1
|
Irving AT, Rozario P, Kong PS, Luko K, Gorman JJ, Hastie ML, Chia WN, Mani S, Lee BPH, Smith GJD, Mendenhall IH, Larman HB, Elledge SJ, Wang LF. Robust dengue virus infection in bat cells and limited innate immune responses coupled with positive serology from bats in IndoMalaya and Australasia. Cell Mol Life Sci 2020; 77:1607-1622. [PMID: 31352533 PMCID: PMC11104837 DOI: 10.1007/s00018-019-03242-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/10/2019] [Accepted: 07/22/2019] [Indexed: 01/19/2023]
Abstract
Natural reservoir hosts can sustain infection of pathogens without succumbing to overt disease. Multiple bat species host a plethora of viruses, pathogenic to other mammals, without clinical symptoms. Here, we detail infection of bat primary cells, immune cells, and cell lines with Dengue virus. While antibodies and viral RNA were previously detected in wild bats, their ability to sustain infection is not conclusive. Old-world fruitbat cells can be infected, producing high titres of virus with limited cellular responses. In addition, there is minimal interferon (IFN) response in cells infected with MOIs leading to dengue production. The ability to support in vitro replication/production raises the possibility of bats as a transient host in the life cycle of dengue or similar flaviviruses. New antibody serology evidence from Asia/Pacific highlights the previous exposure and raises awareness that bats may be involved in flavivirus dynamics and infection of other hosts.
Collapse
Affiliation(s)
| | | | | | | | - Jeffrey J Gorman
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Marcus L Hastie
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Wan Ni Chia
- Duke-NUS Medical School, Singapore, Singapore
| | | | | | | | | | | | - Stephen J Elledge
- Harvard University Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Lin-Fa Wang
- Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
2
|
Wu X, Jia R, Wang M, Chen S, Liu M, Zhu D, Zhao X, Yang Q, Wu Y, Yin Z, Zhang S, Huang J, Zhang L, Liu Y, Yu Y, Pan L, Tian B, Rehman MU, Chen X, Cheng A. Downregulation of microRNA-30a-5p contributes to the replication of duck enteritis virus by regulating Beclin-1-mediated autophagy. Virol J 2019; 16:144. [PMID: 31771604 PMCID: PMC6880601 DOI: 10.1186/s12985-019-1250-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) is increasingly recognized as an important element in regulating virus-host interactions. Our previous results showed that cellular miR-30a-5p was significantly downregulated after duck enteritis virus (DEV) infection cell. However, whehter or not the miR-30a-5p is involved in DEV infection has not been known. METHODS Quantitative reverse-transcription PCR (qRT-PCR) was used to measure the expression levels of miRNAs(miR-30a-5p) and Beclin-1 mRNA. The miR-30a-5p - Beclin-1 target interactions were determined by Dual luciferase reporter assay (DLRA). Western blotting was utilized to analyze Beclin-1-mediated duck embryo fibroblast (DEF) cells autophagy activity. DEV titers were estimated by the median tissue culture infective dose (TCID50). RESULTS The miR-30a-5p was significantly downregulated and the Beclin-1 mRNA was significantly upregulated in DEV-infected DEF cells. DLRA confirmed that miR-30a-5p directly targeted the 3'- UTR of the Beclin-1 gene. Overexpression of miR-30a-5p significantly reduced the expression level of Beclin-1protein (p < 0.05), leading to the decrease of Beclin-1-mediated autophagy activity, which ultimately suppressed DEV replication (P < 0.05). Whereas transfection of miR-30a-5p inhibitor increased Beclin-1-mediated autophagy and triggered DEV replication during the whole process of DEV infection (P < 0.01). CONCLUSIONS This study shows that miR-30a-5p can inhibit DEV replication through reducing autophagy by targeting Beclin-1. These findings suggest a new insight into virus-host interaction during DEV infection and provide a potential new antiviral therapeutic strategy against DEV infection.
Collapse
Affiliation(s)
- Xianglong Wu
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China.
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Ling Zhang
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Yunya Liu
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Yanling Yu
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Leichang Pan
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Bin Tian
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Mujeeb Ur Rehman
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Xiaoyue Chen
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu, 611130, Sichuan Province, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu, 611130, Sichuan Province, China.
| |
Collapse
|
3
|
Tao XL, Zhao W, Tong W, Wang XF, Dou LL, Chen JM, Liu N, Lu Y, Zhang YB, Jin XP, Shen YF, Zhao HY, Jin H, Li YG. The effects of autophagy on the replication of Nelson Bay orthoreovirus. Virol J 2019; 16:90. [PMID: 31319897 PMCID: PMC6639940 DOI: 10.1186/s12985-019-1196-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 06/26/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nelson Bay orthoreovirus (NBV) was first isolated over 40 years ago from a fruit bat in Australia. Normally, NBV does not cause human diseases, but recently several NBV strains have been associated with human respiratory tract infections, thus attracting clinical attention. Autophagy, an evolutionarily conserved process in eukaryotic cells, degrades intracellular substrates, participates in multiple physiological processes, and maintains cellular homeostasis. In addition, autophagy is intimately involved in viral infection. METHODS A new strain of NBV, isolated from a patient with a respiratory tract infection who returned to Japan from Bali, Indonesia, in 2007, was used in this study. NBV was rescued using a reverse genetics system involving cotransfection of BHK cells with 11 plasmids (pT7-L1 MB, pT7-L2 MB, pT7-L3 MB, pT7-M1 MB, pT7-M2 MB, pT7-M3 MB, pT7-S1 MB, pT7-S2 MB, pT7-S3 MB, pT7-S4 MB, and pcDNA3.1-T7), yielding NBV-MB. Recovered viruses were confirmed by immunofluorescence. The effect of NBV-MB on autophagy was evaluated by measuring the LC3-I/II proteins by immunoblot analysis after infection of BHK cells. Furthermore, after treatment with rapamycin (RAPA), 3-methyladenine (3-MA), chloroquine (CQ), or plasmid (GFP-LC3) transfection, the changes in expression of the LC3 gene and the amount of LC3-I/II protein were examined. In addition, variations in viral titer were assayed after treatment of BHK cells with drugs or after transfection with plasmids pCAGM3 and pCAGS3, which encode virus nonstructural proteins μNS and σNS, respectively. RESULTS NBV-MB infection induced autophagy in host cells; however, the level of induction was dependent on viral replication. Induction of autophagy increased viral replication. By contrast, inhibiting autophagy suppressed NBV replication, albeit not significantly. The NBV-MB nonstructural protein μNS was involved in the induction of autophagy with viral infection. CONCLUSIONS NBV-MB infection triggered autophagy. Also, the NBV nonstructural protein μNS may contribute to augmentation of autophagy upon viral infection.
Collapse
Affiliation(s)
- Xiao-Li Tao
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang City, 110013, Liaoning Province, People's Republic of China.,Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Wei Zhao
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Wei Tong
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Xiao-Fang Wang
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Li-Li Dou
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Jiang-Man Chen
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Nian Liu
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Ying Lu
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Yi-Bo Zhang
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Xu-Peng Jin
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Yan-Fei Shen
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Hong-Yan Zhao
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China
| | - Hong Jin
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang City, 110013, Liaoning Province, People's Republic of China.
| | - Yong-Gang Li
- Department of Pathogenic Microbiology, College of Basic Medical Sciences, Jinzhou Medical University, No. 40, the Third Section of SongPo Rd, Jinzhou City, 121200, Liaoning Province, China.
| |
Collapse
|
4
|
Homologs of Human Dengue-Resistance Genes, FKBP1B and ATCAY, Confer Antiviral Resistance in Aedes aegypti Mosquitoes. INSECTS 2019; 10:insects10020046. [PMID: 30717390 PMCID: PMC6409984 DOI: 10.3390/insects10020046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 02/06/2023]
Abstract
Dengue virus (DENV) is transmitted by mosquitoes and is a major public health concern. The study of innate mosquito defense mechanisms against DENV have revealed crucial roles for the Toll, Imd, JAK-STAT, and RNAi pathways in mediating DENV in the mosquito. Often overlooked in such studies is the role of intrinsic cellular defense mechanisms that we hypothesize to work in concert with the classical immune pathways to affect organismal defense. Our understanding of the molecular interaction of DENV with mosquito host cells is limited, and we propose to expand upon the recent results from a genome-scale, small interfering RNA (siRNA)-based study that identified mammalian host proteins associated with resistance to dengue/West Nile virus (DENV/WNV) infection. The study identified 22 human DENV/WNV resistance genes (DVR), and we hypothesized that a subset would be functionally conserved in Aedes aegypti mosquitoes, imparting cellular defense against flaviviruses in this species. We identified 12 homologs of 22 human DVR genes in the Ae. aegypti genome. To evaluate their possible role in cellular resistance/antiviral defense against DENV, we used siRNA silencing targeted against each of the 12 homologs in an Ae. aegypti cell line (Aag2) infected with DENV2 and identified that silencing of the two candidates, AeFKBP1 and AeATCAY, homologs of human FKBP1B and ATCAY, were associated with a viral increase. We then used dsRNA to silence each of the two genes in adult mosquitoes to validate the observed antiviral functions in vivo. Depletion of AeFKBP1 or AeATCAY increased viral dissemination through the mosquito at 14 days post-infection. Our results demonstrated that AeFKBP1 and AeATCAY mediate resistance to DENV akin to what has been described for their homologs in humans. AeFKBP1 and AeATCAY provide a rare opportunity to elucidate a DENV-resistance mechanism that may be evolutionarily conserved between humans and Ae. aegypti.
Collapse
|
5
|
Zhou K, Yao P, He J, Zhao H. Lipophagy in nonliver tissues and some related diseases: Pathogenic and therapeutic implications. J Cell Physiol 2018; 234:7938-7947. [PMID: 30537019 DOI: 10.1002/jcp.27988] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022]
Abstract
Lipid autophagy (lipophagy) is defined as a selective autophagy process in which some intracellular lipid droplets are selectively degraded by autophagic lysosomes pathway. The occurrence of lipophagy was first discovered in liver tissues. Additionally, abundant evidence indicated that the occurrence of hepatic lipophagy has been implicated in many liver diseases including fatty liver diseases, nonalcoholic fatty liver diseases, liver fibrosis, and liver cirrhosis. However, recent studies suggested that hepatic lipophagy occurs not only in liver tissue but also in other nonliver tissues and cells. Furthermore, the occurrence of lipophagy plays a crucial role in nonliver tissues and some related diseases. For instance, lipophagy relieves insulin resistance in adipose tissue from obesity patient with type 2 diabetes. Additionally, lipophagy has the ability to remit neurodegenerative diseases by reducing activity-dependent neurodegeneration in nervous tissue. Lipophagy decreases muscle lipid accumulation and accordingly improves lipid storage myopathy in muscle tissue. Moreover, lipophagy alleviates the malignancy and metastasis of cancer in clear renal cell carcinoma tissue. Lipophagy is also involved in other processes, such as spermatogenesis, osteoblastogenesis, and mucosal ulceration. In conclusion, targeting lipophagy may be a critical regulator and a new therapeutic strategy for nonliver tissues and some related diseases.
Collapse
Affiliation(s)
- Kebing Zhou
- Department of Emergency Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Pingbo Yao
- Department of Emergency Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Jun He
- Department of Emergency Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Hong Zhao
- Department of Basic nursing, Nursing College, University of South China, Hengyang, China
| |
Collapse
|
6
|
Investigating Tick-borne Flaviviral-like Particles as a Delivery System for Gene Therapy. Curr Ther Res Clin Exp 2017; 88:8-17. [PMID: 30093925 PMCID: PMC6076373 DOI: 10.1016/j.curtheres.2017.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2017] [Indexed: 12/30/2022] Open
Abstract
Background Research on the biogenesis of tick-borne encephalitis virus (TBEV) would benefit gene therapy. Due to specific arrangements of genes along the TBEV genome, its viral-like particles (VLPs) could be exploited as shuttles to deliver their replicon, which carries therapeutic genes, to immune system cells. Objective To develop a flaviviral vector for gene delivery as a part of gene therapy research that can be expressed in secretable VLP suicidal shuttles and provide abundant unique molecular and structural data supporting this gene therapy concept. Method TBEV structural gene constructs of a Swedish Torö strain were cloned into plasmids driven by the promoters CAG and CMV and then transfected into various cell lines, including COS-1 and BHK-21. Time-course sampling of the cells, culture fluid, cell lysate supernatant, and pellet specimens were performed. Western blotting and electron microscopy analyses of collected specimens were used to investigate molecular and structural processing of TBEV structural proteins. Results Western blotting analysis showed differences between promoters in directing the gene expression of the VLPs constructs. The premature flaviviral polypeptides as well as mature VLPs could be traced. Using electron microscopy, the premature and mature VLP accumulation in cellular compartments—and also endoplasmic reticulum proliferation as a virus factory platform—were observed in addition to secreted VLPs. Conclusions The abundant virologic and cellular findings in this study show the natural processing and safety of inserting flaviviral structural genes into suicidal VLP shuttles. Thus, we propose that these VLPs are a suitable gene delivering system model in gene therapy.
Collapse
|
7
|
Ho MR, Tsai TT, Chen CL, Jhan MK, Tsai CC, Lee YC, Chen CH, Lin CF. Blockade of dengue virus infection and viral cytotoxicity in neuronal cells in vitro and in vivo by targeting endocytic pathways. Sci Rep 2017; 7:6910. [PMID: 28761128 PMCID: PMC5537343 DOI: 10.1038/s41598-017-07023-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023] Open
Abstract
Dengue virus (DENV) infection in neuronal cells was speculated to trigger neuropathy. Herein, we determined the blockade of DENV infection by targeting endocytic pathways in vitro and in vivo. In DENV-infected mouse brains, we previously showed that viral proteins are expressed in neuronal cells around the hippocampus with accompanying neurotoxicity. DENV caused infection, including entry, double-stranded (ds)RNA replication, protein expression, and virus release, followed by cytotoxicity in the mouse neuronal Neuro-2a cell line. Pharmacologically blocking clathrin-mediated endocytosis of the DENV retarded viral replication. Targeting vacuolar-type H+-ATPase (V-ATPase)-based endosomal acidification effectively blocked the DENV replication process, but had no direct effect on viral translation. Blockade of the clathrin- and V-ATPase-based endocytic pathways also attenuated DENV-induced neurotoxicity. Inhibiting endosomal acidification effectively retarded DENV infection, acute viral encephalitis, and mortality. These results demonstrate that clathrin mediated endocytosis of DENV followed by endosomal acidification-dependent viral replication in neuronal cells, which can lead to neurotoxicity.
Collapse
Affiliation(s)
- Min-Ru Ho
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tsung-Ting Tsai
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chia-Ling Chen
- Translational Research Center, Taipei Medical University, Taipei, 110, Taiwan
| | - Ming-Kai Jhan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Cheng-Chieh Tsai
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
| | - Yi-Chao Lee
- The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
| | - Chun-Han Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
8
|
Welte MA, Gould AP. Lipid droplet functions beyond energy storage. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1260-1272. [PMID: 28735096 PMCID: PMC5595650 DOI: 10.1016/j.bbalip.2017.07.006] [Citation(s) in RCA: 353] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Lipid droplets are cytoplasmic organelles that store neutral lipids and are critically important for energy metabolism. Their function in energy storage is firmly established and increasingly well characterized. However, emerging evidence indicates that lipid droplets also play important and diverse roles in the cellular handling of lipids and proteins that may not be directly related to energy homeostasis. Lipid handling roles of droplets include the storage of hydrophobic vitamin and signaling precursors, and the management of endoplasmic reticulum and oxidative stress. Roles of lipid droplets in protein handling encompass functions in the maturation, storage, and turnover of cellular and viral polypeptides. Other potential roles of lipid droplets may be connected with their intracellular motility and, in some cases, their nuclear localization. This diversity highlights that lipid droplets are very adaptable organelles, performing different functions in different biological contexts. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY, United States.
| | | |
Collapse
|
9
|
Targeting heat shock factor 1 as an antiviral strategy against dengue virus replication in vitro and in vivo. Antiviral Res 2017; 145:44-53. [PMID: 28733114 DOI: 10.1016/j.antiviral.2017.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 11/22/2022]
Abstract
Fever onset is correlated with viremia in dengue virus (DENV) patients. Heat shock factor 1 (HSF1), a heat stress response host transcription factor, plays a crucial role in regulating multiple cellular functions, as well as the onset of infectious diseases. This study evaluated the role of HSF1 in DENV replication as a means of regulating DENV infection in vitro and in vivo. DENV infection activated HSF1 in both Ca2+ and protein kinase A-dependent manners. Inhibiting HSF1 effectively reduced DENV replication, not only in THP-1 cells but also in primary human monocytes. Activated HSF1 contributed to DENV replication by upregulating autophagy-related protein (Atg) 7, as autophagy is crucial for virus replication. Heat stress also activated HSF1, which in turn facilitated DENV replication. Activated HSF1, the increased Atg7, and autophagic induction were founded in the DENV-infected brains and pharmacologically inhibiting HSF1 reduced autophagy, viral protein expression, neuropathy, and mortality. These results provide new insight into HSF1 as a novel host factor for DENV infection through its role in facilitating autophagy-regulated viral replication in the brains.
Collapse
|
10
|
Yin HC, Zhao LL, Li SQ, Niu YJ, Jiang XJ, Xu LJ, Lu TF, Han LX, Liu SW, Chen HY. Autophagy activated by duck enteritis virus infection positively affects its replication. J Gen Virol 2017; 98:486-495. [PMID: 28008822 DOI: 10.1099/jgv.0.000696] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Duck enteritis virus (DEV) is an acute, septic, sexually transmitted disease that occurs in ducks, geese and other poultry. Autophagy is an evolutionarily ancient pathway that is important in many viral infections. Despite extensive study, the interplay between DEV and autophagy of host cells is not clearly understood. In this study, we found that DEV infection triggers autophagy in duck embryo fibroblast (DEF) cells, as demonstrated by the appearance of autophagosome-like double- or single-membrane vesicles in the cytoplasm of host cells and the number of GFP-LC3 dots. In addition, increased conversion of the autophagy marker protein LC3-I and LC3-II and decreased p62/SQSTM1 indicated complete autophagy flux. Heat-inactivated DEV infection did not induce autophagy, suggesting that the trigger of autophagy in DEF cells depended on DEV replication. When autophagy was pharmacologically inhibited by LY294002 or wortmannin, DEV replication decreased. The DEV offspring yield decreased when small interference RNA was used to interfere with autophagy related to the genes Beclin-1 and ATG5. In contrast, after treating DEF cells with rapamycin, an inducer of autophagy, DEV replication increased. These results indicated that DEV infection induced autophagy in DEF cells and autophagy facilitated DEV replication.
Collapse
Affiliation(s)
- Hai-Chang Yin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agriculture Sciences, 678 Haping Road, Harbin 150069, PR China
| | - Li-Li Zhao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agriculture Sciences, 678 Haping Road, Harbin 150069, PR China
| | - Si-Qi Li
- College of Life Science and Technology, Mudanjiang Normal University, 191 Wenhua Street, Mudanjiang 157011, PR China
| | - Yin-Jie Niu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agriculture Sciences, 678 Haping Road, Harbin 150069, PR China
| | - Xin-Jie Jiang
- College of Life Science and Technology, Mudanjiang Normal University, 191 Wenhua Street, Mudanjiang 157011, PR China
| | - Li-Jing Xu
- College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, PR China
| | - Tao-Feng Lu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agriculture Sciences, 678 Haping Road, Harbin 150069, PR China
| | - Ling-Xia Han
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agriculture Sciences, 678 Haping Road, Harbin 150069, PR China
| | - Sheng-Wang Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agriculture Sciences, 678 Haping Road, Harbin 150069, PR China
| | - Hong-Yan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agriculture Sciences, 678 Haping Road, Harbin 150069, PR China
| |
Collapse
|
11
|
Perera N, Miller JL, Zitzmann N. The role of the unfolded protein response in dengue virus pathogenesis. Cell Microbiol 2017; 19. [PMID: 28207988 DOI: 10.1111/cmi.12734] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/12/2017] [Accepted: 02/14/2017] [Indexed: 12/31/2022]
Abstract
Symptomatic dengue virus (DENV) infections range from mild fever to severe haemorrhagic disease and death. Host-viral interactions play a significant role in deciding the fate of the infection. The unfolded protein response (UPR) is a prosurvival cellular reaction induced in response to DENV-mediated endoplasmic reticulum stress. The UPR has complex interactions with the cellular autophagy machinery, apoptosis, and innate immunity. DENV has evolved to manipulate the UPR to facilitate its replication and to evade host immunity. Our knowledge of this intertwined network of events is continuously developing. A better understanding of the UPR mediated antiviral and proviral effects will shed light on dengue disease pathogenesis and may help development of anti-DENV therapeutics. This review summarizes the role of the UPR in viral replication, autophagy, and DENV-induced inflammation to describe how a host response contributes to DENV pathogenesis.
Collapse
Affiliation(s)
- Nilanka Perera
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK.,Department of Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Joanna L Miller
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| | - Nicole Zitzmann
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Pang EL, Loh HS. Towards development of a universal dengue vaccine – How close are we? ASIAN PAC J TROP MED 2017; 10:220-228. [DOI: 10.1016/j.apjtm.2017.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 01/20/2017] [Accepted: 01/26/2017] [Indexed: 11/16/2022] Open
|
13
|
Severson DW, Behura SK. Genome Investigations of Vector Competence in Aedes aegypti to Inform Novel Arbovirus Disease Control Approaches. INSECTS 2016; 7:insects7040058. [PMID: 27809220 PMCID: PMC5198206 DOI: 10.3390/insects7040058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/24/2016] [Accepted: 10/25/2016] [Indexed: 11/16/2022]
Abstract
Dengue (DENV), yellow fever, chikungunya, and Zika virus transmission to humans by a mosquito host is confounded by both intrinsic and extrinsic variables. Besides virulence factors of the individual arboviruses, likelihood of virus transmission is subject to variability in the genome of the primary mosquito vector, Aedes aegypti. The “vectorial capacity” of A. aegypti varies depending upon its density, biting rate, and survival rate, as well as its intrinsic ability to acquire, host and transmit a given arbovirus. This intrinsic ability is known as “vector competence”. Based on whole transcriptome analysis, several genes and pathways have been predicated to have an association with a susceptible or refractory response in A. aegypti to DENV infection. However, the functional genomics of vector competence of A. aegypti is not well understood, primarily due to lack of integrative approaches in genomic or transcriptomic studies. In this review, we focus on the present status of genomics studies of DENV vector competence in A. aegypti as limited information is available relative to the other arboviruses. We propose future areas of research needed to facilitate the integration of vector and virus genomics and environmental factors to work towards better understanding of vector competence and vectorial capacity in natural conditions.
Collapse
Affiliation(s)
- David W Severson
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Susanta K Behura
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
14
|
Zhang X, Jia R, Zhou J, Wang M, Yin Z, Cheng A. Capsid-Targeted Viral Inactivation: A Novel Tactic for Inhibiting Replication in Viral Infections. Viruses 2016; 8:E258. [PMID: 27657114 PMCID: PMC5035972 DOI: 10.3390/v8090258] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/08/2016] [Accepted: 09/15/2016] [Indexed: 12/18/2022] Open
Abstract
Capsid-targeted viral inactivation (CTVI), a conceptually powerful new antiviral strategy, is attracting increasing attention from researchers. Specifically, this strategy is based on fusion between the capsid protein of a virus and a crucial effector molecule, such as a nuclease (e.g., staphylococcal nuclease, Barrase, RNase HI), lipase, protease, or single-chain antibody (scAb). In general, capsid proteins have a major role in viral integration and assembly, and the effector molecule used in CTVI functions to degrade viral DNA/RNA or interfere with proper folding of viral key proteins, thereby affecting the infectivity of progeny viruses. Interestingly, such a capsid-enzyme fusion protein is incorporated into virions during packaging. CTVI is more efficient compared to other antiviral methods, and this approach is promising for antiviral prophylaxis and therapy. This review summarizes the mechanism and utility of CTVI and provides some successful applications of this strategy, with the ultimate goal of widely implementing CTVI in antiviral research.
Collapse
Affiliation(s)
- Xingcui Zhang
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
| | - Renyong Jia
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
| | - Jiakun Zhou
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
| | - Mingshu Wang
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu 611130, Sichuan Province, China.
| | - Anchun Cheng
- Avian Disease Research Center, College of Veterinary Medicine of Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Wenjiang District, Chengdu 611130, Sichuan Province, China.
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang District, Chengdu 611130, Sichuan Province, China.
| |
Collapse
|
15
|
Valadão ALC, Aguiar RS, de Arruda LB. Interplay between Inflammation and Cellular Stress Triggered by Flaviviridae Viruses. Front Microbiol 2016; 7:1233. [PMID: 27610098 PMCID: PMC4996823 DOI: 10.3389/fmicb.2016.01233] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae family comprises several human pathogens, including Dengue, Zika, Yellow Fever, West Nile, Japanese Encephalitis viruses, and Hepatitis C Virus. Those are enveloped, single-stranded positive sense RNA viruses, which replicate mostly in intracellular compartments associated to endoplasmic reticulum (ER) and Golgi complex. Virus replication results in abundant viral RNAs and proteins, which are recognized by cellular mechanisms evolved to prevent virus infection, resulting in inflammation and stress responses. Virus RNA molecules are sensed by Toll-like receptors (TLRs), RIG-I-like receptors (RIG-I and MDA5) and RNA-dependent protein kinases (PKR), inducing the production of inflammatory mediators and interferons. Simultaneously, the synthesis of virus RNA and proteins are distinguished in different compartments such as mitochondria, ER and cytoplasmic granules, triggering intracellular stress pathways, including oxidative stress, unfolded protein response pathway, and stress granules assembly. Here, we review the new findings that connect the inflammatory pathways to cellular stress sensors and the strategies of Flaviviridae members to counteract these cellular mechanisms and escape immune response.
Collapse
Affiliation(s)
- Ana L C Valadão
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Renato S Aguiar
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Luciana B de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Wang K, Wang J, Sun T, Bian G, Pan W, Feng T, Wang P, Li Y, Dai J. Glycosphingolipid GM3 is Indispensable for Dengue Virus Genome Replication. Int J Biol Sci 2016; 12:872-83. [PMID: 27313500 PMCID: PMC4910605 DOI: 10.7150/ijbs.15641] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/03/2016] [Indexed: 01/08/2023] Open
Abstract
Dengue virus (DENV) causes the most prevalent arthropod-borne viral disease of humans worldwide. Glycosphingolipids (GSLs) are involved in virus infection by regulating various steps of viral-host interaction. However, the distinct role of GSLs during DENV infection remains unclear. In this study, we used mouse melanoma B16 cells and their GSL-deficient mutant counterpart GM95 cells to study the influence of GSLs on DENV infection. Surprisingly, GM95 cells were highly resistant to DENV infection compared with B16 cells. Pretreatment of B16 cells with synthetase inhibitor of GM3, the most abundant GSLs in B16 cells, or silencing GM3 synthetase T3GAL5, significantly inhibited DENV infection. DENV attachment and endocytosis were not impaired in GM95 cells, but DENV genome replication was obviously inhibited in GM95 cells compared to B16 cells. Furthermore, GM3 was colocalized with DENV viral replication complex on endoplasmic reticulum (ER) inside the B16 cells. Finally, GM3 synthetase inhibitor significantly reduced the mortality rate of suckling mice that challenged with DENV by impairing the viral replication in mouse brain. Taken together, these data indicated that GM3 was not required for DENV attachment and endocytosis, however, essential for viral genome replication. Targeting GM3 could be a novel strategy to inhibit DENV infection.
Collapse
Affiliation(s)
- Kezhen Wang
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Juanjuan Wang
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Ta Sun
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Gang Bian
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Wen Pan
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Tingting Feng
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Penghua Wang
- 2. Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Yunsen Li
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Jianfeng Dai
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| |
Collapse
|
17
|
Pang EL, Loh HS. Current perspectives on dengue episode in Malaysia. ASIAN PAC J TROP MED 2016; 9:395-401. [PMID: 27086160 DOI: 10.1016/j.apjtm.2016.03.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/20/2016] [Indexed: 11/29/2022] Open
Abstract
Prevalence of dengue transmission has been alarmed by an estimate of 390 million infections per annum. Urban encroachment, ecological disruption and poor sanitation are all contributory factors of increased epidemiology. Complication however arises from the fact that dengue virus inherently exists as four different serotypes. Secondary infection is often manifested in the more severe form, such that antibody-dependent enhancement (ADE) could aggravate ailment by allowing pre-existing antibodies to form complexes with infecting viruses as means of intrusion. Consequently, increased viraemic titter and suppression of antiviral response are observed. Deep concerns are thus expressed in regards to escalating trend of hospitalisation and mortality rates. In Malaysia, situation is exacerbated by improper clinical management and pending vector control operations. As a preparedness strategy against the potential deadly dengue pandemic, the call for development of a durable and cost-effective dengue vaccine against all infecting serotypes is intensified. Even though several vaccine candidates are currently being evaluated in clinical trials, uncertainties in regards to serotypes interference, incomplete protection and dose adequacy have been raised. Instead of sole reliance on outsourcing, production of local vaccine should be considered in coherent to government's efforts to combat against dengue.
Collapse
Affiliation(s)
- Ee Leen Pang
- School of Biosciences, Faculty of Science, The University of Nottingham Malaysia Campus, Jalan Broga, 43500 Semenyih, Selangor, Malaysia
| | - Hwei-San Loh
- School of Biosciences, Faculty of Science, The University of Nottingham Malaysia Campus, Jalan Broga, 43500 Semenyih, Selangor, Malaysia; Biotechnology Research Centre, The University of Nottingham Malaysia Campus, Jalan Broga, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
18
|
Paul B, Tham WL. Controlling Dengue: Effectiveness of Biological Control and Vaccine in Reducing the Prevalence of Dengue Infection in Endemic Areas. Health (London) 2016. [DOI: 10.4236/health.2016.81008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Yamanaka A, Oddgun D, Chantawat N, Okabayashi T, Ramasoota P, Churrotin S, Kotaki T, Kameoka M, Soegijanto S, Konishi E. Dengue virus infection-enhancing antibody activities against Indonesian strains in inhabitants of central Thailand. Microbes Infect 2015; 18:277-84. [PMID: 26645957 DOI: 10.1016/j.micinf.2015.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 11/10/2015] [Accepted: 11/15/2015] [Indexed: 12/30/2022]
Abstract
Dengue virus (DENV) infection-enhancing antibodies are a hypothetic factor to increase the dengue disease severity. In this study, we investigated the enhancing antibodies against Indonesian strains of DENV-1-4 in 50 healthy inhabitants of central Thailand (Bangkok and Uthai Thani). Indonesia and Thailand have seen the highest dengue incidence in Southeast Asia. The infection history of each subject was estimated by comparing his/her neutralizing antibody titers against prototype DENV-1-4 strains. To resolve the difficulty in obtaining foreign live viruses for use as assay antigens, we used a recombinant system to prepare single-round infectious dengue viral particles based on viral sequence information. Irrespective of the previously infecting serotype(s), most serum samples showed significantly higher enhancement titers against Indonesian DENV-2 strains than against Thai DENV-2 strains, whereas the opposite effect was observed for the DENV-3 strains. Equivalent enhancing activities were observed against both DENV-1 and DENV-4. These results suggest that the genotype has an impact on enhancing antibody activities against DENV-2 and DENV-3, because the predominant circulating genotypes of each serotype differ between Indonesia and Thailand.
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan.
| | - Duangjai Oddgun
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Nantarat Chantawat
- Mahidol-Osaka Center for Infectious Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; Mahidol-Osaka Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Tamaki Okabayashi
- Mahidol-Osaka Center for Infectious Diseases, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; Mahidol-Osaka Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Pongrama Ramasoota
- Center of Excellence for Antibody Research, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| | - Siti Churrotin
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60015, Indonesia
| | - Tomohiro Kotaki
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60015, Indonesia; Department of International Health, Kobe University Graduate School of Health Sciences, Hyogo 654-0142, Japan
| | - Masanori Kameoka
- Department of International Health, Kobe University Graduate School of Health Sciences, Hyogo 654-0142, Japan
| | - Soegeng Soegijanto
- Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60015, Indonesia
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
20
|
Islam R, Salahuddin M, Ayubi MS, Hossain T, Majumder A, Taylor-Robinson AW, Mahmud-Al-Rafat A. Dengue epidemiology and pathogenesis: images of the future viewed through a mirror of the past. Virol Sin 2015; 30:326-43. [PMID: 26494479 PMCID: PMC8200867 DOI: 10.1007/s12250-015-3624-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/07/2015] [Indexed: 12/18/2022] Open
Abstract
Every year, millions of individuals throughout the world are seriously affected by dengue virus. The unavailability of a vaccine and of anti-viral drugs has made this mosquito-borne disease a serious health concern. Not only does dengue cause fatalities but it also has a profoundly negative economic impact. In recent decades, extensive research has been performed on epidemiology, vector biology, life cycle, pathogenesis, vaccine development and prevention. Although dengue research is still not at a stage to suggest definite hopes of a cure, encouraging significant advances have provided remarkable progress in the fight against infection. Recent developments indicate that both anti-viral drug and vaccine research should be pursued, in parallel with vector control programs.
Collapse
Affiliation(s)
- Rashedul Islam
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Mohammed Salahuddin
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Md Salahuddin Ayubi
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Tahmina Hossain
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Apurba Majumder
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9100, Bangladesh
| | - Andrew W Taylor-Robinson
- School of Medical & Applied Sciences, Central Queensland University, Rockhampton, 4701, Australia
| | - Abdullah Mahmud-Al-Rafat
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9100, Bangladesh.
- Research and Development (R&D) Department, Incepta Vaccine Limited, Zirabo, Savar, Dhaka, 1341, Bangladesh.
| |
Collapse
|
21
|
Autophagy Activated by Bluetongue Virus Infection Plays a Positive Role in Its Replication. Viruses 2015; 7:4657-75. [PMID: 26287233 PMCID: PMC4576199 DOI: 10.3390/v7082838] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/27/2015] [Accepted: 07/30/2015] [Indexed: 01/19/2023] Open
Abstract
Bluetongue virus (BTV) is an important pathogen of wild and domestic ruminants. Despite extensive study in recent decades, the interplay between BTV and host cells is not clearly understood. Autophagy as a cellular adaptive response plays a part in many viral infections. In our study, we found that BTV1 infection triggers the complete autophagic process in host cells, as demonstrated by the appearance of obvious double-membrane autophagosome-like vesicles, GFP-LC3 dots accumulation, the conversion of LC3-I to LC3-II and increased levels of autophagic flux in BSR cells (baby hamster kidney cell clones) and primary lamb lingual epithelial cells upon BTV1 infection. Moreover, the results of a UV-inactivated BTV1 infection assay suggested that the induction of autophagy was dependent on BTV1 replication. Therefore, we investigated the role of autophagy in BTV1 replication. The inhibition of autophagy by pharmacological inhibitors (3-MA, CQ) and RNA interference (siBeclin1) significantly decreased viral protein synthesis and virus yields. In contrast, treating BSR cells with rapamycin, an inducer of autophagy, promoted viral protein expression and the production of infectious BTV1. These findings lead us to conclude that autophagy is activated by BTV1 and contributes to its replication, and provide novel insights into BTV-host interactions.
Collapse
|
22
|
Meng F, Badierah RA, Almehdar HA, Redwan EM, Kurgan L, Uversky VN. Unstructural biology of the dengue virus proteins. FEBS J 2015; 282:3368-94. [DOI: 10.1111/febs.13349] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 06/01/2015] [Accepted: 06/15/2015] [Indexed: 01/02/2023]
Affiliation(s)
- Fanchi Meng
- Department of Electrical and Computer Engineering; University of Alberta; Edmonton Alberta Canada
| | - Reaid A. Badierah
- Biological Department; Faculty of Science; King Abdulaziz University; Jeddah Saudi Arabia
| | - Hussein A. Almehdar
- Biological Department; Faculty of Science; King Abdulaziz University; Jeddah Saudi Arabia
| | - Elrashdy M. Redwan
- Biological Department; Faculty of Science; King Abdulaziz University; Jeddah Saudi Arabia
- Therapeutic and Protective Proteins Laboratory; Protein Research Department; Genetic Engineering and Biotechnology Research Institute; City for Scientific Research and Technology Applications; New Borg El-Arab Alexandria Egypt
| | - Lukasz Kurgan
- Department of Electrical and Computer Engineering; University of Alberta; Edmonton Alberta Canada
| | - Vladimir N. Uversky
- Biological Department; Faculty of Science; King Abdulaziz University; Jeddah Saudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute; Morsani College of Medicine; University of South Florida; Tampa FL USA
- Laboratory of Structural Dynamics, Stability and Folding of Proteins; Institute of Cytology; Russian Academy of Sciences; St Petersburg Russia
| |
Collapse
|
23
|
Pereira GV, Freitas VA, Oliveira HS, Oliveira LCA, Belchior JC. A photocatalytic process for the eradication of dengue through ˙OH generation in the presence of sunlight and iron oxide. RSC Adv 2014. [DOI: 10.1039/c4ra13435d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|