1
|
Abou Staiteieh S, Faddoul N, Hayar B, Houshaymi B, Darwiche N, Abou Merhi R. Primary effusion lymphoma: therapeutic strategies targeting viral and cellular mechanisms. Expert Rev Anticancer Ther 2025; 25:363-381. [PMID: 40033827 DOI: 10.1080/14737140.2025.2474728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/31/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Primary effusion lymphoma (PEL) is a rare subtype of B-cell lymphoma primarily affecting immunocompromised and elderly individuals. Given the dismal survival rates associated with traditional treatments, studying novel therapeutic approaches to improve patient outcomes is critical. AREAS COVERED This review focuses on developing therapeutic options for PEL that target particular viral and cellular mechanisms involved in PEL pathogenesis. Since the CHOP regimen was associated with a lower median survival rate, alternative treatments, including stem cell transplantation, have also been explored, but have generally produced unsatisfactory results. Therefore, novel therapeutic agents are under investigation, including antiretroviral drugs targeting viral pathways and treatments targeting particular cellular processes, such as DNA damage, epigenetics, apoptotic, and immune-modulatory pathways showing promising outcomes in preclinical and clinical research, increasing PEL treatment efficacy while minimizing toxicity. In this review, we conducted a comprehensive literature search using PubMed, and Google Scholar for studies published between 1989 and 2024. EXPERT OPINION Further research is needed to refine the appropriate combination methods and strategies behind drug interactions. Targeted therapies could be investigated further to improve therapeutic efficacy and reduce toxicity in this type of lymphoma.
Collapse
Affiliation(s)
- Soumaiah Abou Staiteieh
- Genomics and Surveillance Biotherapy Laboratory, Biology Department, Faculty of Sciences, R. Hariri Campus, Lebanese University, Hadath, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Nouria Faddoul
- Genomics and Surveillance Biotherapy Laboratory, Biology Department, Faculty of Sciences, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| | - Berthe Hayar
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Bilal Houshaymi
- Genomics and Surveillance Biotherapy Laboratory, Biology Department, Faculty of Sciences, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Raghida Abou Merhi
- Genomics and Surveillance Biotherapy Laboratory, Biology Department, Faculty of Sciences, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| |
Collapse
|
2
|
Dutta S, Ganguly A, Ghosh Roy S. An Overview of the Unfolded Protein Response (UPR) and Autophagy Pathways in Human Viral Oncogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:81-131. [PMID: 38782502 DOI: 10.1016/bs.ircmb.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Autophagy and Unfolded Protein Response (UPR) can be regarded as the safe keepers of cells exposed to intense stress. Autophagy maintains cellular homeostasis, ensuring the removal of foreign particles and misfolded macromolecules from the cytoplasm and facilitating the return of the building blocks into the system. On the other hand, UPR serves as a shock response to prolonged stress, especially Endoplasmic Reticulum Stress (ERS), which also includes the accumulation of misfolded proteins in the ER. Since one of the many effects of viral infection on the host cell machinery is the hijacking of the host translational system, which leaves in its wake a plethora of misfolded proteins in the ER, it is perhaps not surprising that UPR and autophagy are common occurrences in infected cells, tissues, and patient samples. In this book chapter, we try to emphasize how UPR, and autophagy are significant in infections caused by six major oncolytic viruses-Epstein-Barr (EBV), Human Papilloma Virus (HPV), Human Immunodeficiency Virus (HIV), Human Herpesvirus-8 (HHV-8), Human T-cell Lymphotropic Virus (HTLV-1), and Hepatitis B Virus (HBV). Here, we document how whole-virus infection or overexpression of individual viral proteins in vitro and in vivo models can regulate the different branches of UPR and the various stages of macro autophagy. As is true with other viral infections, the relationship is complicated because the same virus (or the viral protein) exerts different effects on UPR and Autophagy. The nature of this response is determined by the cell types, or in some cases, the presence of diverse extracellular stimuli. The vice versa is equally valid, i.e., UPR and autophagy exhibit both anti-tumor and pro-tumor properties based on the cell type and other factors like concentrations of different metabolites. Thus, we have tried to coherently summarize the existing knowledge, the crux of which can hopefully be harnessed to design vaccines and therapies targeted at viral carcinogenesis.
Collapse
Affiliation(s)
- Shovan Dutta
- Center for Immunotherapy & Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar, Jharkhand, India
| | - Sounak Ghosh Roy
- Henry M Jackson for the Advancement of Military Medicine, Naval Medical Research Command, Silver Spring, MD, United States.
| |
Collapse
|
3
|
Wen KW, Wang L, Menke JR, Damania B. Cancers associated with human gammaherpesviruses. FEBS J 2022; 289:7631-7669. [PMID: 34536980 PMCID: PMC9019786 DOI: 10.1111/febs.16206] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/10/2021] [Accepted: 09/16/2021] [Indexed: 01/14/2023]
Abstract
Epstein-Barr virus (EBV; human herpesvirus 4; HHV-4) and Kaposi sarcoma-associated herpesvirus (KSHV; human herpesvirus 8; HHV-8) are human gammaherpesviruses that have oncogenic properties. EBV is a lymphocryptovirus, whereas HHV-8/KSHV is a rhadinovirus. As lymphotropic viruses, EBV and KSHV are associated with several lymphoproliferative diseases or plasmacytic/plasmablastic neoplasms. Interestingly, these viruses can also infect epithelial cells causing carcinomas and, in the case of KSHV, endothelial cells, causing sarcoma. EBV is associated with Burkitt lymphoma, classic Hodgkin lymphoma, nasopharyngeal carcinoma, plasmablastic lymphoma, lymphomatoid granulomatosis, leiomyosarcoma, and subsets of diffuse large B-cell lymphoma, post-transplant lymphoproliferative disorder, and gastric carcinoma. KSHV is implicated in Kaposi sarcoma, primary effusion lymphoma, multicentric Castleman disease, and KSHV-positive diffuse large B-cell lymphoma. Pathogenesis by these two herpesviruses is intrinsically linked to viral proteins expressed during the lytic and latent lifecycles. This comprehensive review intends to provide an overview of the EBV and KSHV viral cycles, viral proteins that contribute to oncogenesis, and the current understanding of the pathogenesis and clinicopathology of their related neoplastic entities.
Collapse
Affiliation(s)
- Kwun Wah Wen
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158
| | - Linlin Wang
- Department of Laboratory Medicine, University of California, San Francisco, CA 94158
| | - Joshua R. Menke
- Department of Pathology, Stanford University, Palo Alto, CA 94304
| | - Blossom Damania
- Department of Microbiology & Immunology & Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
4
|
Kadota A, Moriguchi M, Watanabe T, Sekine Y, Nakamura S, Yasuno T, Ohe T, Mashino T, Fujimuro M. A pyridinium‑type fullerene derivative suppresses primary effusion lymphoma cell viability via the downregulation of the Wnt signaling pathway through the destabilization of β‑catenin. Oncol Rep 2022; 47:46. [PMID: 35014678 PMCID: PMC8771160 DOI: 10.3892/or.2022.8257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/18/2021] [Indexed: 11/16/2022] Open
Abstract
Primary effusion lymphoma (PEL) is defined as a rare subtype of non-Hodgkin's B cell lymphoma, which is caused by Kaposi's sarcoma-associated herpesvirus (KSHV) in immunosuppressed patients. PEL is an aggressive type of lymphoma and is frequently resistant to conventional chemotherapeutics. Therefore, the discovery of novel drug candidates for the treatment of PEL is of utmost importance. In order to discover potential novel anti-tumor compounds against PEL, the authors previously developed a pyrrolidinium-type fullerene derivative, 1,1,1′,1′-tetramethyl [60]fullerenodipyrrolidinium diiodide (derivative #1), which induced the apoptosis of PEL cells via caspase-9 activation. In the present study, the growth inhibitory effects of pyrrolidinium-type (derivatives #1 and #2), pyridinium-type (derivatives #3 and #5 to #9) and anilinium-type fullerene derivatives (derivative #4) against PEL cells were evaluated. This analysis revealed a pyridinium-type derivative (derivative #5; 3- 5′-(etho-xycarbonyl)-1′,5′-dihydro-2′H-[5,6]fullereno-C60-Ih-[1,9-c]pyrrol-2′-yl]-1-methylpyridinium iodide), which exhibited antitumor activity against PEL cells via the downregulation of Wnt/β-catenin signaling. Derivative #5 suppressed the viability of KSHV-infected PEL cells compared with KSHV-uninfected B-lymphoma cells. Furthermore, derivative #5 induced the destabilization of β-catenin and suppressed β-catenin-TCF4 transcriptional activity in PEL cells. It is known that the constitutive activation of Wnt/β-catenin signaling is essential for the growth of KSHV-infected cells. The Wnt/β-catenin activation in KSHV-infected cells is mediated by KSHV latency-associated nuclear antigen (LANA). The data demonstrated that derivative #5 increased β-catenin phosphorylation, which resulted in β-catenin polyubiquitination and subsequent degradation. Thus, derivative #5 overcame LANA-mediated β-catenin stabilization. Furthermore, the administration of derivative #5 suppressed the development of PEL cells in the ascites of SCID mice with tumor xenografts derived from PEL cells. On the whole, these findings provide evidence that the pyridinium-type fullerene derivative #5 exhibits antitumor activity against PEL cells in vitro and in vivo. Thus, derivative #5 may be utilized as a novel therapeutic agent for the treatment of PEL.
Collapse
Affiliation(s)
- Ayano Kadota
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607‑8412, Japan
| | - Misato Moriguchi
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607‑8412, Japan
| | - Tadashi Watanabe
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607‑8412, Japan
| | - Yuichi Sekine
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607‑8412, Japan
| | - Shigeo Nakamura
- Department of Chemistry, Nippon Medical School, Musashino, Tokyo 180‑0023, Japan
| | - Takumi Yasuno
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, Tokyo 105‑8512, Japan
| | - Tomoyuki Ohe
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, Tokyo 105‑8512, Japan
| | - Tadahiko Mashino
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Keio University, Tokyo 105‑8512, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607‑8412, Japan
| |
Collapse
|
5
|
Calderon A, Soldan SS, De Leo A, Deng Z, Frase DM, Anderson EM, Zhang Y, Vladimirova O, Lu F, Leung JC, Murphy ME, Lieberman PM. Identification of Mubritinib (TAK 165) as an inhibitor of KSHV driven primary effusion lymphoma via disruption of mitochondrial OXPHOS metabolism. Oncotarget 2020; 11:4224-4242. [PMID: 33245718 PMCID: PMC7679036 DOI: 10.18632/oncotarget.27815] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
KSHV-associated cancers have poor prognoses and lack therapeutics that selectively target viral gene functions. We developed a screening campaign to identify known drugs that could be repurposed for the treatment of KSHV-associated cancers. We focused on primary effusion lymphoma (PEL), which has particularly poor treatment outcomes. We developed a luciferase reporter assay to test the ability of drugs to inhibit DNA binding of the KSHV LANA DNA binding domain (DBD). In parallel, we screened drugs for selective inhibition of a KSHV+ PEL cells. While potent hits were identified in each assay, only one hit, Mubritinib, was found to score in both assays. Mubritinib caused PEL cells to undergo cell cycle arrest with accumulation of sub-G1 population and Annexin V. Mubritinib inhibited LANA binding to KSHV terminal repeat (TR) DNA in KSHV+ PEL cells, but did not lead to KSHV lytic cycle reactivation. Mubritinib was originally identified as a receptor tyrosine kinase (RTK) inhibitor selective for HER2/ErbB2. But recent studies have revealed that Mubritinib can also inhibit the electron transport chain (ETC) complex at nanomolar concentrations. We found that other related ETC complex inhibitors (Rotenone and Deguelin) exhibited PEL cell growth inhibition while RTK inhibitors failed. Seahorse analysis demonstrated that Mubritinib selectively inhibits the maximal oxygen consumption (OCR) in PEL cells and metabolomics revealed changes in ATP/ADP and ATP/AMP ratios. These findings indicate that PEL cells are selectively sensitive to ETC complex inhibitors and provide a rationale for repurposing Mubritinib for selective treatment of PEL.
Collapse
Affiliation(s)
| | | | | | - Zhong Deng
- The Wistar Institute, Philadelphia, PA 19146, USA
| | | | | | - Yue Zhang
- The Wistar Institute, Philadelphia, PA 19146, USA
| | | | - Fang Lu
- The Wistar Institute, Philadelphia, PA 19146, USA
| | | | | | | |
Collapse
|
6
|
MG132 exerts anti-viral activity against HSV-1 by overcoming virus-mediated suppression of the ERK signaling pathway. Sci Rep 2020; 10:6671. [PMID: 32317666 PMCID: PMC7174428 DOI: 10.1038/s41598-020-63438-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/27/2020] [Indexed: 01/18/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) causes a number of clinical manifestations including cold sores, keratitis, meningitis and encephalitis. Although current drugs are available to treat HSV-1 infection, they can cause side effects such as nephrotoxicity. Moreover, owing to the emergence of drug-resistant HSV-1 strains, new anti-HSV-1 compounds are needed. Because many viruses exploit cellular host proteases and encode their own viral proteases for survival, we investigated the inhibitory effects of a panel of protease inhibitors (TLCK, TPCK, E64, bortezomib, or MG132) on HSV-1 replication and several host cell signaling pathways. We found that HSV-1 infection suppressed c-Raf-MEK1/2-ERK1/2-p90RSK signaling in host cells, which facilitated viral replication. The mechanism by which HSV-1 inhibited ERK signaling was mediated through the polyubiquitination and proteasomal degradation of Ras-guanine nucleotide-releasing factor 2 (Ras-GRF2). Importantly, the proteasome inhibitor MG132 inhibited HSV-1 replication by reversing ERK suppression in infected cells, inhibiting lytic genes (ICP5, ICP27 and UL42) expression, and overcoming the downregulation of Ras-GRF2. These results indicate that the suppression of ERK signaling via proteasomal degradation of Ras-GRF2 is necessary for HSV-1 infection and replication. Given that ERK activation by MG132 exhibits anti-HSV-1 activity, these results suggest that the proteasome inhibitor could serve as a novel therapeutic agent against HSV-1 infection.
Collapse
|
7
|
Ishiura Y, Ishimaru H, Watanabe T, Fujimuro M. Sulforaphane Exhibits Cytotoxic Effects against Primary Effusion Lymphoma Cells by Suppressing p38MAPK and AKT Phosphorylation. Biol Pharm Bull 2020; 42:2109-2112. [PMID: 31787726 DOI: 10.1248/bpb.b19-00659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Primary effusion lymphoma (PEL) is a rare subtype of non-Hodgkin's B-cell lymphoma and is caused by Kaposi's sarcoma-associated herpesvirus (KSHV) in immunosuppressed patients. PEL is an aggressive lymphoma and is frequently resistant to conventional chemotherapies. Sulforaphane (SFN), a natural compound found in cruciferous vegetables and broccoli sprouts, modulates signaling pathways and epigenetic gene expression. However, the anti-proliferative effects of SFN on PEL cells and the underlying mechanisms have not been identified. Here, we found that SFN decreased the viability of KSHV-infected PEL cells compared to KSHV-uninfected B-lymphoma cells. The anti-proliferative effects of SFN on PEL cells were mediated by apoptosis with activating caspases. In addition, SFN inhibited the phosphorylation of p38 mitogen-activated protein kinase (p38MAPK) and AKT in PEL cells. We also showed that p38MAPK and AKT inhibitors reduced PEL cell growth. The constitutive and/or transient activation of p38MAPK and AKT signaling are necessary for the survival and proliferation of PEL cells. Our data and previous literature indicate that SFN represses the phosphorylation of p38MAPK and AKT, which results in PEL cell apoptosis. Moreover, we investigated whether MG132 or sangivamycin (Sangi) in combination with SFN potentiated the cytotoxic effects of SFN on PEL cells. Compared to treatment with SFN alone, the addition of MG132 or Sangi enhanced the cytotoxic activity of SFN in a synergistic manner. In conclusion, the anti-proliferative effects of SFN indicate its potential as a new substance for the treatment of PEL.
Collapse
Affiliation(s)
- Yuki Ishiura
- Department of Cell Biology, Kyoto Pharmaceutical University
| | | | | | | |
Collapse
|
8
|
Johnston BP, McCormick C. Herpesviruses and the Unfolded Protein Response. Viruses 2019; 12:E17. [PMID: 31877732 PMCID: PMC7019427 DOI: 10.3390/v12010017] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Herpesviruses usurp cellular stress responses to promote viral replication and avoid immune surveillance. The unfolded protein response (UPR) is a conserved stress response that is activated when the protein load in the ER exceeds folding capacity and misfolded proteins accumulate. The UPR aims to restore protein homeostasis through translational and transcriptional reprogramming; if homeostasis cannot be restored, the UPR switches from "helper" to "executioner", triggering apoptosis. It is thought that the burst of herpesvirus glycoprotein synthesis during lytic replication causes ER stress, and that these viruses may have evolved mechanisms to manage UPR signaling to create an optimal niche for replication. The past decade has seen considerable progress in understanding how herpesviruses reprogram the UPR. Here we provide an overview of the molecular events of UPR activation, signaling and transcriptional outputs, and highlight key evidence that herpesviruses hijack the UPR to aid infection.
Collapse
Affiliation(s)
- Benjamin P. Johnston
- Department of Microbiology & Immunology, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada;
- Beatrice Hunter Cancer Research Institute, 5850 College Street, Halifax, NS B3H 4R2, Canada
| | - Craig McCormick
- Department of Microbiology & Immunology, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada;
- Beatrice Hunter Cancer Research Institute, 5850 College Street, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
9
|
The Ubiquitin-Proteasome System Is Necessary for Efficient Replication of Human Astrovirus. J Virol 2018; 92:JVI.01809-17. [PMID: 29093085 DOI: 10.1128/jvi.01809-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 12/25/2022] Open
Abstract
Astroviruses, members of the family Astroviridae, represent an important cause of human gastroenteritis in the world. The cellular factors required for astrovirus replication have been poorly studied. In this work, we evaluated the relevance of the ubiquitin-proteasome system (UPS) in the replication of Yuc8, a human astrovirus serotype 8 strain. We found that proteasome inhibitors decrease the production of infectious viral progeny at a step in the replication cycle subsequent to virus entry. The inhibition of proteasome activity decreases viral RNA levels and viral protein synthesis; similarly, the inhibition of ubiquitination by chemical inhibitors or RNA interference (RNAi) reduces the production of viral progeny as well as viral protein synthesis. The effect on viral progeny production induced by proteasome inhibitors is not explained by a reduction in the pool of monoubiquitin or the induction of early apoptosis or autophagy. Our observations are consistent with the need of the proteolytic activity of the UPS for the efficient replication of the virus and suggest that UPS is necessary for the production of genomic and subgenomic RNA but not for antigenomic RNA.IMPORTANCE Astroviruses are a major cause of gastroenteritis in young humans and animals, and recently, it was associated with fatal encephalitis in humans. The role of the ubiquitin-proteasome system in the replication of these viruses has not been studied previously. In this work, we present evidence that supports that the proteolytic activity of the proteasome is necessary for efficient viral progeny production and that this proteolytic system is required for the accumulation of both genomic and subgenomic viral RNAs.
Collapse
|
10
|
Tsai CY, Chen CY, Chiou YH, Shyu HW, Lin KH, Chou MC, Huang MH, Wang YF. Epigallocatechin-3-Gallate Suppresses Human Herpesvirus 8 Replication and Induces ROS Leading to Apoptosis and Autophagy in Primary Effusion Lymphoma Cells. Int J Mol Sci 2017; 19:ijms19010016. [PMID: 29267216 PMCID: PMC5795967 DOI: 10.3390/ijms19010016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 12/02/2022] Open
Abstract
Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, has been shown to induce cell death in cancer cells. Primary effusion lymphoma (PEL) is an aggressive neoplasm caused by human herpesvirus 8 (HHV8). In this study, we examined the role of EGCG on PEL cells in cell death and HHV8 replication. We performed trypan blue exclusion assay to assess the cell viability of PEL cells, flow cytometry analysis to examine the cell cycle distribution and reactive oxygen species (ROS) generation, caspase-3 activity to assay apoptosis, acridine orange staining to determine autophagy, and immunoblotting to detect the protein levels involved in apoptosis and autophagy as well as mitogen activated protein kinases (MAPKs) activation upon EGCG treatment. The expression of the HHV8 lytic gene was determined by luciferase reporter assay and reverse transcription-PCR, and viral progeny production was determined by PCR. Results revealed that EGCG induced cell death and ROS generation in PEL cells in a dose-dependent manner. N-acetylcysteine (NAC) inhibited the EGCG-induced ROS and rescued the cell from EGCG-induced cell death. Even though EGCG induced ROS generation in PEL cells, it reduced the production of progeny virus from PEL cells without causing HHV8 reactivation. These results suggest that EGCG may represent a novel strategy for the treatment of HHV8 infection and HHV8-associated lymphomas.
Collapse
Affiliation(s)
- Ching-Yi Tsai
- Department of Medical Laboratory Science and Biotechnology, Fooyin-University, Kaohsiung 83102, Taiwan.
| | - Chang-Yu Chen
- Department of Medical Laboratory Science and Biotechnology, Fooyin-University, Kaohsiung 83102, Taiwan.
| | - Yee-Hsuan Chiou
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung 83102, Taiwan.
| | - Huey-Wen Shyu
- Department of Medical Laboratory Science and Biotechnology, Fooyin-University, Kaohsiung 83102, Taiwan.
| | - Kuan-Hua Lin
- Department of Medical Laboratory Science and Biotechnology, Fooyin-University, Kaohsiung 83102, Taiwan.
| | - Miao-Chen Chou
- Department of Medical Laboratory Science and Biotechnology, Fooyin-University, Kaohsiung 83102, Taiwan.
| | - Mei-Han Huang
- Department of Medical Laboratory Science and Biotechnology, Fooyin-University, Kaohsiung 83102, Taiwan.
| | - Yi-Fen Wang
- Department of Medical Laboratory Science and Biotechnology, Fooyin-University, Kaohsiung 83102, Taiwan.
| |
Collapse
|
11
|
Bulman Page PC, Goodyear RL, Horton AE, Chan Y, Karim R, O’Connell MA, Hamilton C, Slawin AMZ, Buckley BR, Allin SM. Formal Total Synthesis of (+)-C9-Deoxyomuralide from l-Leucine Using a Double Sacrificial Chirality Transfer Approach. J Org Chem 2017; 82:12209-12223. [DOI: 10.1021/acs.joc.7b02078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | - Benjamin R. Buckley
- Department
of Chemistry, Loughborough University, Loughborough, Leicestershire LE11 3TU, United Kingdom
| | - Steven M. Allin
- School of Science & Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| |
Collapse
|
12
|
Nakata S, Watanabe T, Nakagawa K, Takeda H, Ito A, Fujimuro M. The dynamics of histone H2A ubiquitination in HeLa cells exposed to rapamycin, ethanol, hydroxyurea, ER stress, heat shock and DNA damage. Biochem Biophys Res Commun 2016; 472:46-52. [DOI: 10.1016/j.bbrc.2016.02.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/15/2016] [Indexed: 11/26/2022]
|
13
|
Shigemi Z, Furukawa Y, Hosokawa K, Minami S, Matsuhiro J, Nakata S, Watanabe T, Kagawa H, Nakagawa K, Takeda H, Fujimuro M. Diallyl trisulfide induces apoptosis by suppressing NF-κB signaling through destabilization of TRAF6 in primary effusion lymphoma. Int J Oncol 2015; 48:293-304. [PMID: 26647777 DOI: 10.3892/ijo.2015.3247] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/02/2015] [Indexed: 11/06/2022] Open
Abstract
The allyl sulfides, including diallyl sulfide (DAS), diallyl disulfide (DAD), and diallyl trisulfide (DAT), contained in garlic and members of the Allium family, have a variety of pharmacological activities. Therefore, allyl sulfides have been evaluated as potential novel chemotherapeutic agents. Here, we found that DAT inhibited nuclear factor-κB (NF-κB) signaling and induced apoptosis in primary effusion lymphoma (PEL), a subtype of non-Hodgkin's B-cell lymphoma caused by Kaposi's sarcoma-associated herpesvirus (KSHV). We examined the cytotoxic effects of DAS, DAD and DAT on PEL cells. DAT significantly reduced the viability of PEL cells compared with uninfected B-lymphoma cells, and induced the apoptosis of PEL cells by activating caspase-9. DAT induced stabilization of IκBα, and suppressed NF-κB transcriptional activity in PEL cells. We examined the mechanism underlying DAT-mediated IκBα stabilization. The results indicated that DAT stabilized IκBα by inhibiting the phosphorylation of IκBα by the IκB kinase (IKK) complex. Furthermore, DAT induced proteasomal degradation of TRAF6, and DAT suppressed IKKβ-phosphorylation through downregulation of TRAF6. It is known that activation of NF-κB is essential for survival of PEL cells. In fact, the NF-κB inhibitor BAY11-7082 induced apoptosis in PEL cells. In addition, DAT suppressed the production of progeny virus from PEL cells. The administration of DAT suppressed the development of PEL cells and ascites in SCID mice xenografted with PEL cells. These findings provide evidence that DAT has antitumor activity against PEL cells in vitro and in vivo, suggesting it to be a novel therapeutic agent for the treatment of PEL.
Collapse
Affiliation(s)
- Zenpei Shigemi
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Yoshiki Furukawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Kohei Hosokawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Setsuya Minami
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Jumpei Matsuhiro
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Shiori Nakata
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Tadashi Watanabe
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Hiroki Kagawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| | - Koji Nakagawa
- Department of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kitaku, Sapporo 060-0812, Japan
| | - Hiroshi Takeda
- Department of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kitaku, Sapporo 060-0812, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Yamashinaku, Kyoto 607-8412, Japan
| |
Collapse
|
14
|
Resveratrol induces cell death and inhibits human herpesvirus 8 replication in primary effusion lymphoma cells. Chem Biol Interact 2015; 242:372-9. [PMID: 26549478 DOI: 10.1016/j.cbi.2015.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 09/18/2015] [Accepted: 10/31/2015] [Indexed: 11/23/2022]
Abstract
Resveratrol (3,4',5-trihydroxy-trans-stilbene) has been reported to inhibit proliferation of various cancer cells. However, the effects of resveratrol on the human herpesvirus 8 (HHV8) harboring primary effusion lymphoma (PEL) cells remains unclear. The anti-proliferation effects and possible mechanisms of resveratrol in the HHV8 harboring PEL cells were examined in this study. Results showed that resveratrol induced caspase-3 activation and the formation of acidic vacuoles in the HHV8 harboring PEL cells, indicating resveratrol treatment could cause apoptosis and autophagy in PEL cells. In addition, resveratrol treatment increased ROS generation but did not lead to HHV8 reactivation. ROS scavenger (N-acetyl cysteine, NAC) could attenuate both the resveratrol induced caspase-3 activity and the formation of acidic vacuoles, but failed to attenuate resveratrol induced PEL cell death. Caspase inhibitor, autophagy inhibitors and necroptosis inhibitor could not block resveratrol induced PEL cell death. Moreover, resveratrol disrupted HHV8 latent infection, inhibited HHV8 lytic gene expression and decreased virus progeny production. Overexpression of HHV8-encoded viral FLICE inhibitory protein (vFLIP) could partially block resveratrol induced cell death in PEL cells. These data suggest that resveratrol-induced cell death in PEL cells may be mediated by disruption of HHV8 replication. Resveratrol may be a potential anti-HHV8 drug and an effective treatment for HHV8-related tumors.
Collapse
|
15
|
Pyrrolidinium fullerene induces apoptosis by activation of procaspase-9 via suppression of Akt in primary effusion lymphoma. Biochem Biophys Res Commun 2014; 451:93-100. [DOI: 10.1016/j.bbrc.2014.07.068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/15/2014] [Indexed: 11/23/2022]
|
16
|
Primary effusion lymphoma in an elderly patient effectively treated by lenalidomide: case report and review of literature. Blood Cancer J 2014; 4:e190. [PMID: 24608734 PMCID: PMC3972705 DOI: 10.1038/bcj.2014.6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 12/27/2022] Open
Abstract
Primary effusion lymphoma (PEL) is a rare aggressive subset of non-Hodgkin B-cell lymphoma. It is caused by Kaposi sarcoma-associated herpesvirus/human herpesvirus type 8 (KSHV/HHV8). It occurs mainly, but not exclusively, in HIV-positive patients. PEL predominantly develops in serous cavities and occasionally in extracavitary regions. PEL carries a very poor prognosis with a median survival time of <6 months. Indeed, currently used treatment modalities such as CHOP chemotherapy are far from achieving complete and sustainable remission. Therefore, there is no clear standard of care established in the treatment of PEL patients, stressing the need for novel-targeted approaches. Here, we have attempted a comprehensive assessment of the treatment of PEL, discussed avant-garde therapies and updated the state of preclinical research with promising clinical applications in the field. These include inhibitors of viral replication, modulators of cell signaling and inflammation, nuclear factor kappa B (NF-κB) and histone deacetylase inhibitors, and recently the combination of arsenic trioxide and interferon-alpha. Some of these targeted therapies have not yet reached clinical studies, although others were used in a few individual case reports with low numbers of patients. We also describe the first case of a 77-year-old, HIV-negative, HHV8-positive patient diagnosed with PEL limited to the pleural and peritoneal cavities. He received lenalidomide 25 mg/day for 21 days every 28 days. Treatment was well tolerated with no side effects. He rapidly improved after 1 month of treatment and progressively achieved complete remission persistent after 18 months of therapy. We believe that this review will bridge an important gap between classical chemotherapy and modern approaches of targeted therapy. Finally, our findings warrant further evaluation of lenalidomide in future prospective clinical studies.
Collapse
|
17
|
Wakao K, Watanabe T, Takadama T, Ui S, Shigemi Z, Kagawa H, Higashi C, Ohga R, Taira T, Fujimuro M. Sangivamycin induces apoptosis by suppressing Erk signaling in primary effusion lymphoma cells. Biochem Biophys Res Commun 2014; 444:135-40. [PMID: 24434142 DOI: 10.1016/j.bbrc.2014.01.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/08/2014] [Indexed: 01/24/2023]
Abstract
Sangivamycin, a structural analog of adenosine and antibiotic exhibiting antitumor and antivirus activities, inhibits protein kinase C and the synthesis of both DNA and RNA. Primary effusion lymphoma (PEL) is an aggressive neoplasm caused by Kaposi's sarcoma-associated herpesvirus (KSHV) in immunosuppressed patients and HIV-infected homosexual males. PEL cells are derived from post-germinal center B cells, and are infected with KSHV. Herein, we asked if sangivamycin might be useful to treat PEL. We found that sangivamycin killed PEL cells, and we explored the underlying mechanism. Sangivamycin treatment drastically decreased the viability of PEL cell lines compared to KSHV-uninfected B lymphoma cell lines. Sangivamycin induced the apoptosis of PEL cells by activating caspase-7 and -9. Further, sangivamycin suppressed the phosphorylation of Erk1/2 and Akt, thus inhibiting activation of the proteins. Inhibitors of Akt and MEK suppressed the proliferation of PEL cells compared to KSHV-uninfected cells. It is known that activation of Erk and Akt signaling inhibits apoptosis and promotes proliferation in PEL cells. Our data therefore suggest that sangivamycin induces apoptosis by inhibiting Erk and Akt signaling in such cells. We next investigated whether sangivamycin, in combination with an HSP90 inhibitor geldanamycin (GA) or valproate (valproic acid), potentiated the cytotoxic effects of the latter drugs on PEL cells. Compared to treatment with GA or valproate alone, the addition of sangivamycin enhanced cytotoxic activity. Our data thus indicate that sangivamycin may find clinical utility as a novel anti-cancer agent targeting PEL.
Collapse
Affiliation(s)
- Kazufumi Wakao
- Department of Biotechnology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Kofu-shi 400-8511, Japan
| | - Tadashi Watanabe
- Department of Cell Biology, Kyoto Pharmaceutical University, Misasagi-Shichonocho 1, Yamashinaku, Kyoto 607-8412, Japan
| | - Tadatoshi Takadama
- Department of Biotechnology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Kofu-shi 400-8511, Japan
| | - Sadaharu Ui
- Department of Biotechnology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Kofu-shi 400-8511, Japan
| | - Zenpei Shigemi
- Department of Cell Biology, Kyoto Pharmaceutical University, Misasagi-Shichonocho 1, Yamashinaku, Kyoto 607-8412, Japan
| | - Hiroki Kagawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Misasagi-Shichonocho 1, Yamashinaku, Kyoto 607-8412, Japan
| | - Chizuka Higashi
- Department of Molecular Cell Biology, Faculty of Medicine, University of Yamanashi, Chuoh-shi 409-3898, Japan
| | - Rie Ohga
- Department of Molecular Cell Biology, Faculty of Medicine, University of Yamanashi, Chuoh-shi 409-3898, Japan
| | - Takahiro Taira
- Department of Molecular Cell Biology, Faculty of Medicine, University of Yamanashi, Chuoh-shi 409-3898, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Misasagi-Shichonocho 1, Yamashinaku, Kyoto 607-8412, Japan.
| |
Collapse
|
18
|
Jiang D, Wang X, Liu X, Li F. Gene delivery of cyclin-dependent kinase inhibitors p21Waf1 and p27Kip1 suppresses proliferation of MCF-7 breast cancer cells in vitro. Breast Cancer 2013; 21:614-23. [PMID: 23338153 DOI: 10.1007/s12282-012-0438-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 12/21/2012] [Indexed: 11/24/2022]
Abstract
BACKGROUND Because tumorigenesis depends on a variety of oncogenes, symphyseal study of combined genes may lead to more significant knowledge about tumorigenesis and progression. Combined deficiency of p21 and p27 proteins in mice is linked to more aggressive spontaneous tumorigenesis. We investigated the effect of the transfected p21 (Waf1) -p27 (Kip1) gene on centrosome duplication, cell proliferation, and apoptosis of MCF-7, a breast cancer cell line. METHODS The pIRES-p21 (Waf1) , pIRES-p27 (Kip1) , and pIRES-p21 (Waf1) -p27 (Kip1) genes were transfected into MCF-7 cells by lipofection. The effect on proliferation was evaluated by MTT assay and clone-formation assay. Cell cycle and apoptosis were analyzed by flow cytometry. Apoptosis was tested by flow cytometry and TUNEL assay. Centrosome duplication was detected by use of indirect immunofluorescence microscopy. RESULTS The results showed that the pIRES-p21 (Waf1) , pIRES-p27 (Kip1) , and pIRES-p21 (Waf1) -p27 (Kip1) significantly inhibited proliferation of MCF-7 cells, followed by accumulation of MCF-7 cells in cycle G1, induced apoptosis, and a decrease in the proportion of MCF-7 cells which contained abnormal centrosomes. Compared with p21 (Waf1) or p27 (Kip1) alone, combination of p21 (Waf1) and p27 (Kip1) had a much more significant effect (P < 0.05). CONCLUSION Altogether, these results indicate that the p21 (Waf1) -p27 (Kip1) gene combination has a more obvious antitumor effect than p21 (Waf1) or p27 (Kip1) alone. This study provides preclinical evidence that combination of p21 (Waf1) and p27 (Kip1) could be a novel and promising therapeutic approach to treatment of breast cancer with suppressed p21 (Waf1) and p27 (Kip1) expression.
Collapse
Affiliation(s)
- Dandan Jiang
- Breast Surgery, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, 266003, Shandong, China
| | | | | | | |
Collapse
|
19
|
Gao C, Chen G, Liu L, Li X, He J, Jiang L, Zhu J, Xu Y. Impact of high glucose and proteasome inhibitor MG132 on histone H2A and H2B ubiquitination in rat glomerular mesangial cells. J Diabetes Res 2013; 2013:589474. [PMID: 23738337 PMCID: PMC3657404 DOI: 10.1155/2013/589474] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 01/12/2013] [Accepted: 02/17/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Hyperglycemia plays a pivotal role in the development of diabetic nephropathy (DN) and may be related to epigenetic metabolic memory. One of the most crucial epigenetic mechanisms is histone modification, which is associated with the expression of a fibrosis factor in vascular injury. Aim .In this study, we investigated the ubiquitination of histones H2A and H2B to explore the epigenetic mechanisms of DN. MATERIALS AND METHODS The GMCs were cultured as follows: normal group, high glucose group, mannitol group, and intervention group. After 12 hr, 24 hr, and 48 hr, histones ubiquitination, transforming growth factor-β (TGF-β), and fibronectin (FN) were measured using WB, RT-PCR, and IF. RESULT High glucose can induce the upregulation of FN. H2A ubiquitination in GMCs increased in high glucose group (P < 0.01), whereas it decreased significantly in intervention group (P < 0.05). In contrast, H2B ubiquitination decreased with an increasing concentration of glucose, but it was recovered in the intervention group (P < 0.05). Expression of TGF-β changed in response to abnormal histone ubiquitination. CONCLUSIONS The high glucose may induce H2A ubiquitination and reduce H2B ubiquitination in GMCs. The changes of histone ubiquitination may be due in part to DN by activating TGF-β signaling pathway.
Collapse
Affiliation(s)
- Chenlin Gao
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
| | - Guo Chen
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
| | - Li Liu
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
| | - Xia Li
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
| | - Jianhua He
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
| | - Lan Jiang
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
| | - Jianhua Zhu
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
| | - Yong Xu
- Department of Endocrinology, Affiliated Hospital of Luzhou Medical College, Luzhou 646000, China
- *Yong Xu:
| |
Collapse
|
20
|
Kaposi's sarcoma-associated herpesvirus suppression of DUSP1 facilitates cellular pathogenesis following de novo infection. J Virol 2012; 87:621-35. [PMID: 23097457 DOI: 10.1128/jvi.01441-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), and KSHV activation of mitogen-activated protein kinases (MAPKs) initiates a number of key pathogenic determinants of KS. Direct inhibition of signal transduction as a therapeutic approach presents several challenges, and a better understanding of KSHV-induced mechanisms regulating MAPK activation may facilitate the development of new treatment or prevention strategies for KS. MAPK phosphatases, including dual-specificity phosphatase-1 (DUSP1), negatively regulate signal transduction and cytokine activation through MAPK dephosphorylation or interference with effector molecule binding to MAPKs, including the extracellular signal-regulated kinase (ERK). We found that ERK-dependent latent viral gene expression, the induction of promigratory factors, and cell invasiveness following de novo infection of primary human endothelial cells are in part dependent on KSHV suppression of DUSP1 expression during de novo infection. KSHV-encoded miR-K12-11 upregulates the expression of xCT (an amino acid transporter and KSHV fusion/entry receptor), and existing data indicate a role for xCT in the regulation of 14-3-3β, a transcriptional repressor of DUSP1. We found that miR-K12-11 induces endothelial cell secretion of promigratory factors and cell invasiveness through upregulation of xCT-dependent, 14-3-3β-mediated suppression of DUSP1. Finally, proof-of-principle experiments revealed that pharmacologic upregulation of DUSP1 inhibits the induction of promigratory factors and cell invasiveness during de novo KSHV infection. These data reveal an indirect role for miR-K12-11 in the regulation of DUSP1 and downstream pathogenesis.
Collapse
|