1
|
Ameya G, Birri DJ. The molecular mechanisms of virus-induced human cancers. Microb Pathog 2023; 183:106292. [PMID: 37557930 DOI: 10.1016/j.micpath.2023.106292] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 08/11/2023]
Abstract
Cancer is a serious public health problem globally. Many human cancers are induced by viruses. Understanding of the mechanisms by which oncogenic (tumorigenic) viruses induce cancer is essential in the prevention and control of cancer. This review covers comprehensive characteristics and molecular mechanisms of the main virus-attributed cancers caused by human papillomavirus, hepatitis B virus, hepatitis C virus, Epstein-Barr virus, human herpesvirus type 8, human T-cell lymphotropic virus, human polyomaviruses, Merkel cell polyomavirus, and HIV. Oncogenic viruses employ biological processes to replicate and avoid detection by host cell immune systems. Tumorigenic infectious agents activate oncogenes in a variety of ways, allowing the pathogen to block host tumour suppressor proteins, inhibit apoptosis, enhance cell proliferation, and promote invasion of host cells. Furthermore, this review assesses many pathways of viruses linked to cancer, including host cellular communication perturbation, DNA damage mechanisms, immunity, and microRNA targets that promote the beginning and progression of cancer. The current cancer prevention is primarily focused on non-communicable diseases, but infection-attributable cancer also needs attention to significantly reduce the rising cancer burden and related deaths.
Collapse
Affiliation(s)
- Gemechu Ameya
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Kotebe Metropolitan University, Addis Ababa, Ethiopia; Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Dagim Jirata Birri
- Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia.
| |
Collapse
|
2
|
Olson AT, Kang Y, Ladha AM, Zhu S, Lim CB, Nabet B, Lagunoff M, Gujral TS, Geballe AP. Polypharmacology-based kinome screen identifies new regulators of KSHV reactivation. PLoS Pathog 2023; 19:e1011169. [PMID: 37669313 PMCID: PMC10503724 DOI: 10.1371/journal.ppat.1011169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/15/2023] [Accepted: 08/16/2023] [Indexed: 09/07/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) causes several human diseases including Kaposi's sarcoma (KS), a leading cause of cancer in Africa and in patients with AIDS. KS tumor cells harbor KSHV predominantly in a latent form, while typically <5% contain lytic replicating virus. Because both latent and lytic stages likely contribute to cancer initiation and progression, continued dissection of host regulators of this biological switch will provide insights into fundamental pathways controlling the KSHV life cycle and related disease pathogenesis. Several cellular protein kinases have been reported to promote or restrict KSHV reactivation, but our knowledge of these signaling mediators and pathways is incomplete. We employed a polypharmacology-based kinome screen to identify specific kinases that regulate KSHV reactivation. Those identified by the screen and validated by knockdown experiments included several kinases that enhance lytic reactivation: ERBB2 (HER2 or neu), ERBB3 (HER3), ERBB4 (HER4), MKNK2 (MNK2), ITK, TEC, and DSTYK (RIPK5). Conversely, ERBB1 (EGFR1 or HER1), MKNK1 (MNK1) and FRK (PTK5) were found to promote the maintenance of latency. Mechanistic characterization of ERBB2 pro-lytic functions revealed a signaling connection between ERBB2 and the activation of CREB1, a transcription factor that drives KSHV lytic gene expression. These studies provided a proof-of-principle application of a polypharmacology-based kinome screen for the study of KSHV reactivation and enabled the discovery of both kinase inhibitors and specific kinases that regulate the KSHV latent-to-lytic replication switch.
Collapse
Affiliation(s)
- Annabel T. Olson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Yuqi Kang
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Anushka M. Ladha
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Songli Zhu
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Chuan Bian Lim
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Behnam Nabet
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Michael Lagunoff
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Adam P. Geballe
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
3
|
Li Y, Patterson MR, Morgan EL, Wasson CW, Ryder EL, Barba‐Moreno D, Scarth JA, Wang M, Macdonald A. CREB1 activation promotes human papillomavirus oncogene expression and cervical cancer cell transformation. J Med Virol 2023; 95:e29025. [PMID: 37565725 PMCID: PMC10952218 DOI: 10.1002/jmv.29025] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/03/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023]
Abstract
Human papillomaviruses (HPVs) infect the oral and anogenital mucosa and can cause cancer. The high-risk (HR)-HPV oncoproteins, E6 and E7, hijack cellular factors to promote cell proliferation, delay differentiation and induce genomic instability, thus predisposing infected cells to malignant transformation. cAMP response element (CRE)-binding protein 1 (CREB1) is a master transcription factor that can function as a proto-oncogene, the abnormal activity of which is associated with multiple cancers. However, little is known about the interplay between HPV and CREB1 activity in cervical cancer or the productive HPV lifecycle. We show that CREB is activated in productively infected primary keratinocytes and that CREB1 expression and phosphorylation is associated with the progression of HPV+ cervical disease. The depletion of CREB1 or inhibition of CREB1 activity results in decreased cell proliferation and reduced expression of markers of epithelial to mesenchymal transition, coupled with reduced migration in HPV+ cervical cancer cell lines. CREB1 expression is negatively regulated by the tumor suppressor microRNA, miR-203a, and CREB1 phosphorylation is controlled through the MAPK/MSK pathway. Crucially, CREB1 directly binds the viral promoter to upregulate transcription of the E6/E7 oncogenes, establishing a positive feedback loop between the HPV oncoproteins and CREB1. Our findings demonstrate the oncogenic function of CREB1 in HPV+ cervical cancer and its relationship with the HPV oncogenes.
Collapse
Affiliation(s)
- Yigen Li
- School of Molecular and Cellular Biology, Faculty of Biological SciencesUniversity of LeedsLeedsWest YorkshireUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsWest YorkshireUK
| | - Molly R. Patterson
- School of Molecular and Cellular Biology, Faculty of Biological SciencesUniversity of LeedsLeedsWest YorkshireUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsWest YorkshireUK
| | | | - Christopher W. Wasson
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and HealthUniversity of LeedsLeedsWest YorkshireUK
| | - Emma L. Ryder
- School of Molecular and Cellular Biology, Faculty of Biological SciencesUniversity of LeedsLeedsWest YorkshireUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsWest YorkshireUK
| | - Diego Barba‐Moreno
- School of Molecular and Cellular Biology, Faculty of Biological SciencesUniversity of LeedsLeedsWest YorkshireUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsWest YorkshireUK
| | - James A. Scarth
- School of Molecular and Cellular Biology, Faculty of Biological SciencesUniversity of LeedsLeedsWest YorkshireUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsWest YorkshireUK
| | - Miao Wang
- School of Molecular and Cellular Biology, Faculty of Biological SciencesUniversity of LeedsLeedsWest YorkshireUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsWest YorkshireUK
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Faculty of Biological SciencesUniversity of LeedsLeedsWest YorkshireUK
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsWest YorkshireUK
| |
Collapse
|
4
|
Olson AT, Kang Y, Ladha AM, Lim CB, Lagunoff M, Gujral TS, Geballe AP. Polypharmacology-based kinome screen identifies new regulators of KSHV reactivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526589. [PMID: 36778430 PMCID: PMC9915688 DOI: 10.1101/2023.02.01.526589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) causes several human diseases including Kaposi's sarcoma (KS), a leading cause of cancer in Africa and in patients with AIDS. KS tumor cells harbor KSHV predominantly in a latent form, while typically <5% contain lytic replicating virus. Because both latent and lytic stages likely contribute to cancer initiation and progression, continued dissection of host regulators of this biological switch will provide insights into fundamental pathways controlling the KSHV life cycle and related disease pathogenesis. Several cellular protein kinases have been reported to promote or restrict KSHV reactivation, but our knowledge of these signaling mediators and pathways is incomplete. We employed a polypharmacology-based kinome screen to identifiy specific kinases that regulate KSHV reactivation. Those identified by the screen and validated by knockdown experiments included several kinases that enhance lytic reactivation: ERBB2 (HER2 or neu ), ERBB3 (HER3), ERBB4 (HER4), MKNK2 (MNK2), ITK, TEC, and DSTYK (RIPK5). Conversely, ERBB1 (EGFR1 or HER1), MKNK1 (MNK1) and FRK (PTK5) were found to promote the maintenance of latency. Mechanistic characterization of ERBB2 pro-lytic functions revealed a signaling connection between ERBB2 and the activation of CREB1, a transcription factor that drives KSHV lytic gene expression. These studies provided a proof-of-principle application of a polypharmacology-based kinome screen for the study of KSHV reactivation and enabled the discovery of both kinase inhibitors and specific kinases that regulate the KSHV latent-to-lytic replication switch. Author Summary Kaposi's sarcoma-associated herpesvirus (KSHV) causes Kaposi's sarcoma, a cancer particularly prevalent in Africa. In cancer cells, the virus persists in a quiescent form called latency, in which only a few viral genes are made. Periodically, the virus switches into an active replicative cycle in which most of the viral genes are made and new virus is produced. What controls the switch from latency to active replication is not well understood, but cellular kinases, enzymes that control many cellular processes, have been implicated. Using a cell culture model of KSHV reactivation along with an innovative screening method that probes the effects of many cellular kinases simultaneously, we identified drugs that significantly limit KSHV reactivation, as well as specific kinases that either enhance or restrict KSHV replicative cycle. Among these were the ERBB kinases which are known to regulate growth of cancer cells. Understanding how these and other kinases contribute to the switch leading to production of more infectious virus helps us understand the mediators and mechanisms of KSHV diseases. Additionally, because kinase inhibitors are proving to be effective for treating other diseases including some cancers, identifying ones that restrict KSHV replicative cycle may lead to new approaches to treating KSHV-related diseases.
Collapse
Affiliation(s)
- Annabel T. Olson
- Division of Human Biology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Yuqi Kang
- Division of Human Biology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Anushka M. Ladha
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Chuan Bian Lim
- Division of Human Biology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Michael Lagunoff
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Taran S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Adam P. Geballe
- Division of Human Biology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Herrera-Ortíz A, Meng W, Gao SJ. Nitric oxide is induced and required for efficient Kaposi's sarcoma-associated herpesvirus lytic replication. J Med Virol 2021; 93:6323-6332. [PMID: 34297418 DOI: 10.1002/jmv.27228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/08/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus associated with several human malignancies. KSHV lytic replication promotes the spread of infection and progression of KSHV-associated malignancies; however, the mechanism regulating KSHV lytic replication remains unclear. In this study, we investigated the role of nitric oxide (NO) in KSHV lytic replication. In the TREx BCBL1-RTA KSHV lytic replication cell system, induction of KSHV lytic replication increased intracellular and extracellular NO. Chemical inhibition of NO production resulted in a lower level of KSHV lytic replication as shown by a reduced level of infectious virions, and decreased levels of viral lytic transcripts and proteins. In a second KSHV lytic replication system of iSLK-RGB-BAC16 cells, we confirmed that KSHV lytic replication increased NO production. Chemical inhibition of NO production resulted in reduced numbers of cells expressing enhanced green fluorescent protein and blue fluorescent protein, two reporters that closely track the expression of KSHV early and late genes, respectively. Consistent with these results, inhibition of NO production resulted in reduced levels of infectious virions, and viral lytic transcripts and proteins. Importantly, exogenous addition of a NO donor was sufficient to enhance the full KSHV lytic replication program. These results demonstrate that NO is required for efficient KSHV lytic replication, and NO plays a crucial role in the KSHV life cycle and KSHV-induced malignancies.
Collapse
Affiliation(s)
- Antonia Herrera-Ortíz
- Department of Immunology and Molecular Microbiology, University of Southern California Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| | - Wen Meng
- Department of Microbiology and Molecular Genetics, Cancer Virology Program, UPMC Hillman Caner Center, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shou-Jiang Gao
- Department of Immunology and Molecular Microbiology, University of Southern California Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Microbiology and Molecular Genetics, Cancer Virology Program, UPMC Hillman Caner Center, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Sevim H, Çelik H, Düşünceli L, Ceyhan CS, Molotkova A, Nakazawa K, Graham GT, Petro JR, Toretsky JA, Üren A. Clofarabine induces ERK/MSK/CREB activation through inhibiting CD99 on Ewing sarcoma cells. PLoS One 2021; 16:e0253170. [PMID: 34133426 PMCID: PMC8208565 DOI: 10.1371/journal.pone.0253170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/29/2021] [Indexed: 01/17/2023] Open
Abstract
Clofarabine, an FDA approved purine analog, is used in the treatment of relapsed or refractory acute lymphoblastic leukemia. Clofarabine acts by inhibiting DNA synthesis. We demonstrated that clofarabine may have a novel function though inhibiting CD99, a transmembrane protein highly expressed on Ewing Sarcoma (ES) cells. CD99 is a validated target in ES whose inhibition may lead to a high therapeutic index for patients. Here we present additional data to support the hypothesis that clofarabine acts on CD99 and regulates key signaling pathways in ES. Cellular thermal shift assay indicated a direct interaction between clofarabine and CD99 in ES cell lysates. Clofarabine induced ES cell death does not require clofarabine's conversion to its active form by deoxycytidine kinase. A phosphokinase array screen with clofarabine and a CD99 blocking antibody identified alterations in signaling pathways. CD99 inhibition with clofarabine in ES cells caused rapid and sustained phosphorylation of ERK, MSK, and CREB. However, activation of this pathway did not correlate with clofarabine induced ES cell death. In summary, we demonstrated that clofarabine may activate ERK, MSK, and CREB phosphorylation through CD99 within minutes, however this paradoxical activation and subsequent ES cell death requires additional investigation.
Collapse
Affiliation(s)
- Handan Sevim
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Haydar Çelik
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Levent Düşünceli
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Ceyda S. Ceyhan
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Anna Molotkova
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Kay Nakazawa
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Garrett T. Graham
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Jeffrey R. Petro
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Jeffrey A. Toretsky
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| | - Aykut Üren
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
7
|
Chander Y, Kumar R, Khandelwal N, Singh N, Shringi BN, Barua S, Kumar N. Role of p38 mitogen-activated protein kinase signalling in virus replication and potential for developing broad spectrum antiviral drugs. Rev Med Virol 2021; 31:1-16. [PMID: 33450133 DOI: 10.1002/rmv.2217] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) play a key role in complex cellular processes such as proliferation, development, differentiation, transformation and apoptosis. Mammals express at least four distinctly regulated groups of MAPKs which include extracellular signal-related kinases (ERK)-1/2, p38 proteins, Jun amino-terminal kinases (JNK1/2/3) and ERK5. p38 MAPK is activated by a wide range of cellular stresses and modulates activity of several downstream kinases and transcription factors which are involved in regulating cytoskeleton remodeling, cell cycle modulation, inflammation, antiviral response and apoptosis. In viral infections, activation of cell signalling pathways is part of the cellular defense mechanism with the basic aim of inducing an antiviral state. However, viruses can exploit enhanced cell signalling activities to support various stages of their replication cycles. Kinase activity can be inhibited by small molecule chemical inhibitors, so one strategy to develop antiviral drugs is to target these cellular signalling pathways. In this review, we provide an overview on the current understanding of various cellular and viral events regulated by the p38 signalling pathway, with a special emphasis on targeting these events for antiviral drug development which might identify candidates with broad spectrum activity.
Collapse
Affiliation(s)
- Yogesh Chander
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Ram Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Nitin Khandelwal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Biotechnology, GLA University, Mathura, India
| | - Namita Singh
- Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Brij Nandan Shringi
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| | - Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| |
Collapse
|
8
|
Ouwendijk WJD, Dekker LJM, van den Ham HJ, Lenac Rovis T, Haefner ES, Jonjic S, Haas J, Luider TM, Verjans GMGM. Analysis of Virus and Host Proteomes During Productive HSV-1 and VZV Infection in Human Epithelial Cells. Front Microbiol 2020; 11:1179. [PMID: 32547533 PMCID: PMC7273502 DOI: 10.3389/fmicb.2020.01179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/08/2020] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) and varicella-zoster virus (VZV) are two closely related human alphaherpesviruses that persistently infect most adults worldwide and cause a variety of clinically important diseases. Herpesviruses are extremely well adapted to their hosts and interact broadly with cellular proteins to regulate virus replication and spread. However, it is incompletely understood how HSV-1 and VZV interact with the host proteome during productive infection. This study determined the temporal changes in virus and host protein expression during productive HSV-1 and VZV infection in the same cell type. Results demonstrated the temporally coordinated expression of HSV-1 and VZV proteins in infected cells. Analysis of the host proteomes showed that both viruses affected extracellular matrix composition, transcription, RNA processing and cell division. Moreover, the prominent role of epidermal growth factor receptor (EGFR) signaling during productive HSV-1 and VZV infection was identified. Stimulation and inhibition of EGFR leads to increased and decreased virus replication, respectively. Collectively, the comparative temporal analysis of viral and host proteomes in productively HSV-1 and VZV-infected cells provides a valuable resource for future studies aimed to identify target(s) for antiviral therapy development.
Collapse
Affiliation(s)
- Werner J. D. Ouwendijk
- Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
- *Correspondence: Werner J. D. Ouwendijk,
| | | | - Henk-Jan van den Ham
- Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
- Enpicom B.V., ‘s-Hertogenbosch, Netherlands
| | - Tihana Lenac Rovis
- Center for Proteomics and Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Erik S. Haefner
- Experimental and Translational Oncology, University Medical Center Mainz, Mainz, Germany
| | - Stipan Jonjic
- Center for Proteomics and Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jürgen Haas
- Division of Infection and Pathway Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | | |
Collapse
|
9
|
Dupont L, Du L, Poulter M, Choi S, McIntosh M, Reeves MB. Src family kinase activity drives cytomegalovirus reactivation by recruiting MOZ histone acetyltransferase activity to the viral promoter. J Biol Chem 2019; 294:12901-12910. [PMID: 31273084 PMCID: PMC6721939 DOI: 10.1074/jbc.ra119.009667] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/29/2019] [Indexed: 12/25/2022] Open
Abstract
Human cytomegalovirus (HCMV) latency and reactivation rely on a complex interplay between cellular differentiation, cell signaling pathways, and viral gene functions. HCMV reactivation in dendritic cells (DCs) is triggered by IL-6 and extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase signaling. However, activation of the same pathway fails to reactivate HCMV in other myeloid cell types, despite this signaling axis being active in those cells. We hypothesized that IL-6-induced ERK activation initiates the changes in chromatin structure required for viral reactivation but that a concomitant signal is necessary to complete the changes in chromatin structure required for gene expression to occur. Using a differential phosphoproteomics approach in cells that do or do not support IL-6-induced viral reactivation, we identified the concomitant activation of an Src family kinase (SFK), hematopoietic cell kinase (HCK), specifically in DCs in response to IL-6. Pharmacological and genetic inhibition of HCK activity indicated that HCK is required for HCMV reactivation. Furthermore, the HCK/SFK activity was linked to recruitment of the monocytic leukemia zinc finger protein (MOZ) histone acetyltransferase to the viral promoter, which promoted histone acetylation after ERK-mediated histone phosphorylation. Importantly, pharmacological and genetic inhibition of MOZ activity prevented reactivation. These results provide an explanation for the selective activation of viral gene expression in DCs by IL-6, dependent on concomitant SFK and ERK signaling. They also reveal a previously unreported role for SFK activity in the regulation of chromatin structure at promoters in eukaryotic cells via MOZ histone acetyltransferase activity.
Collapse
Affiliation(s)
- Liane Dupont
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Lily Du
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Madeleine Poulter
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Stephanie Choi
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Megan McIntosh
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Matthew B. Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom, Supported by Wellcome Trust Grant WT/204870/Z/16/Z. To whom correspondence should be addressed. Tel.:
44-203-1086783; E-mail:
| |
Collapse
|
10
|
He M, Cheng F, da Silva SR, Tan B, Sorel O, Gruffaz M, Li T, Gao SJ. Molecular Biology of KSHV in Relation to HIV/AIDS-Associated Oncogenesis. Cancer Treat Res 2019; 177:23-62. [PMID: 30523620 DOI: 10.1007/978-3-030-03502-0_2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Discovered in 1994, Kaposi's sarcoma-associated herpesvirus (KSHV) has been associated with four human malignancies including Kaposi's sarcoma, primary effusion lymphoma, a subset of multicentric Castleman's disease, and KSHV inflammatory cytokine syndrome. These malignancies mostly occur in immunocompromised patients including patients with acquired immunodeficiency syndrome and often cause significant mortality because of the lack of effective therapies. Significant progresses have been made to understand the molecular basis of KSHV infection and KSHV-induced oncogenesis in the last two decades. This chapter provides an update on the recent advancements focusing on the molecular events of KSHV primary infection, the mechanisms regulating KSHV life cycle, innate and adaptive immunity, mechanism of KSHV-induced tumorigenesis and inflammation, and metabolic reprogramming in KSHV infection and KSHV-transformed cells.
Collapse
Affiliation(s)
- Meilan He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Océane Sorel
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Marion Gruffaz
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Tingting Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
11
|
Gao R, Li T, Tan B, Ramos da Silva S, Jung JU, Feng P, Gao SJ. FoxO1 Suppresses Kaposi's Sarcoma-Associated Herpesvirus Lytic Replication and Controls Viral Latency. J Virol 2019; 93:JVI.01681-18. [PMID: 30404794 PMCID: PMC6340022 DOI: 10.1128/jvi.01681-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 10/31/2018] [Indexed: 02/05/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) has latent and lytic replication phases, both of which contribute to the development of KSHV-induced malignancies. Among the numerous factors identified to regulate the KSHV life cycle, oxidative stress, caused by imbalanced clearing and production of reactive oxygen species (ROS), has been shown to robustly disrupt KSHV latency and induce viral lytic replication. In this study, we identified an important role of the antioxidant defense factor forkhead box protein O1 (FoxO1) in the KSHV life cycle. Either chemical inhibition of the FoxO1 function or knockdown of FoxO1 expression led to an increase in the intracellular ROS level that was subsequently sufficient to disrupt KSHV latency and induce viral lytic reactivation. On the other hand, treatment with N-acetyl-l-cysteine (NAC), an oxygen free radical scavenger, led to a reduction in the FoxO1 inhibition-induced ROS level and, ultimately, the attenuation of KSHV lytic reactivation. These findings reveal that FoxO1 plays a critical role in keeping KSHV latency in check by maintaining the intracellular redox balance.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with several cancers, including Kaposi's sarcoma (KS). Both the KSHV latent and lytic replication phases are important for the development of KS. Identification of factors regulating the KSHV latent phase-to-lytic phase switch can provide insights into the pathogenesis of KSHV-induced malignancies. In this study, we show that the antioxidant defense factor forkhead box protein O1 (FoxO1) maintains KSHV latency by suppressing viral lytic replication. Inhibition of FoxO1 disrupts KSHV latency and induces viral lytic replication by increasing the intracellular ROS level. Significantly, treatment with an oxygen free radical scavenger, N-acetyl-l-cysteine (NAC), attenuated the FoxO1 inhibition-induced intracellular ROS level and KSHV lytic replication. Our works reveal a critical role of FoxO1 in suppressing KSHV lytic replication, which could be targeted for antiviral therapy.
Collapse
Affiliation(s)
- Ruoyun Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Tingting Li
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Suzane Ramos da Silva
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Pinghui Feng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
12
|
High expression of mitogen-activated and stress-activated protein kinase 1 indicates poor prognosis in patients with glioma. Neuroreport 2018; 29:1249-1255. [PMID: 30020192 DOI: 10.1097/wnr.0000000000001090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mitogen-activated and stress-activated protein kinase 1 (MSK1), which belongs to the subfamily of MAPK-activated protein kinase, plays an important role in cell proliferation and neoplastic transformation. It has been recently reported that MSK1 overexpression was closely related to the progression of some tumors such as colorectal cancer. However, the clinical significance of MSK1 in glioma has not been addressed. To investigate the potential role of MSK1 in glioma, we first examined the expression pattern of MSK1 in glioma tissues and normal brain tissues using quantitative RT-PCR, and the results showing that MSK1 expression was significantly elevated in glioma tissues compared with normal brain tissues. The clinical relevance of MSK1 expression level was then analyzed, and we found that high expression of MSK1 was closely related to the larger tumor size and advanced WHO grade. Univariate and multivariate analyses revealed that glioma patients with higher expression of MSK1 had poorer overall survival, and MSK1 was identified as an independent unfavorable prognosis factor. In addition, the effects of MSK1 on glioma cells were tested through cellular experiments, and we demonstrated that MSK1 can promote proliferation and invasion capacities of tumor cells. In conclusion, patients with glioma with higher MSK1 expression were more predisposed to poorer clinical outcomes and unfavorable prognosis, indicating the potential role of MSK1 as a novel clinical biomarker and therapeutic target.
Collapse
|
13
|
KSHV-induced ligand mediated activation of PDGF receptor-alpha drives Kaposi's sarcomagenesis. PLoS Pathog 2018; 14:e1007175. [PMID: 29985958 PMCID: PMC6053240 DOI: 10.1371/journal.ppat.1007175] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/19/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022] Open
Abstract
Kaposi’s sarcoma (KS) herpesvirus (KSHV) causes KS, an angiogenic AIDS-associated spindle-cell neoplasm, by activating host oncogenic signaling cascades through autocrine and paracrine mechanisms. Tyrosine kinase receptor (RTK) proteomic arrays, identified PDGF receptor-alpha (PDGFRA) as the predominantly-activated RTK in KSHV-induced mouse KS-tumors. We show that: 1) KSHV lytic replication and the vGPCR can activate PDGFRA through upregulation of its ligands PDGFA/B, which increase c-myc, VEGF and KSHV gene expression in infected cells 2) KSHV infected spindle cells of most AIDS-KS lesions display robust phospho-PDGFRA staining 3) blocking PDGFRA-signaling with N-acetyl-cysteine, RTK-inhibitors Imatinib and Sunitinib, or dominant-negative PDGFRA inhibits tumorigenesis 4) PDGFRA D842V activating-mutation confers resistance to Imatinib in mouse-KS tumorigenesis. Our data show that KSHV usurps sarcomagenic PDGFRA signaling to drive KS. This and the fact that PDGFRA drives non-viral sarcomas highlights the importance for KSHV-induced ligand-mediated activation of PDGFRA in KS sarcomagenesis and shows that this oncogenic axis could be successfully blocked to impede KS tumor growth. Signaling mimicry is a key mechanism whereby oncoviruses can usurp host-regulatory pathways leading to acquisition of tissue-specific cancer hallmarks. A critical question in the KS field is the identification of this host pathways activated by KSHV that could provide novel insights on KSHV-pathobiology, elucidating new druggable pathways. Here we show that KSHV lytic replication as well as the KSHV-oncogene vGPCR activates PDGFRA signaling through upregulation of its ligands PDGFA/B, and that blocking of PDGFRA signaling is anti-tumorigenic. This indicates that approaches that fully and stably inhibit PDGFR-signaling could lead to successful treatments for KS, validating this receptor-ligand signaling-axis as a therapeutic target.
Collapse
|
14
|
Effects of CREB1 gene silencing on cognitive dysfunction by mediating PKA-CREB signaling pathway in mice with vascular dementia. Mol Med 2018; 24:18. [PMID: 30134805 PMCID: PMC6016877 DOI: 10.1186/s10020-018-0020-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022] Open
Abstract
Background As a form of dementia primarily affecting the elderly, vascular dementia (VD) is characterized by changes in the supply of blood to the brain, resulting in cognitive impairment. The aim of the present study was to explore the effects involved with cyclic adenosine monophosphate (cAMP) response element-binding (CREB)1 gene silencing on cognitive dysfunction through meditation of the protein kinase A (PKA)-CREB signaling pathway in mice with VD. Methods Both the Morris water maze test and the step down test were applied to assess the cognitive function of the mice with VD. Immunohistochemical and TUNEL staining techniques were employed to evaluate the positive expression rates of the protein CREB1 and Cleaved Caspase-3, as well as neuronal apoptosis among hippocampal tissues in a respective manner. Flow cytometry was applied to determine the proliferation index and apoptosis rate of the hippocampal cells among each group. Reverse transcription quantitative polymerase chain reaction and Western blot analysis methods were applied to detect the expressions of cAMP, PKA and CREB in hippocampal cells. Results Compared with the normal group, all the other groups exhibited impaired cognitive function, reduced cell numbers in the CAI area, positive expressions of CREB1 as well as positive optical density (OD) values. Furthermore, increased Cleaved Caspase-3 positive expression, OD value, proliferation index, apoptosis rate of hippocampal cells and neurons, were observed in the other groups when compared with the normal group, as well as lower expressions of cAMP, PKA and CREB1 and p-CREB1 (the shCREB1–1, H89 and shCREB1–1 + H89 groups < the VD group). Conclusion The key findings of the present study demonstrated that CREB1 gene silencing results in aggravated VD that occurs as a result of inhibiting the PKA-CREB signaling pathway, thus exasperating cognitive dysfunction.
Collapse
|
15
|
Berard A, Kroeker A, McQueen P, Coombs KM. Methods and approaches to disease mechanisms using systems kinomics. Synth Syst Biotechnol 2018; 3:34-43. [PMID: 29911197 PMCID: PMC5884222 DOI: 10.1016/j.synbio.2017.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/04/2017] [Accepted: 12/13/2017] [Indexed: 02/06/2023] Open
Abstract
All cellular functions, ranging from regular cell maintenance and homeostasis, specialized functions specific to cellular types, or generating responses due to external stimulus, are mediated by proteins within the cell. Regulation of these proteins allows the cell to alter its behavior under different circumstances. A major mechanism of protein regulation is utilizing protein kinases and phosphatases; enzymes that catalyze the transfer of phosphates between substrates [1]. Proteins involved in phosphate signaling are well studied and include kinases and phosphatases that catalyze opposing reactions regulating both structure and function of the cell. Kinomics is the study of kinases, phosphatases and their targets, and has been used to study the functional changes in numerous diseases and infectious diseases with aims to delineate the cellular functions affected. Identifying the phosphate signaling pathways changed by certain diseases or infections can lead to novel therapeutic targets. However, a daunting 518 putative protein kinase genes have been identified [2], indicating that this protein family is very large and complex. Identifying which enzymes are specific to a particular disease can be a laborious task. In this review, we will provide information on large-scale systems biology methodologies that allow global screening of the kinome to more efficiently identify which kinase pathways are pertinent for further study.
Collapse
Affiliation(s)
- Alicia Berard
- Department of Medical Microbiology, University of Manitoba, Winnipeg, R3E 0J9, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | | | - Peter McQueen
- Department of Medical Microbiology, University of Manitoba, Winnipeg, R3E 0J9, Canada
- JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Canada
| | - Kevin M. Coombs
- Department of Medical Microbiology, University of Manitoba, Winnipeg, R3E 0J9, Canada
| |
Collapse
|
16
|
Cheng F, Ramos da Silva S, Huang IC, Jung JU, Gao SJ. Suppression of Zika Virus Infection and Replication in Endothelial Cells and Astrocytes by PKA Inhibitor PKI 14-22. J Virol 2018; 92:JVI.02019-17. [PMID: 29212931 PMCID: PMC5790943 DOI: 10.1128/jvi.02019-17] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 02/05/2023] Open
Abstract
The recent outbreak of Zika virus (ZIKV), a reemerging flavivirus, and its associated neurological disorders, such as Guillain-Barré (GB) syndrome and microcephaly, have generated an urgent need to develop effective ZIKV vaccines and therapeutic agents. Here, we used human endothelial cells and astrocytes, both of which represent key cell types for ZIKV infection, to identify potential inhibitors of ZIKV replication. Because several pathways, including the AMP-activated protein kinase (AMPK), protein kinase A (PKA), and mitogen-activated protein kinase (MAPK) signaling pathways, have been reported to play important roles in flavivirus replication, we tested inhibitors and agonists of these pathways for their effects on ZIKV replication. We identified the PKA inhibitor PKI 14-22 (PKI) to be a potent inhibitor of ZIKV replication. PKI effectively suppressed the replication of ZIKV from both the African and Asian/American lineages with a high efficiency and minimal cytotoxicity. While ZIKV infection does not induce PKA activation, endogenous PKA activity is essential for supporting ZIKV replication. Interestingly, in addition to PKA, PKI also inhibited another unknown target(s) to block ZIKV replication. PKI inhibited ZIKV replication at the postentry stage by preferentially affecting negative-sense RNA synthesis as well as viral protein translation. Together, these results have identified a potential inhibitor of ZIKV replication which could be further explored for future therapeutic application.IMPORTANCE There is an urgent need to develop effective vaccines and therapeutic agents against Zika virus (ZIKV) infection, a reemerging flavivirus associated with neurological disorders, including Guillain-Barré (GB) syndrome and microcephaly. By screening for inhibitors of several cellular pathways, we have identified the PKA inhibitor PKI 14-22 (PKI) to be a potent inhibitor of ZIKV replication. We show that PKI effectively suppresses the replication of all ZIKV strains tested with minimal cytotoxicity to human endothelial cells and astrocytes, two key cell types for ZIKV infection. Furthermore, we show that PKI inhibits ZIKV negative-sense RNA synthesis and viral protein translation. This study has identified a potent inhibitor of ZIKV infection which could be further explored for future therapeutic application.
Collapse
Affiliation(s)
- Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - I-Chueh Huang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
17
|
Tan B, Liu H, Zhang S, da Silva SR, Zhang L, Meng J, Cui X, Yuan H, Sorel O, Zhang SW, Huang Y, Gao SJ. Viral and cellular N 6-methyladenosine and N 6,2'-O-dimethyladenosine epitranscriptomes in the KSHV life cycle. Nat Microbiol 2018; 3:108-120. [PMID: 29109479 PMCID: PMC6138870 DOI: 10.1038/s41564-017-0056-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 10/09/2017] [Indexed: 02/05/2023]
Abstract
N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am) modifications (m6A/m) of messenger RNA mediate diverse cellular functions. Oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) has latent and lytic replication phases that are essential for the development of KSHV-associated cancers. To date, the role of m6A/m in KSHV replication and tumorigenesis is unclear. Here, we provide mechanistic insights by examining the viral and cellular m6A/m epitranscriptomes during KSHV latent and lytic infection. KSHV transcripts contain abundant m6A/m modifications during latent and lytic replication, and these modifications are highly conserved among different cell types and infection systems. Knockdown of YTHDF2 enhanced lytic replication by impeding KSHV RNA degradation. YTHDF2 binds to viral transcripts and differentially mediates their stability. KSHV latent infection induces 5' untranslated region (UTR) hypomethylation and 3'UTR hypermethylation of the cellular epitranscriptome, regulating oncogenic and epithelial-mesenchymal transition pathways. KSHV lytic replication induces dynamic reprogramming of epitranscriptome, regulating pathways that control lytic replication. These results reveal a critical role of m6A/m modifications in KSHV lifecycle and provide rich resources for future investigations.
Collapse
Affiliation(s)
- Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hui Liu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- School of Information and Electrical Engineering, China University of Mining and Technology, Xuzhou, Jiangsu, China
| | - Songyao Zhang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA
- School of Automation, Northwestern Polytechnic University, Xi'an, Shaanxi, China
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lin Zhang
- School of Information and Electrical Engineering, China University of Mining and Technology, Xuzhou, Jiangsu, China
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Jia Meng
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, China
| | - Xiaodong Cui
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Hongfeng Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Océane Sorel
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shao-Wu Zhang
- School of Automation, Northwestern Polytechnic University, Xi'an, Shaanxi, China
| | - Yufei Huang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, USA.
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
18
|
Yuan H, He M, Cheng F, Bai R, da Silva SR, Aguiar RCT, Gao SJ. Tenovin-6 inhibits proliferation and survival of diffuse large B-cell lymphoma cells by blocking autophagy. Oncotarget 2017; 8:14912-14924. [PMID: 28118604 PMCID: PMC5362454 DOI: 10.18632/oncotarget.14741] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is one of the most aggressive non-Hodgkin lymphomas. It is curable but one-third of cases are refractory to therapy or relapse after initial response highlighting the urgent need for developing novel therapeutic approaches. Targeting sirtuins, particularly SIRT1 by genetic approaches or using pharmaceutical inhibitor tenovin-6, has shown promising therapeutic potential in various hematopoietic malignancies. However, it remains unknown whether these approaches are effective for DLBCL. In this study, we have found that tenovin-6 potently inhibits the proliferation and survival of DLBCL cells. Surprisingly, specific knockdown of SIRT1/2/3 has no effect on DLBCL. Mechanistically, tenovin-6 increases the level of microtubule-associated protein 1 light chain 3B (LC3B)-II in a SIRT1/2/3- and p53-independent manner in DLBCL cell lines. Tenovin-6-mediated increase of LC3B-II is through inhibition of classical autophagy pathway. Furthermore, inhibition of the autophagy pathway by using other inhibitors or by knocking down key genes in the pathway impairs cell proliferation and survival of DLBCL cells. These results indicate that targeting the autophagic pathway could be a novel therapeutic strategy for DLBCL and that precaution should be taken to interpret data where tenovin-6 was used as an inhibitor of sirtuins.
Collapse
Affiliation(s)
- Hongfeng Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Meilan He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rosemary Bai
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ricardo C T Aguiar
- Department of Medicine and Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,South Texas Veterans Health Care System, Audie Murphy VA Hospital, San Antonio, TX, USA
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
19
|
Ho J, Moyes DL, Tavassoli M, Naglik JR. The Role of ErbB Receptors in Infection. Trends Microbiol 2017; 25:942-952. [PMID: 28522156 PMCID: PMC7126822 DOI: 10.1016/j.tim.2017.04.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 04/11/2017] [Accepted: 04/24/2017] [Indexed: 01/18/2023]
Abstract
Members of the epidermal growth factor receptor family (ErbB family) possess a wide distribution and diverse functions ranging from cellular growth to migration and apoptosis. Though highly implicated in a variety of cancers, their involvement in infectious disease is less recognised. A growing body of evidence now highlights the importance of the ErbB family in a variety of infections. Their role as growth factor receptors, along with other characteristics, such as surface expression and continuous intracellular trafficking, make this receptor family ideally placed for exploitation by pathogens. Herein, we review our current understanding of the role of the ErbB family in the context of infectious disease, exploring the mechanisms that govern pathogen exploitation of this system. A wide and diverse variety of microbes have each evolved distinct mechanisms to exploit ErbB receptors, highlighting this receptor kinase family as a critical factor in initiation and maintenance of pathogen infections. ErbB family members are utilised by pathogens attempting to gain cellular entry, subvert immune responses, and manipulate the cell cycle of infected host cells. These events support and are necessary for pathogen persistence. Pathogen-mediated ErbB-exploitation may contribute to cellular transformation and oncogenesis in a variety of cancers. The use of existing FDA-approved drugs that target ErbB receptors and associated signalling components may offer potential future therapies against infection.
Collapse
Affiliation(s)
- Jemima Ho
- Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK.
| | - David L Moyes
- Centre for Host Microbiome interactions, Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Mahvash Tavassoli
- Department of Molecular Oncology, Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Julian R Naglik
- Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| |
Collapse
|
20
|
He M, Tan B, Vasan K, Yuan H, Cheng F, Ramos da Silva S, Lu C, Gao SJ. SIRT1 and AMPK pathways are essential for the proliferation and survival of primary effusion lymphoma cells. J Pathol 2017; 242:309-321. [PMID: 28393364 DOI: 10.1002/path.4905] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/20/2017] [Accepted: 03/20/2017] [Indexed: 12/19/2022]
Abstract
Primary effusion lymphoma (PEL) is a rare and aggressive B-cell lymphoma with a dismal prognosis caused by infection of Kaposi's sarcoma-associated herpesvirus. Despite the findings that numerous viral genes and cellular pathways are essential for the proliferation and survival of PEL cells, there is currently no effective therapeutic treatment for PEL. Here, we report that the metabolic sensor SIRT1 is functionally required for sustaining the proliferation and survival of PEL cells. Knockdown of SIRT1 with specific shRNAs or inhibition of SIRT1 with an inhibitor (tenovin-6) induced cell cycle arrest and apoptosis in PEL cells. We detected high levels of AMPK activation in PEL cells, reflected in AMPKα1 phosphorylation at T174. Knockdown or inhibition of SIRT1 reduced AMPK activation, indicating that SIRT1 was required for AMPK activation. Interestingly, knockdown of AMPK with specific shRNAs or inhibition of AMPK with the inhibitor compound C recapitulated the phenotype of SIRT1, and induced cell cycle arrest and apoptosis, whereas overexpression of a constitutively active AMPK construct rescued the cytotoxic effect of SIRT1 knockdown. Remarkably, treatment with tenovin-6 effectively inhibited the initiation and progression of PEL, and significantly extended the survival of mice in a murine PEL model. Taken together, these results illustrate that the SIRT1-AMPK axis is essential for maintaining the proliferation and survival of PEL and identify SIRT1 and AMPK as potential therapeutic targets, and tenovin-6 as a candidate therapeutic agent for PEL patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Meilan He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brandon Tan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Karthik Vasan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hongfeng Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Fan Cheng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chun Lu
- Department of Microbiology, Nanjing Medical University, Nanjing, PR China
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
21
|
Li S, Bai L, Dong J, Sun R, Lan K. Kaposi's Sarcoma-Associated Herpesvirus: Epidemiology and Molecular Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1018:91-127. [PMID: 29052134 DOI: 10.1007/978-981-10-5765-6_7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV), also known as Human herpesvirus 8 (HHV-8), is a member of the lymphotropic gammaherpesvirus subfamily and a human oncogenic virus. Since its discovery in AIDS-associated KS tissues by Drs. Yuan Chang and Patrick Moore, much progress has been made in the past two decades. There are four types of KS including classic KS, endemic KS, immunosuppressive therapy-related KS, and AIDS-associated KS. In addition to KS, KSHV is also involved in the development of primary effusion lymphoma (PEL) and certain types of multicentric Castleman's disease. KSHV manipulates numerous viral proteins to promote the progression of angiogenesis and tumorigenesis. In this chapter, we review the epidemiology and molecular biology of KSHV and the mechanisms underlying KSHV-induced diseases.
Collapse
Affiliation(s)
- Shasha Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Lei Bai
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Jiazhen Dong
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Rui Sun
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, People's Republic of China.
| |
Collapse
|
22
|
Kaposi's sarcoma-associated herpesvirus: the role of lytic replication in targeted therapy. Curr Opin Infect Dis 2016; 28:611-24. [PMID: 26524334 DOI: 10.1097/qco.0000000000000213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW To discuss the role of Kaposi's sarcoma-associated herpesvirus (KSHV) lytic replication in viral-associated diseases and assess the progress on targeting KSHV lytic replication as a strategy to prevent KSHV-related malignancies. RECENT FINDINGS New inhibitors of viral lytic replication are being developed as well as novel modalities are being investigated to target cellular processes that the virus hijacks during its life cycle. Research has also focused on reactivating viral lytic replication in latently infected tumour cells (lytic induction therapy) to promote death of tumour cells. SUMMARY KSHV is linked to three malignancies: Kaposi sarcoma, primary effusion lymphoma, and multicentric Castleman disease. Despite significant progress in understanding KSHV pathobiology, no therapeutic guidelines for the management of KSHV-related diseases exist, and current treatments are suboptimal and associated with toxicity. Antiherpesvirus drugs have shown inconsistent results in KSHV-associated malignancies that harbour the virus in a latent state. However, lytic replication plays a crucial role in the process of tumorigenesis. Therefore, not only antiviral agents directed against the virus replicative cycle but also agents that target cellular processes that are activated by the virus are being investigated. Antivirals may also be used in combination with inducers of the viral lytic stage.
Collapse
|
23
|
Atak A, Mukherjee S, Jain R, Gupta S, Singh VA, Gahoi N, K P M, Srivastava S. Protein microarray applications: Autoantibody detection and posttranslational modification. Proteomics 2016; 16:2557-2569. [PMID: 27452627 DOI: 10.1002/pmic.201600104] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 07/09/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022]
Abstract
The discovery of DNA microarrays was a major milestone in genomics; however, it could not adequately predict the structure or dynamics of underlying protein entities, which are the ultimate effector molecules in a cell. Protein microarrays allow simultaneous study of thousands of proteins/peptides, and various advancements in array technologies have made this platform suitable for several diagnostic and functional studies. Antibody arrays enable researchers to quantify the abundance of target proteins in biological fluids and assess PTMs by using the antibodies. Protein microarrays have been used to assess protein-protein interactions, protein-ligand interactions, and autoantibody profiling in various disease conditions. Here, we summarize different microarray platforms with focus on its biological and clinical applications in autoantibody profiling and PTM studies. We also enumerate the potential of tissue microarrays to validate findings from protein arrays as well as other approaches, highlighting their significance in proteomics.
Collapse
Affiliation(s)
- Apurva Atak
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shuvolina Mukherjee
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Rekha Jain
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shabarni Gupta
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Vedita Anand Singh
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Nikita Gahoi
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Manubhai K P
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Sanjeeva Srivastava
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| |
Collapse
|
24
|
Suppression of Kaposi's Sarcoma-Associated Herpesvirus Infection and Replication by 5'-AMP-Activated Protein Kinase. J Virol 2016; 90:6515-6525. [PMID: 27147746 DOI: 10.1128/jvi.00624-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 04/28/2016] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED The host intracellular antiviral restriction factors inhibit viral infection and replication. The 5'-AMP-activated protein kinase (AMPK) is a cellular energy sensor regulating metabolic homeostasis. Activated AMPK inhibits the replication of numerous RNA viruses but enhances the entry of vaccinia virus. However, the role of AMPK in herpesvirus infection is unclear. In this study, we showed that the constitutive AMPK activity restricted Kaposi's sarcoma-associated herpesvirus (KSHV) lytic replication in primary human umbilical vein endothelial cells while KSHV infection did not markedly affect the endogenous AMPK activity. Knockdown of the AMPKα1 considerably enhanced the expression of viral lytic genes and the production of infectious virions, while overexpression of a constitutively active AMPK had the opposite effects. Accordingly, an AMPK inhibitor, compound C, augmented viral lytic gene expressions and virion productions but an AMPK agonist, 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), suppressed both. Furthermore, a common diabetes drug, metformin, which carries an AMPK-agonistic activity, drastically inhibited the expression of viral lytic genes and the production of infectious virions, suggesting the use of metformin as a therapeutic agent for KSHV infection and replication. Together, these results identify the host AMPK as a KSHV restriction factor that can serve as a potential therapeutic target. IMPORTANCE Host cells encode specific proteins to restrict viral infection and replication. Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumor virus associated with several cancers. In this study, we have identified 5'-AMP-activated protein kinase (AMPK), a cellular energy sensor, as a restriction factor of KSHV lytic replication during primary infection. Activation of AMPK suppresses, while inhibition of AMPK enhances, KSHV lytic replication by regulating the expression of viral genes. AICAR and metformin, both of which are AMPK agonists currently used in clinics for the treatment of conditions associated with metabolic disorders, inhibit KSHV lytic replication. Thus, our work has identified AMPK as a potential therapeutic target and AICAR and metformin as potential therapeutic agents for KSHV-associated cancers.
Collapse
|
25
|
Reyskens KMSE, Arthur JSC. Emerging Roles of the Mitogen and Stress Activated Kinases MSK1 and MSK2. Front Cell Dev Biol 2016; 4:56. [PMID: 27376065 PMCID: PMC4901046 DOI: 10.3389/fcell.2016.00056] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/30/2016] [Indexed: 01/17/2023] Open
Abstract
Mitogen- and stress-activated kinases (MSK) 1 and 2 are nuclear proteins activated downstream of the ERK1/2 or p38 MAPK pathways. MSKs phosphorylate multiple substrates, including CREB and Histone H3, and their major role is the regulation of specific subsets of Immediate Early genes (IEG). While MSKs are expressed in multiple tissues, their levels are high in immune and neuronal cells and it is in these systems most is known about their function. In immunity, MSKs have predominantly anti-inflammatory roles and help regulate production of the anti-inflammatory cytokine IL-10. In the CNS they are implicated in neuronal proliferation and synaptic plasticity. In this review we will focus on recent advances in understanding the roles of MSKs in the innate immune system and neuronal function.
Collapse
Affiliation(s)
- Kathleen M S E Reyskens
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee Dundee, UK
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee Dundee, UK
| |
Collapse
|
26
|
Purushothaman P, Dabral P, Gupta N, Sarkar R, Verma SC. KSHV Genome Replication and Maintenance. Front Microbiol 2016; 7:54. [PMID: 26870016 PMCID: PMC4740845 DOI: 10.3389/fmicb.2016.00054] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/12/2016] [Indexed: 12/04/2022] Open
Abstract
Kaposi's sarcoma associated herpesvirus (KSHV) or human herpesvirus 8 (HHV8) is a major etiological agent for multiple severe malignancies in immune-compromised patients. KSHV establishes lifetime persistence in the infected individuals and displays two distinct life cycles, generally a prolonged passive latent, and a short productive or lytic cycle. During latent phase, the viral episome is tethered to the host chromosome and replicates once during every cell division. Latency-associated nuclear antigen (LANA) is a predominant multifunctional nuclear protein expressed during latency, which plays a central role in episome tethering, replication and perpetual segregation of the episomes during cell division. LANA binds cooperatively to LANA binding sites (LBS) within the terminal repeat (TR) region of the viral episome as well as to the cellular nucleosomal proteins to tether viral episome to the host chromosome. LANA has been shown to modulate multiple cellular signaling pathways and recruits various cellular proteins such as chromatin modifying enzymes, replication factors, transcription factors, and cellular mitotic framework to maintain a successful latent infection. Although, many other regions within the KSHV genome can initiate replication, KSHV TR is important for latent DNA replication and possible segregation of the replicated episomes. Binding of LANA to LBS favors the recruitment of various replication factors to initiate LANA dependent DNA replication. In this review, we discuss the molecular mechanisms relevant to KSHV genome replication, segregation, and maintenance of latency.
Collapse
Affiliation(s)
- Pravinkumar Purushothaman
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Prerna Dabral
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Namrata Gupta
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Roni Sarkar
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, Reno Reno, NV, USA
| | - Subhash C Verma
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, Reno Reno, NV, USA
| |
Collapse
|