocarcinomas [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5110. doi:10.1158/1538-7445.AM2017-5110
- Published
- 2017
- Full Text
- View/download PDF
45. Correction for Sun et al., Kaposi’s sarcoma-associated herpesvirus LANA recruits the DNA polymerase clamp loader to mediate efficient replication and virus persistence
- Author
-
Toshiki Tsurimoto, She Chen, Qiming Sun, Franceline Juillard, Shijun Li, Erika De León Vázquez, Kenneth M. Kaye, and Lin Li
- Subjects
Multidisciplinary ,Clamp ,biology ,DNA polymerase ,Replication (statistics) ,medicine ,biology.protein ,Kaposi's sarcoma-associated herpesvirus ,Viral persistence ,medicine.disease_cause ,Virology ,Corrections - Published
- 2014
46. Kaposi's sarcoma-associated herpesvirus LANA recruits the DNA polymerase clamp loader to mediate efficient replication and virus persistence
- Author
-
Kenneth M. Kaye, Shijun Li, She Chen, Toshiki Tsurimoto, Franceline Juillard, Lin Li, Erika De León Vázquez, and Qiming Sun
- Subjects
DNA Replication ,viruses ,Eukaryotic DNA replication ,DNA-Directed DNA Polymerase ,Virus Replication ,DNA polymerase delta ,DNA replication factor CDT1 ,Replication factor C ,Control of chromosome duplication ,Cell Line, Tumor ,Proliferating Cell Nuclear Antigen ,Humans ,Replication Protein C ,Antigens, Viral ,Sarcoma, Kaposi ,Multidisciplinary ,biology ,DNA replication ,virus diseases ,Nuclear Proteins ,biochemical phenomena, metabolism, and nutrition ,Biological Sciences ,Virology ,Proliferating cell nuclear antigen ,Virus Latency ,Gene Knockdown Techniques ,DNA, Viral ,Herpesvirus 8, Human ,Host-Pathogen Interactions ,biology.protein ,Origin recognition complex - Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) latently infects tumor cells and persists as a multiple-copy, extrachromosomal, circular episome. To persist, the viral genome must replicate with each cell cycle. The KSHV latency-associated nuclear antigen (LANA) mediates viral DNA replication and persistence, but little is known regarding the underlying mechanisms. We find that LANA recruits replication factor C (RFC), the DNA polymerase clamp [proliferating cell nuclear antigen (PCNA)] loader, to drive DNA replication efficiently. Mutated LANA lacking RFC interaction was deficient for LANA-mediated DNA replication and episome persistence. RFC depletion had a negative impact on LANA's ability to replicate and maintain viral DNA in cells containing artificial KSHV episomes or in infected cells, leading to loss of virus. LANA substantially increased PCNA loading onto DNA in vitro and recruited RFC and PCNA to KSHV DNA in cells. These findings suggest that PCNA loading is a rate-limiting step in DNA replication that is incompatible with viral survival. LANA enhancement of PCNA loading permits efficient virus replication and persistence, revealing a previously unidentified mechanism for KSHV latency.
- Published
- 2014
47. Kaposi's Sarcoma-Associated Herpesvirus Latency-Associated Nuclear Antigen 1 Mediates Episome Persistence through cis -Acting Terminal Repeat (TR) Sequence and Specifically Binds TR DNA
- Author
-
Mary E. Ballestas and Kenneth M. Kaye
- Subjects
DNA Replication ,viruses ,Molecular Sequence Data ,Immunology ,Biology ,medicine.disease_cause ,Microbiology ,chemistry.chemical_compound ,Plasmid ,Virology ,Extrachromosomal DNA ,medicine ,Kaposi's sarcoma-associated herpesvirus ,Nuclear protein ,Antigens, Viral ,Mitosis ,Metaphase ,Base Sequence ,Terminal Repeat Sequences ,DNA replication ,Nuclear Proteins ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Precipitin Tests ,Molecular biology ,Virus-Cell Interactions ,chemistry ,Insect Science ,DNA, Viral ,Herpesvirus 8, Human ,DNA ,Plasmids - Abstract
Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) (also known as human herpesvirus 8) latently infects KS tumors, primary effusion lymphomas (PELs), and PEL cell lines. In latently infected cells, KSHV DNA is maintained as circularized, extrachromosomal episomes. To persist in proliferating cells, KSHV episomes must replicate and efficiently segregate to progeny nuclei. In uninfected B-lymphoblastoid cells, KSHV latency-associated nuclear antigen (LANA1) is necessary and sufficient for persistence of artificial episomes containing specific KSHV DNA. In previous work, the cis -acting sequence required for episome persistence contained KSHV terminal-repeat (TR) DNA and unique KSHV sequence. We now show that cis -acting KSHV TR DNA is necessary and sufficient for LANA1-mediated episome persistence. Furthermore, LANA1 binds TR DNA in mobility shift assays and a 20-nucleotide LANA1 binding sequence has been identified. Since LANA1 colocalizes with KSHV episomes along metaphase chromosomes, these results are consistent with a model in which LANA1 may bridge TR DNA to chromosomes during mitosis to efficiently segregate KSHV episomes to progeny nuclei.
- Published
- 2001
- Full Text
- View/download PDF
48. ONCE-DAILY DOSING OF AMINOGLYCOSIDE ANTIBIOTICS
- Author
-
David N. Fisman and Kenneth M. Kaye
- Subjects
Adult ,Microbiology (medical) ,medicine.drug_class ,Antibiotics ,Pharmacology ,Drug Administration Schedule ,Microbiology ,medicine ,Tobramycin ,Humans ,Dosing ,Child ,business.industry ,Aminoglycoside ,Infant ,Bacterial Infections ,Anti-Bacterial Agents ,Aminoglycosides ,Treatment Outcome ,Infectious Diseases ,Amikacin ,Streptomycin ,Gentamicin ,Netilmicin ,business ,medicine.drug - Abstract
The aminoglycosides are a class of bactericidal antibiotics characterized by the presence of a six-carbon aminocyclitol ring covalently bonded to multiple amino sugar groups. 24 Aminoglycosides commonly used for the treatment of serious bacterial infections in the United States include gentamicin, tobramycin, amikacin, netilmicin, and streptomycin. 18 These drugs act in part by impairing bacterial protein synthesis through irreversible binding to the 30S subunit of the bacterial ribosome. 24 Since the introduction of streptomycin in the 1940s, aminoglycosides have proved extremely useful in the treatment of infections caused by gram-negative bacilli, including Pseudomonas aeruginosa, and infections caused by staphylococci, mycobacteria, and several other pathogens. 18 The Food and Drug Administration (FDA) approved dosing regimens for aminoglycoside antibiotics require multiple daily doses in individuals with normal renal function. Improvements in the understanding of the pharmacodynamics of aminoglycoside efficacy and mechanisms of toxicity, however, have prompted the evaluation of once-daily dosing regimens in clinical studies. In the pages that follow, we review the rationale behind once-daily dosing of aminoglycoside therapy as well as clinical data on the efficacy and toxicity of once-daily dosing of aminoglycosides. We also review practical aspects of dosing and monitoring once-daily aminoglycoside therapy and issues complicating the use of these regimens in special populations (including children, adults with an altered volume of distribution for aminoglycosides, and individuals with renal dysfunction) and in certain illnesses (including bacterial endocarditis, neutropenia and fever, and cystic fibrosis).
- Published
- 2000
- Full Text
- View/download PDF
49. Epstein – Barr virus-encoded LMP1 and CD40 mediate IL-6 production in epithelial cells via an NF-κB pathway involving TNF receptor-associated factors
- Author
-
Liz Hodgkin, Maria Stack, Christopher W. Dawson, Aristides G. Eliopoulos, Lawrence S. Young, Kenneth M. Kaye, Sim K. Sihota, and Martin Rowe
- Subjects
TRAF3 ,Herpesvirus 4, Human ,Cancer Research ,TRAF2 ,medicine.medical_treatment ,Molecular Sequence Data ,Transfection ,Epithelium ,Receptors, Tumor Necrosis Factor ,Viral Matrix Proteins ,Tumor Cells, Cultured ,otorhinolaryngologic diseases ,Genetics ,medicine ,Humans ,Amino Acid Sequence ,CD40 Antigens ,Molecular Biology ,Transcription factor ,Ovarian Neoplasms ,Binding Sites ,CD40 ,TNF Receptor-Associated Factor 3 ,biology ,Interleukin-6 ,NF-kappa B ,Proteins ,Cell Transformation, Viral ,TNF Receptor-Associated Factor 2 ,TNF Receptor-Associated Factor 1 ,Protein Structure, Tertiary ,Cell biology ,stomatognathic diseases ,Cytokine ,Urinary Bladder Neoplasms ,biology.protein ,Cancer research ,Female ,Signal transduction ,Cell Division ,HeLa Cells - Abstract
Expression of the Epstein-Barr virus (EBV) transforming LMP1 in B cells activates the transcription factor NF-kappaB and induces phenotypic changes through two distinct domains in the cytoplasmic C-terminus of the protein. The aa 187-231 domain of LMP1, which is important for growth transformation, binds tumour necrosis factor (TNF) receptor associated factor (TRAF) 1 and TRAF3 and this interaction mediates subsequent signalling events. The TRAFs also associate with CD40, a member of the TNFR family, which upon ligation activates NF-kappaB and induces phenotypic changes similar to those mediated by LMP1. This study demonstrates that LMP1 expression in carcinoma cell lines and SV40-transformed keratinocytes results in induction of the pleiotropic cytokine interleukin 6 (IL6), an effect which is also observed upon CD40 ligation. The mechanism by which either LMP1 expression or CD40 ligation induces IL6 production was found to be NF-kappaB-dependent. Mutational analysis identified domains in the C-terminus of LMP1 which are important for NF-kappaB activation and IL6 secretion. LMP1 and CD40 share a common PxQxT core TRAF binding motif and mutations in or adjacent to this sequence impaired the ability of LMP1 or CD40 to induce NF-kappaB activation and IL6 secretion. The importance of TRAF interactions in mediating these effects was confirmed using dominant negative TRAF2 and TRAF3 mutants which also identified differences in the signalling events mediated by the two NF-kappaB activating domains of LMP1. A20, an anti-apoptotic protein which interacts with TRAF2 and blocks CD40-mediated NF-kappaB activity, also blocked NF-kappaB and IL6 secretion in LMP1-transfected epithelial cells. These results suggest that LMP1 regulates IL6 production in epithelial cells in a manner similar to CD40 ligation and implicate TRAFs as common mediators in the transduction of signals generated via the CD40 and LMP1 pathways. As a role for IL6 in regulating epithelial cell growth has previously been suggested, the control of IL6 secretion via the CD40 and LMP1 pathways may have implications for the growth of both normal and transformed epithelial cells.
- Published
- 1997
- Full Text
- View/download PDF
50. The Epstein–Barr virus LMP1 amino acid sequence that engages tumor necrosis factor receptor associated factors is critical for primary B lymphocyte growth transformation
- Author
-
Kenneth M. Izumi, Elliott Kieff, and Kenneth M. Kaye
- Subjects
Herpesvirus 4, Human ,Lymphoma ,Biology ,medicine.disease_cause ,Virus ,Viral Matrix Proteins ,Epitopes ,hemic and lymphatic diseases ,otorhinolaryngologic diseases ,medicine ,Gene ,Peptide sequence ,Sequence Deletion ,B-Lymphocytes ,Mutation ,Multidisciplinary ,TNF Receptor-Associated Factor 3 ,Proteins ,Epstein–Barr virus latent membrane protein 1 ,Biological Sciences ,Cell Transformation, Viral ,Hematopoietic Stem Cells ,TNF Receptor-Associated Factor 2 ,TNF Receptor-Associated Factor 1 ,Epstein–Barr virus ,Virology ,stomatognathic diseases ,Cell Transformation, Neoplastic ,Cell culture ,Protein Binding ,Binding domain - Abstract
Epstein–Barr virus (EBV) latent membrane protein 1 (LMP1) is essential for transforming primary B lymphocytes into lymphoblastoid cell lines. EBV recombinants with LMP1 genes truncated after the proximal 45 codons of the LMP1 carboxyl terminus are adequate for transformation. The proximal 45 residues include a domain that engages the tumor necrosis factor receptor associated factors (TRAFs). We investigated the importance of the TRAF binding domain by assaying the transforming ability of recombinant EBV genomes with a deletion of LMP1 codons 185–211. This mutation eliminates TRAF association in yeast and in lymphoblasts but does not affect LMP1 stability or localization. Specifically mutated recombinant EBV genomes were generated by transfecting P3HR-1 cells with overlapping EBV cosmids. Infection of primary B lymphocytes resulted in cell lines that were coinfected with an LMP1Δ185–211 EBV recombinant and P3HR-1 EBV, which has a wild-type LMP1 gene but is transformation defective due to another deletion. Despite the equimolar mixture of wild-type and mutated LMP1 genes in virus preparations from five coinfected cell lines, only the wild-type LMP1 gene was found in 412 cell lines obtained after transformation of primary B lymphocytes. No transformed cell line had only the LMP1Δ185–211 gene. An EBV recombinant with a Flag-tagged LMP1 gene passaged in parallel segregated from the coinfecting P3HR-1. These data indicate that the LMP1 TRAF binding domain is critical for primary B lymphocyte growth transformation.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.