1
|
Singh G, Singh K, Sinha RA, Singh A, Khushi, Kumar A. Japanese encephalitis virus infection causes reactive oxygen species-mediated skeletal muscle damage. Eur J Neurosci 2024; 60:4843-4860. [PMID: 39049535 DOI: 10.1111/ejn.16469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024]
Abstract
Skeletal muscle wasting is a clinically proven pathology associated with Japanese encephalitis virus (JEV) infection; however, underlying factors that govern skeletal muscle damage are yet to be explored. The current study aims to investigate the pathobiology of skeletal muscle damage using a mouse model of JEV infection. Our study reveals a significant increment in viral copy number in skeletal muscle post-JEV infection, which is associated with enhanced skeletal muscle cell death. Molecular and biochemical analysis confirms NOX2-dependent generation of reactive oxygen species, leading to autophagy flux inhibition and cell apoptosis. Along with this, an alteration in mitochondrial dynamics (change in fusion and fission process) and a decrease in the total number of mitochondria copies were found during JEV disease progression. The study represents the initial evidence of skeletal muscle damage caused by JEV and provides insights into potential avenues for therapeutic advancement.
Collapse
Affiliation(s)
- Gajendra Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Kulwant Singh
- Stem Cell Research Center, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Anjali Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Khushi
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Alok Kumar
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| |
Collapse
|
2
|
Zhang Y, Zhang X, Yang X, Lv L, Wang Q, Zeng S, Zhang Z, Dorf M, Li S, Zhao L, Fu B. AP3B1 facilitates PDIA3/ERP57 function to regulate rabies virus glycoprotein selective degradation and viral entry. Autophagy 2024:1-19. [PMID: 39128851 DOI: 10.1080/15548627.2024.2390814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024] Open
Abstract
Rabies virus causes an estimated 59,000 annual fatalities worldwide and promising therapeutic treatments are necessary to develop. In this study, affinity tag-purification mass spectrometry was employed to delineate RABV glycoprotein and host protein interactions, and PDIA3/ERP57 was identified as a potential inhibitor of RABV infection. PDIA3 restricted RABV infection with follow mechanisms: PDIA3 mediated the degradation of RABV G protein by targeting lysine 332 via the selective macroautophagy/autophagy pathway; The PDIA3 interactor, AP3B1 (adaptor related protein complex 3 subunit beta 1) was indispensable in PDIA3-triggered selective degradation of the G protein; Furthermore, PDIA3 competitively bound with NCAM1/NCAM (neural cell adhesion molecule 1) to block RABV G, hindering viral entry into host cells. PDIA3 190-199 aa residues bound to the RABV G protein were necessary and sufficient to defend against RABV. These results demonstrated the therapeutic potential of biologics that target PDIA3 or utilize PDIA3 190-199 aa peptide to treat clinical rabies.Abbreviation: aa: amino acids; ANXA2: annexin A2; AP-MS: affinity tag purification-mass spectrometry; AP3B1: adaptor related protein complex 3 subunit beta 1; ATP6V1A: ATPase H+ transporting V1 subunit A; ATP6V1H: ATPase H+ transporting V1 subunit H; BafA1: bafilomycin A1; CHX: cycloheximide; co-IP: co-immunoprecipitation; DDX17: DEAD-box helicase 17; DmERp60: drosophila melanogaster endoplasmic reticulum p60; EBOV: Zaire ebolavirus virus; EV: empty vector; GANAB: glucosidase II alpha subunit; G protein: glycoprotein; GRM2/mGluR2: glutamate metabotropic receptor 2; HsPDIA3: homo sapiens protein disulfide isomerase family A member 3; IAV: influenza virus; ILF2: interleukin enhancer binding factor 2; KO: knockout; MAGT1: magnesium transporter 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MmPDIA3: mus musculus protein disulfide isomerase associated 3; NCAM1/NCAM: neural cell adhesion molecule 1; NGFR/p75NTR: nerve growth factor receptor; NGLY1: N-glycanase 1; OTUD4: OTU deubiquitinase 4; PDI: protein disulfide isomerase; PPIs: protein-protein interactions; RABV: rabies virus; RUVBL2: RuvB like AAA ATPase 2; SCAMP3: secretory carrier membrane protein 3; ScPdi1: Saccharomyces cerevisiae s288c protein disulfide isomerase 1; SLC25A6: solute carrier family 25 member 6; SQSTM1/p62: sequestosome 1; VSV: vesicular stomatitis virus.
Collapse
Affiliation(s)
- Yuelan Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xinyi Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xue Yang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Linyue Lv
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Qinyang Wang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Shaowei Zeng
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Zhuyou Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Martin Dorf
- Department of Microbiology & Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Shitao Li
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, USA
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
| | - Bishi Fu
- Department of Rheumatology and Immunology, State Key Laboratory of Virology, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Yuan Y, Fang A, Wang Z, Chen H, Fu ZF, Zhou M, Zhao L. The matrix protein of lyssavirus hijacks autophagosome for efficient egress by recruiting NEDD4 through its PPxY motif. Autophagy 2024; 20:1723-1740. [PMID: 38566321 PMCID: PMC11262214 DOI: 10.1080/15548627.2024.2338575] [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: 05/23/2023] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
Lyssaviruses are well-known worldwide and often cause fatal encephalitis. Previous studies have shown that autophagy is beneficial for the replication of rabies virus (RABV), the representative lyssavirus, but the detailed mechanism remains obscure. In this study, we showed that the rabies virus matrix protein (RABV-M) used its PPxY motif to interact with the E3 ubiquitin-protein ligase NEDD4. NEDD4 then recruited MAP1LC3/LC3 via its LC3-interacting region (LIR). Interestingly, after binding to the ubiquitinated RABV-M, NEDD4 could bind more LC3 and enhance autophagosome accumulation, while NEDD4 knockdown significantly reduced M-induced autophagosome accumulation. Further study revealed that RABV-M prevented autophagosome-lysosome fusion and facilitated viral budding. Inhibition of RABV-M-induced autophagosome accumulation reduced the production of extracellular virus-like particles. We also found that M proteins of most lyssaviruses share the same mechanism to accumulate autophagosome by hijacking NEDD4. Collectively, this study revealed a novel strategy for lyssaviruses to achieve efficient viral replication by exploiting the host autophagy system.Abbreviations: ABLV: Australian bat lyssavirus; ATG5: autophagy related 5; Baf A1:bafilomycin A1;co-IP: co-immunoprecipitation; CQ: chloroquine; DAPI:4',6-diamidino-2'-phenylindole; DMSO: dimethyl sulfoxide; EBLV:European bat lyssavirus; GFP: green fluorescent protein; GST:glutathione S-transferase; hpi: hours post-infection; hpt: hourspost-transfection; LIR: LC3-interactingregion;MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; mCherry:red fluorescent protein; MOI: multiplicity of infection; NC: negativecontrol; MVB: multivesicular body; NEDD4: neural precursorcell-expressed developmentally down-regulated 4; RABV: rabies virus;SQSTM1/p62: sequestosome 1; VLP: virus-like particle; VPS4B: vacuolarprotein sorting 4B; TEM: transmission electron microscopy; WB:western blotting; WT: wild-type; μm: micrometer; μM: micromole.
Collapse
Affiliation(s)
- Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, China
| | - An Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, China
| | - Zhihui Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
4
|
Yuan Y, Fang A, Zhang M, Zhou M, Fu ZF, Zhao L. Lassa virus Z protein hijacks the autophagy machinery for efficient transportation by interrupting CCT2-mediated cytoskeleton network formation. Autophagy 2024:1-18. [PMID: 39007910 DOI: 10.1080/15548627.2024.2379099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024] Open
Abstract
The Lassa virus (LASV) is a widely recognized virulent pathogen that frequently results in lethal viral hemorrhagic fever (VHF). Earlier research has indicated that macroautophagy/autophagy plays a role in LASV replication, but, the precise mechanism is unknown. In this present study, we show that LASV matrix protein (LASV-Z) is essential for blocking intracellular autophagic flux. LASV-Z hinders actin and tubulin folding by interacting with CCT2, a component of the chaperonin-containing T-complexes (TRiC). When the cytoskeleton is disrupted, lysosomal enzyme transit is hampered. In addition, cytoskeleton disruption inhibits the merge of autophagosomes with lysosomes, resulting in autophagosome accumulation that promotes the budding of LASV virus-like particles (VLPs). Inhibition of LASV-Z-induced autophagosome accumulation blocks the LASV VLP budding process. Furthermore, it is found that glutamine at position 29 and tyrosine at position 48 on LASV-Z are important in interacting with CCT2. When these two sites are mutated, LASV-mut interacts with CCT2 less efficiently and can no longer inhibit the autophagic flux. These findings demonstrate a novel strategy for LASV-Z to hijack the host autophagy machinery to accomplish effective transportation.Abbreviation: 3-MA: 3-methyladenine; ATG5: autophagy related 5; ATG7: autophagy related 7; Baf-A1: bafilomycin A1; CCT2: chaperonin containing TCP1 subunit 2; co-IP: co-immunoprecipitation; CTSD: cathepsin D; DAPI: 4',6-diamidino-2'-phenylindole; DMSO: dimethyl sulfoxide; EGFR: epidermal growth factor receptor; GFP: green fluorescent protein; hpi: hours post-infection; hpt: hours post-transfection; LAMP1: lysosomal-associated membrane protein 1; LASV: lassa virus; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; mCherry: red fluorescent protein; PM: plasma membrane; SQSTM1/p62: sequestosome 1; STX6: syntaxin 6; VLP: virus-like particle; TEM: transmission electron microscopy; TRiC: chaperonin-containing T-complex; WB: western blotting; μm: micrometer; μM: micromole.
Collapse
Affiliation(s)
- Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - An Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mai Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
5
|
Jin F, Jiang X, Ni X, Yu S, Wu F, Shi X, Mao D, Wang H, Shi Q, Liu Y, Xu Q. Alpha-Hederin induces incomplete autophagic injury in non-small cell lung cancer by interfering with the lysosomal acidification. Sci Rep 2024; 14:13258. [PMID: 38858422 PMCID: PMC11164981 DOI: 10.1038/s41598-024-63348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024] Open
Abstract
Lung cancer is the most common oncological disease worldwide, with non-small cell lung cancer accounting for approximately 85% of lung cancer cases. α-Hederin is a monodesmosidic triterpenoid saponin isolated from the leaves of Hedera helix L. or Nigella sativa and has been extensively studied for its antitumor activity against a variety of tumor cells. It has been suggested that α-Hederin is a potential regulator of autophagy and has high promise for application. However, the specific mechanism and characteristics of α-Hederin in regulating autophagy are not well understood. In this study, we confirmed the potential of α-Hederin application in lung cancer treatment and comprehensively explored the mechanism and characteristics of α-Hederin in regulating autophagy in lung cancer cells. Our results suggest that α-Hederin is an incomplete autophagy inducer that targets mTOR to activate the classical autophagic pathway, inhibits lysosomal acidification without significantly affecting the processes of autophagosome transport, lysosome biogenesis, autophagosome and lysosome fusion, and finally leads to impaired autophagic flux and triggers autophagic damage in NSCLC.
Collapse
Affiliation(s)
- Feng Jin
- Department of Respiratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Xiaomin Jiang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China
| | - Xiaochen Ni
- Department of Respiratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Shilong Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou, 225001, China
| | - Feng Wu
- Department of Respiratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Xinlin Shi
- Department of Respiratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Defang Mao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Haibo Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China
| | - Qingtong Shi
- Department of Thoracic Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China
| | - Qin Xu
- Department of Respiratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
6
|
He H, Cai T, Chen Q, Chen Z, Zhang B, Chen C, Wang Y, Liu Y, Wang Y, Luo Y, Huang S, Luo J, Guo X. TRIM44 Promotes Rabies Virus Replication by Autophagy-Dependent Mechanism. Int J Mol Sci 2024; 25:4616. [PMID: 38731834 PMCID: PMC11083291 DOI: 10.3390/ijms25094616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Tripartite motif (TRIM) proteins are a multifunctional E3 ubiquitin ligase family that participates in various cellular processes. Recent studies have shown that TRIM proteins play important roles in regulating host-virus interactions through specific pathways, but their involvement in response to rabies virus (RABV) infection remains poorly understood. Here, we identified that several TRIM proteins are upregulated in mouse neuroblastoma cells (NA) after infection with the rabies virus using RNA-seq sequencing. Among them, TRIM44 was found to regulate RABV replication. This is supported by the observations that downregulation of TRIM44 inhibits RABV replication, while overexpression of TRIM44 promotes RABV replication. Mechanistically, TRIM44-induced RABV replication is brought about by activating autophagy, as inhibition of autophagy with 3-MA attenuates TRIM44-induced RABV replication. Additionally, we found that inhibition of autophagy with rapamycin reverses the TRIM44-knockdown-induced decrease in LC3B expression and autophagosome formation as well as RABV replication. The results suggest that TRIM44 promotes RABV replication by an autophagy-dependent mechanism. Our work identifies TRIM44 as a key host factor for RABV replication, and targeting TRIM44 expression may represent an effective therapeutic strategy.
Collapse
Affiliation(s)
- Hongling He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Ting Cai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Qiaozhu Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Zilian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Changyi Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Yueze Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Yan Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Yueming Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (H.H.); (T.C.); (Q.C.); (Z.C.); (B.Z.); (C.C.); (Y.W.); (Y.L.); (Y.W.); (Y.L.)
| |
Collapse
|
7
|
Han Y, Wang C, Lu K, Dong X, Chang Z, Zhang R, Hou Q, Wang X, Xiao S, Liu H, Yang Z. Bovine parainfluenza type 3 virus induces incomplete autophagy to promote viral replication by activated beclin1 in vitro. Vet Microbiol 2024; 290:109972. [PMID: 38183839 DOI: 10.1016/j.vetmic.2023.109972] [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: 10/12/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
Bovine Parainfluenza virus Type 3 (BPIV3) is one of the most important pathogens in cattle, capable of causing severe respiratory symptoms. Numerous studies have shown that autophagy plays a diverse role in the infection process of various pathogens. The influence of autophagy machinery on BPIV3 infection has not yet been confirmed. In the present study, we initially demonstrated that the expression of LC3 was significantly increased and exhibited a notable increase in double or single-membrane vesicles under a transmission electron microscope during BPIV3 infection. These observations unequivocally establish the induction of steady-state autophagy in vitro consequent to BPIV3 infection. Furthermore, quantification of autophagic flux substantiates the induction of an incomplete autophagic process during BPIV3 infection. Additionally, through targeted interventions, we demonstrate the regulatory impact of pharmacological agents influencing autophagy and RNA interference targeting an autophagy-associated protein on viral replication. Intriguingly, our data revealed that BPIV3 infection enhanced the phosphorylation of rapamycin kinase (mTOR). This result demonstrated that mTOR does not operate as a counteractive regulator of BPIV3-induced autophagy. Instead, we discern an augmentation in the expression of Beclin1, a key autophagy initiator, which complexes with Vps34, constituting a Class III phosphatidylinositol 3-kinase. This phenomenon serves as a hallmark in the inaugural phase of autophagy initiation during BPIV3 infection. Collectively, these discernments underscore that BPIV3 infection actively stimulates autophagy, thereby enhancing viral replication through the activation of Beclin1, independently of the mTOR signaling pathway. This nuanced comprehension significantly contributes to unraveling the intricate molecular mechanisms governing BPIV3-induced autophagy.
Collapse
Affiliation(s)
- Yu Han
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Chongyang Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province 311300, China
| | - Kejia Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyu Dong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhengwu Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Riteng Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qili Hou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
8
|
Li S, Xu B, Luo Y, Luo J, Huang S, Guo X. Autophagy and Apoptosis in Rabies Virus Replication. Cells 2024; 13:183. [PMID: 38247875 PMCID: PMC10814280 DOI: 10.3390/cells13020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/28/2023] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Rabies virus (RABV) is a single-stranded negative-sense RNA virus belonging to the Rhabdoviridae family and Lyssavirus genus, which is highly neurotropic and can infect almost all warm-blooded animals, including humans. Autophagy and apoptosis are two evolutionarily conserved and genetically regulated processes that maintain cellular and organismal homeostasis, respectively. Autophagy recycles unnecessary or dysfunctional intracellular organelles and molecules in a cell, whereas apoptosis eliminates damaged or unwanted cells in an organism. Studies have shown that RABV can induce both autophagy and apoptosis in target cells. To advance our understanding of pathogenesis of rabies, this paper reviews the molecular mechanisms of autophagy and apoptosis induced by RABV and the effects of the two cellular events on RABV replication.
Collapse
Affiliation(s)
- Saisai Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| | - Bowen Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China;
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (Y.L.)
| |
Collapse
|
9
|
Yin J, Wang S, Ren S, Liang Z, Ge J, Sun Y, Yin X, Wang X. TMP269, a small molecule inhibitor of class IIa HDAC, suppresses RABV replication in vitro. Front Microbiol 2023; 14:1284439. [PMID: 38107853 PMCID: PMC10722228 DOI: 10.3389/fmicb.2023.1284439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
TMP269, a small molecular inhibitor of IIa histone deacetylase, plays a vital role in cancer therapeutic. However, the effect of TMP269 on the regulation of viral replication has not been studied. In the present study, we found that TMP269 treatment significantly inhibited RABV replication at concentrations without significant cytotoxicity in a dose-dependent manner. In addition, TMP269 can reduce the viral titers and protein levels of RABV at an early stage in the viral life cycle. RNA sequencing data revealed that immune-related pathways and autophagy-related genes were significantly downregulated after RABV infection treated with TMP269. Further exploration shows that autophagy enhances RABV replication in HEK-293T cells, while TMP269 can inhibit autophagy to decrease RABV replication. Together, these results provide a novel treatment strategy for rabies.
Collapse
Affiliation(s)
- Juanbin Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shasha Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shanhui Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhengji Liang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Junwei Ge
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
10
|
Bastos V, Pacheco V, Rodrigues ÉDL, Moraes CNS, Nóbile AL, Fonseca DLM, Souza KBS, do Vale FYN, Filgueiras IS, Schimke LF, Giil LM, Moll G, Cabral-Miranda G, Ochs HD, Vasconcelos PFDC, de Melo GD, Bourhy H, Casseb LMN, Cabral-Marques O. Neuroimmunology of rabies: New insights into an ancient disease. J Med Virol 2023; 95:e29042. [PMID: 37885152 DOI: 10.1002/jmv.29042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023]
Abstract
Rabies is an ancient neuroinvasive viral (genus Lyssavirus, family Rhabdoviridae) disease affecting approximately 59,000 people worldwide. The central nervous system (CNS) is targeted, and rabies has a case fatality rate of almost 100% in humans and animals. Rabies is entirely preventable through proper vaccination, and thus, the highest incidence is typically observed in developing countries, mainly in Africa and Asia. However, there are still cases in European countries and the United States. Recently, demographic, increasing income levels, and the coronavirus disease 2019 (COVID-19) pandemic have caused a massive raising in the animal population, enhancing the need for preventive measures (e.g., vaccination, surveillance, and animal control programs), postexposure prophylaxis, and a better understanding of rabies pathophysiology to identify therapeutic targets, since there is no effective treatment after the onset of clinical manifestations. Here, we review the neuroimmune biology and mechanisms of rabies. Its pathogenesis involves a complex and poorly understood modulation of immune and brain functions associated with metabolic, synaptic, and neuronal impairments, resulting in fatal outcomes without significant histopathological lesions in the CNS. In this context, the neuroimmunological and neurochemical aspects of excitatory/inhibitory signaling (e.g., GABA/glutamate crosstalk) are likely related to the clinical manifestations of rabies infection. Uncovering new links between immunopathological mechanisms and neurochemical imbalance will be essential to identify novel potential therapeutic targets to reduce rabies morbidity and mortality.
Collapse
Affiliation(s)
- Victor Bastos
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Vinicius Pacheco
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Érika D L Rodrigues
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Cássia N S Moraes
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Adriel L Nóbile
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
| | - Dennyson Leandro M Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo, São Paulo, Brazil
| | - Kamilla B S Souza
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Fernando Y N do Vale
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
| | - Igor S Filgueiras
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Lasse M Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | | | - Hans D Ochs
- School of Medicine and Seattle Children's Research Institute, University of Washington, Seattle, Washington, USA
| | - Pedro F da Costa Vasconcelos
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
- Department of Pathology, University of the State of Pará, Belem, Brazil
| | - Guilherme D de Melo
- Lyssavirus Epidemiology and Neuropathology Unit, WHO Collaborating Centre for Reference and Research on Rabies, Institut Pasteur, Université Paris Cité, Paris, France
| | - Hervé Bourhy
- Lyssavirus Epidemiology and Neuropathology Unit, WHO Collaborating Centre for Reference and Research on Rabies, Institut Pasteur, Université Paris Cité, Paris, France
| | - Livia M N Casseb
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Otavio Cabral-Marques
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
- Department of Immunology, University of São Paulo, São Paulo, Brazil
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
- Laboratory of Medical Investigation 29, School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Xie Y, Chi YL, Liu SQ, Zhu WY. BCX4430 inhibits the replication of rabies virus by suppressing mTOR-dependent autophagy invitro. Virology 2023; 585:21-31. [PMID: 37267717 DOI: 10.1016/j.virol.2023.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Rabies is a fatal neurological infectious disease caused by rabies virus (RABV). However, no effective anti-RABV drugs for treatment during the symptomatic phase are available. The novel adenosine nucleoside analog galidesivir (BCX4430) has broad-spectrum activity against a wide variety of highly pathogenic RNA viruses. In this study, we observed no apparent cytotoxicity of BCX4430 at the highest concentration of 250 μΜ, and which was displayed stronger antiviral activity against different virulent RABV in N2a or BHK-21 cells until 72 hpi. Meanwhile, BCX4430 showed greater anti-RABV activity than T-705 and anti-RABV activity similar to that of ribavirin in N2a cells. Furthermore, BCX4430 dose- and time-dependently inhibited RABV replication via mTOR-dependent autophagy inhibition in N2a cells with increased phospho-mTOR and phospho-SQSTM1 and decreased LC3-II levels. Taken together, these findings suggest that BCX4430 has potent anti-RABV activity in vitro and might provide a basis for the development of novel drug therapies against RABV.
Collapse
Affiliation(s)
- Yuan Xie
- Key Laboratory of Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, NHC Key Laboratory of Biosafety, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China
| | - Ying Lin Chi
- Key Laboratory of Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, NHC Key Laboratory of Biosafety, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China; School of Public Health, Baotou Medical College, Baotou, 014040, Inner Mongolia, PR China
| | - Shu Qing Liu
- Key Laboratory of Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, NHC Key Laboratory of Biosafety, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China.
| | - Wu Yang Zhu
- Key Laboratory of Medical Virology, Ministry of Health, National Institute for Viral Disease Control and Prevention, NHC Key Laboratory of Biosafety, Chinese Center for Disease Control and Prevention, Beijing, 102206, PR China.
| |
Collapse
|
12
|
Zhang B, Cai T, He H, Huang X, Luo Y, Huang S, Luo J, Guo X. TRIM25 Suppresses Rabies Virus Fixed HEP-Flury Strain Production by Activating RIG-1-Mediated Type I Interferons. Genes (Basel) 2023; 14:1555. [PMID: 37628607 PMCID: PMC10454932 DOI: 10.3390/genes14081555] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Rabies remains a great threat to public health worldwide. So far, the mechanism of rabies virus (RABV) infection is not fully understood, and there is no effective treatment for rabies. Identifying more host restriction factors of RABV will spur the development of novel therapeutic interventions against rabies. Accumulating studies suggest that tripartite motif-containing (TRIM) proteins have great effects on virus replication. TRIMs control the antiviral responses through either direct interaction with viral proteins or indirect regulation of innate immune signaling molecules in the host. The role of TRIM25 in rabies virus (RABV) infection is poorly understood. Using next-generation sequencing, we found that TRIM25 is upregulated during HEP-Flury infection. Knockdown of TRIM25 enhances HEP-Flury production, while overexpression of TRIM25 suppresses HEP-Flury replication. Knockdown of interferon α and interferon β weakens the anti-RABV response induced by TRIM25 overexpression, and potentiates RABV production. Furthermore, we found that TRIM25 regulates type-I interferon response by targeting retinoic acid-inducible gene I (RIG-I) during HEP-Flury infection. Knockdown of RIG-I weakens the anti-HEP-Flury response induced by TRIM25 overexpression, indicating that TRIM25 regulates RABV production via the RIG-I-IFN axis. In addition, we observed that TRIM25 does not directly interact with HEP-Flury structural proteins, suggesting that TRIM25 regulates HEP-Flury production indirectly. Taken together, our work identifies TRIM25 as a new host factor involved in HEP-Flury infection, which may be a potential target for the development of antiviral drugs against RABV.
Collapse
Affiliation(s)
- Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Ting Cai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Hongling He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Xuezhe Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| |
Collapse
|
13
|
Hou P, Guo Y, Jin H, Sun J, Bai Y, Li W, Li L, Cao Z, Wu F, Zhang H, Li Y, Yang S, Xia X, Huang P, Wang H. Bif-1c Attenuates Viral Proliferation by Regulating Autophagic Flux Blockade Induced by the Rabies Virus CVS-11 Strain in N2a Cells. Microbiol Spectr 2023; 11:e0307922. [PMID: 37014208 PMCID: PMC10269655 DOI: 10.1128/spectrum.03079-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Bax-interacting factor-1 (Bif-1) is a multifunctional protein involved in apoptosis, autophagy, and mitochondrial morphology. However, the associations between Bif-1 and viruses are poorly understood. As discrete Bif-1 isoforms are selectively expressed and exert corresponding effects, we evaluated the effects of neuron-specific/ubiquitous Bif-1 isoforms on rabies virus (RABV) proliferation. First, infection with the RABV CVS-11 strain significantly altered Bif-1 expression in mouse neuroblastoma (N2a) cells, and Bif-1 knockdown in turn promoted RABV replication. Overexpression of neuron-specific Bif-1 isoforms (Bif-1b/c/e) suppressed RABV replication. Moreover, our study showed that Bif-1c colocalized with LC3 and partially alleviated the incomplete autophagic flux induced by RABV. Taken together, our data reveal that neuron-specific Bif-1 isoforms impair the RABV replication process by abolishing autophagosome accumulation and blocking autophagic flux induced by the RABV CVS-11 strain in N2a cells. IMPORTANCE Autophagy can be triggered by viral infection and replication. Autophagosomes are generated and affect RABV replication, which differs by viral strain and infected cell type. Bax-interacting factor-1 (Bif-1) mainly has a proapoptotic function but is also involved in autophagosome formation. However, the association between Bif-1-involved autophagy and RABV infection remains unclear. In this study, our data reveal that a neuron-specific Bif-1 isoform, Bif-1c, impaired viral replication by unchoking autophagosome accumulation induced by RABV in N2a cells to a certain extent. Our study reveals for the first time that Bif-1 is involved in modulating autophagic flux and plays a crucial role in RABV replication, establishing Bif-1 as a potential therapeutic target for rabies.
Collapse
Affiliation(s)
- Pengfei Hou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yidi Guo
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hongli Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jingxuan Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yujie Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wujian Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ling Li
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Zengguo Cao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fangfang Wu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Haili Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuanyuan Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Pei Huang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hualei Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
14
|
Zhang Y, Mo R, Sun S, Cui Z, Liang B, Li E, Wang T, Feng Y, Yang S, Yan F, Zhao Y, Xia X. Bacillus subtilis vector based oral rabies vaccines induced potent immune response and protective efficacy in mice. Front Microbiol 2023; 14:1126533. [PMID: 36846792 PMCID: PMC9948087 DOI: 10.3389/fmicb.2023.1126533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Rabies is a worldwide epidemic that poses a serious threat to global public health. At present, rabies in domestic dogs, cats, and some pets can be effectively prevented and controlled by intramuscular injection of rabies vaccine. But for some inaccessible animals, especially stray dogs, and wild animals, it is difficult to prevent with intramuscular injection. Therefore, it is necessary to develop a safe and effective oral rabies vaccine. Methods We constructed recombinant Bacillus subtilis (B. subtilis) expressing two different strains of rabies virus G protein, named CotG-E-G and CotG-C-G, immunogenicity was studied in mice. Results The results showed that CotG-E-G and CotG-C-G could significantly increase the specific SIgA titers in feces, serum IgG titers, and neutralizing antibodies. ELISpot experiments showed that CotG-E-G and CotG-C-G could also induce Th1 and Th2 to mediate the secretion of immune-related IFN-γ and IL-4. Collectively, our results suggested that recombinant B. subtilis CotG-E-G and CotG-C-G have excellent immunogenicity and are expected to be novel oral vaccine candidates for the prevention and control of wild animal rabies.
Collapse
Affiliation(s)
- Ying Zhang
- Northeast Forestry University College of Wildlife and Protected Area, Harbin, China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Ruo Mo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Sheng Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Zhanding Cui
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Bo Liang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Ye Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,Feihu Yan,✉
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,Yongkun Zhao,✉
| | - Xianzhu Xia
- Northeast Forestry University College of Wildlife and Protected Area, Harbin, China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China,*Correspondence: Xianzhu Xia,✉
| |
Collapse
|
15
|
Gérard FCA, Bourhis JM, Mas C, Branchard A, Vu DD, Varhoshkova S, Leyrat C, Jamin M. Structure and Dynamics of the Unassembled Nucleoprotein of Rabies Virus in Complex with Its Phosphoprotein Chaperone Module. Viruses 2022; 14:v14122813. [PMID: 36560817 PMCID: PMC9786881 DOI: 10.3390/v14122813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
As for all non-segmented negative RNA viruses, rabies virus has its genome packaged in a linear assembly of nucleoprotein (N), named nucleocapsid. The formation of new nucleocapsids during virus replication in cells requires the production of soluble N protein in complex with its phosphoprotein (P) chaperone. In this study, we reconstituted a soluble heterodimeric complex between an armless N protein of rabies virus (RABV), lacking its N-terminal subdomain (NNT-ARM), and a peptide encompassing the N0 chaperon module of the P protein. We showed that the chaperone module undergoes a disordered-order transition when it assembles with N0 and measured an affinity in the low nanomolar range using a competition assay. We solved the crystal structure of the complex at a resolution of 2.3 Å, unveiling the details of the conserved interfaces. MD simulations showed that both the chaperon module of P and RNA-mediated polymerization reduced the ability of the RNA binding cavity to open and close. Finally, by reconstituting a complex with full-length P protein, we demonstrated that each P dimer could independently chaperon two N0 molecules.
Collapse
Affiliation(s)
- Francine C. A. Gérard
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Jean-Marie Bourhis
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Caroline Mas
- Integrated Structural Biology Grenoble (ISBG), Université Grenoble Alpes, CNRS, CEA, EMBL, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Anaïs Branchard
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Duc Duy Vu
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Sylvia Varhoshkova
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Cédric Leyrat
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
- Correspondence: (C.L.); (M.J.)
| | - Marc Jamin
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, 71 Avenue des Martyrs, 38000 Grenoble, France
- Correspondence: (C.L.); (M.J.)
| |
Collapse
|
16
|
Zhang H, Huang J, Song Y, Liu X, Qian M, Huang P, Li Y, Zhao L, Wang H. Regulation of innate immune responses by rabies virus. Animal Model Exp Med 2022; 5:418-429. [PMID: 36138548 PMCID: PMC9610147 DOI: 10.1002/ame2.12273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/31/2022] [Indexed: 11/10/2022] Open
Abstract
Rabies virus (RABV) is an infectious and neurotropic pathogen that causes rabies and infects humans and almost all warm-blooded animals, posing a great threat to people and public safety. It is well known that innate immunity is the critical first line of host defense against viral infection. It monitors the invading pathogens by recognizing the pathogen-associated molecular patterns and danger-associated molecular patterns through pattern-recognition receptors, leading to the production of type I interferons (IFNα/β), inflammatory cytokines, and chemokines, or the activation of autophagy or apoptosis to inhibit virus replication. In the case of RABV, the innate immune response is usually triggered when the skin or muscle is bitten or scratched. However, RABV has evolved many ways to escape or even hijack innate immune response to complete its own replication and eventually invades the central nervous system (CNS). Once RABV reaches the CNS, it cannot be wiped out by the immune system or any drugs. Therefore, a better understanding of the interplay between RABV and innate immunity is necessary to develop effective strategies to combat its infection. Here, we review the innate immune responses induced by RABV and illustrate the antagonism mechanisms of RABV to provide new insights for the control of rabies.
Collapse
Affiliation(s)
- Haili Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Jingbo Huang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Yumeng Song
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Xingqi Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Meichen Qian
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Pei Huang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Yuanyuan Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Ling Zhao
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
| | - Hualei Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of EducationInstitute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| |
Collapse
|
17
|
Yuan Y, Fang A, Wang Z, Tian B, Zhang Y, Sui B, Luo Z, Li Y, Zhou M, Chen H, Fu ZF, Zhao L. Trim25 restricts rabies virus replication by destabilizing phosphoprotein. CELL INSIGHT 2022; 1:100057. [PMID: 37193556 PMCID: PMC10120326 DOI: 10.1016/j.cellin.2022.100057] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/21/2022] [Accepted: 09/25/2022] [Indexed: 05/18/2023]
Abstract
Tripartite motif-containing protein 25 (Trim25) is an E3 ubiquitin ligase that activates retinoid acid-inducible gene I (RIG-I) and promotes the antiviral interferon response. Recent studies have shown that Trim25 can bind and degrade viral proteins, suggesting a different mechanism of Trim25 on its antiviral effects. In this study, Trim25 expression was upregulated in cells and mouse brains after rabies virus (RABV) infection. Moreover, expression of Trim25 limited RABV replication in cultured cells. Overexpression of Trim25 caused attenuated viral pathogenicity in a mouse model that was intramuscularly injected with RABV. Further experiments confirmed that Trim25 inhibited RABV replication via two different mechanisms: an E3 ubiquitin ligase-dependent mechanism and an E3 ubiquitin ligase-independent mechanism. Specifically, the CCD domain of Trim25 interacted with RABV phosphoprotein (RABV-P) at amino acid (AA) position at 72 and impaired the stability of RABV-P via complete autophagy. This study reveals a novel mechanism by which Trim25 restricts RABV replication by destabilizing RABV-P, which is independent of its E3 ubiquitin ligase activity.
Collapse
Affiliation(s)
- Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - An Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zongmei Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bin Tian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yuan Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Baokun Sui
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhaochen Luo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yingying Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| |
Collapse
|
18
|
Jiang H, Kan X, Ding C, Sun Y. The Multi-Faceted Role of Autophagy During Animal Virus Infection. Front Cell Infect Microbiol 2022; 12:858953. [PMID: 35402295 PMCID: PMC8990858 DOI: 10.3389/fcimb.2022.858953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/01/2022] [Indexed: 01/17/2023] Open
Abstract
Autophagy is a process of degradation to maintain cellular homeostatic by lysosomes, which ensures cellular survival under various stress conditions, including nutrient deficiency, hypoxia, high temperature, and pathogenic infection. Xenophagy, a form of selective autophagy, serves as a defense mechanism against multiple intracellular pathogen types, such as viruses, bacteria, and parasites. Recent years have seen a growing list of animal viruses with autophagy machinery. Although the relationship between autophagy and human viruses has been widely summarized, little attention has been paid to the role of this cellular function in the veterinary field, especially today, with the growth of serious zoonotic diseases. The mechanisms of the same virus inducing autophagy in different species, or different viruses inducing autophagy in the same species have not been clarified. In this review, we examine the role of autophagy in important animal viral infectious diseases and discuss the regulation mechanisms of different animal viruses to provide a potential theoretical basis for therapeutic strategies, such as targets of new vaccine development or drugs, to improve industrial production in farming.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai, China
| | - Xianjin Kan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai, China
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- *Correspondence: Yingjie Sun, ; Chan Ding,
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai, China
- *Correspondence: Yingjie Sun, ; Chan Ding,
| |
Collapse
|
19
|
Zhang Q, Cao S, Qiu F, Kang N. Incomplete autophagy: Trouble is a friend. Med Res Rev 2022; 42:1545-1587. [PMID: 35275411 DOI: 10.1002/med.21884] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/26/2022] [Accepted: 02/22/2022] [Indexed: 01/18/2023]
Abstract
Incomplete autophagy is an impaired self-eating process of intracellular macromolecules and organelles in which accumulated autophagosomes do not fuse with lysosomes for degradation, resulting in the blockage of autophagic flux. In this review, we summarized the literature over the past decade describing incomplete autophagy, and found that different from the double-edged sword effect of general autophagy on promoting cell survival or death, incomplete autophagy plays a crucial role in disrupting cellular homeostasis, and promotes only cell death. What matters is that incomplete autophagy is closely relevant to the pathogenesis and progression of various human diseases, which, meanwhile, intimately linking to the pharmacologic and toxicologic effects of several compounds. Here, we comprehensively reviewed the latest progress of incomplete autophagy on molecular mechanisms and signaling pathways. Moreover, implications of incomplete autophagy for pharmacotherapy are also discussed, which has great relevance for our understanding of the distinctive role of incomplete autophagy in cellular physiology and disease. Consequently, targeting incomplete autophagy may contribute to the development of novel generation therapeutic agents for diverse human diseases.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Shijie Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,Department of Medicinal Chemistry, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ning Kang
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
20
|
Harsha PK, Ranganayaki S, Yale G, Dey G, Mangalaparthi KK, Yarlagadda A, Chandrasekhar Sagar BK, Mahadevan A, Srinivas Bharath MM, Mani RS. Mitochondrial Dysfunction in Rabies Virus-Infected Human and Canine Brains. Neurochem Res 2022; 47:1610-1636. [PMID: 35229271 DOI: 10.1007/s11064-022-03556-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 11/26/2022]
Abstract
Rabies is a fatal encephalitis caused by the Rabies lyssavirus (RABV). The presence of minimal neuropathological changes observed in rabies indicates that neuronal dysfunction, rather than neuronal death contributes to the fatal outcome. The role of mitochondrial changes has been suggested as a possible mechanism for neuronal dysfunction in rabies. However, these findings are mostly based on studies that have employed experimental models and laboratory-adapted virus. Studies on brain tissues from naturally infected human and animal hosts are lacking. The current study investigated the role of mitochondrial changes in rabies by morphological, biochemical and proteomic analysis of RABV-infected human and canine brains. Morphological analysis showed minimal inflammation with preserved neuronal and disrupted mitochondrial structure in both human and canine brains. Proteomic analysis revealed involvement of mitochondrial processes (oxidative phosphorylation, cristae formation, homeostasis and transport), synaptic proteins and autophagic pathways, with over-expression of subunits of mitochondrial respiratory complexes. Consistent with these findings, human and canine brains displayed elevated activities of complexes I (p < 0.05), IV (p < 0.05) and V (p < 0.05). However, this did not result in elevated ATP production (p < 0.0001), probably due to lowered mitochondrial membrane potential as noted in RABV-infected cells in culture. These could lead to mitochondrial dysfunction and mitophagy as indicated by expression of FKBP8 (p < 0.05) and PINK1 (p < 0.001)/PARKIN (p > 0.05) and ensuing autophagy, as shown by the status of LCIII (p < 0.05), LAMP1 (p < 0.001) and pertinent ultrastructural markers. We propose that altered mitochondrial bioenergetics and cristae architecture probably induce mitophagy, leading to autophagy and consequent neuronal dysfunction in rabies.
Collapse
Affiliation(s)
- Pulleri Kandi Harsha
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Sathyanarayanan Ranganayaki
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | | | - Gourav Dey
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | | | - Anusha Yarlagadda
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - B K Chandrasekhar Sagar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.
| | - Reeta S Mani
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.
| |
Collapse
|
21
|
Ozkaraca M, Ozdemir S, Comakli S, Timurkan MO. Roles of apoptosis and autophagy in natural rabies infections. VET MED-CZECH 2022; 67:1-12. [PMID: 39169958 PMCID: PMC11334964 DOI: 10.17221/221/2020-vetmed] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 08/24/2021] [Indexed: 08/23/2024] Open
Abstract
The aim of this study was to investigate the activity of apoptosis and autophagy in animals (cows, horses, donkeys, dogs and cats) naturally infected with rabies by using immunohistochemistry, immunofluorescence, and qPCR. The mRNA transcript levels of caspase-3, Bax, Bcl2 and LC3B were determined with qPCR. Caspase-3 and AIF immunopositivity were not observed in the immunohistochemical and immunofluorescence staining, whereas LC3B immunopositivity was determined intensively in the infected animals compared to the control groups. LC3B immunopositivity was detected in the cytoplasm of the Purkinje cells in the cerebellum of the cows, horses and donkeys, and also in the cytoplasm of the neurons in the cornu ammonis of the dogs and cats. While the expression levels of caspase-3 and Bax were downregulated, the Bcl2 expression was up-regulated in the infected animals compared to the uninfected animals. In addition, the LC3B levels were found to be significantly higher in the infected animals. To the best of our knowledge, this work represents the first report of neuronal death in the central nervous system by autophagy, rather than by caspase-dependent or AIF-containing caspase-independent apoptosis.
Collapse
Affiliation(s)
- Mustafa Ozkaraca
- Department of Pathology, Faculty of Veterinary Medicine, Cumhuriyet University, Sivas, Turkey
| | - Selcuk Ozdemir
- Department of Genetic, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Selim Comakli
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Mehmet Ozkan Timurkan
- Department of Virology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
22
|
The role of dancing duplexes in biology and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021. [PMID: 34656330 DOI: 10.1016/bs.pmbts.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Across species, a common protein assembly arises: proteins containing structured domains separated by long intrinsically disordered regions, and dimerized through a self-association domain or through strong protein interactions. These systems are termed "IDP duplexes." These flexible dimers have roles in diverse pathologies including development of cancer, viral infections, and neurodegenerative disease. Here we discuss the role of disorder in IDP duplexes with similar domain architectures that bind hub protein, LC8. LC8-binding IDP duplexes are categorized into three groups: IDP duplexes that contain a self-association domain that is extended by LC8 binding, IDP duplexes that have no self-association domain and are dimerized through binding several copies of LC8, and multivalent LC8-binders that also have a self-association domain. Additionally, we discuss non-LC8-binding IDP duplexes with similar domain organizations, including the Nucleocapsid protein of SARS-CoV-2. We propose that IDP duplexes have structural features that are essential in many biological processes and that improved understanding of their structure function relationship will provide new therapeutic opportunities.
Collapse
|
23
|
Abstract
Viruses are obligatory intracellular parasites that use cell proteins to take the control of the cell functions in order to accomplish their life cycle. Studying the viral-host interactions would increase our knowledge of the viral biology and mechanisms of pathogenesis. Studies on pathogenesis mechanisms of lyssaviruses, which are the causative agents of rabies, have revealed some important host protein partners for viral proteins, especially for most studied species, i.e. RABV. In this review article, the key physical lyssavirus-host protein interactions, their contributions to rabies infection, and their exploitation are discussed to improve the knowledge about rabies pathogenesis.
Collapse
|
24
|
Zhao W, Su J, Wang N, Zhao N, Su S. Expression Profiling and Bioinformatics Analysis of CircRNA in Mice Brain Infected with Rabies Virus. Int J Mol Sci 2021; 22:ijms22126537. [PMID: 34207166 PMCID: PMC8234020 DOI: 10.3390/ijms22126537] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/22/2022] Open
Abstract
Rabies virus (RABV) induces acute, fatal encephalitis in mammals including humans. The circRNAs are important in virus infection process, but whether circRNAs regulated RABV infection remains largely unknown. Here, mice brain with or without the RABV CVS-11 strain were subjected to RNA sequencing and a total of 30,985 circRNAs were obtained. Among these, 9021 candidates were shared in both groups, and 14,610 and 7354 circRNAs were expressed specifically to the control and experimental groups, indicating that certain circRNAs were specifically inhibited or induced on RABV infection. The circRNAs mainly derived from coding exons. In total, 636 circRNAs were differentially expressed in RABV infection, of which 426 significantly upregulated and 210 significantly downregulated (p < 0.05 and fold change ≥2). The expression of randomly selected 6 upregulated and 6 downregulated circRNAs was tested by RT-qPCR, and the expression trend of the 11 out of 12 circRNAs was consistent in RT- qPCR and RNA-seq analysis. Rnase R-resistant assay and Sanger sequencing were conducted to verify the circularity of circRNAs. GO analysis demonstrated that source genes of all differentially regulated circRNAs were mainly related to cell plasticity and synapse function. Both KEGG and GSEA analysis revealed that these source genes were engaged in the cGMP–PKG and MAPK signaling pathway, and HTLV-I infection. Also, pathways related to glucose metabolism and synaptic functions were enriched in KEGG analysis. The circRNA–miRNA–mRNA network was built with 25 of 636 differentially expressed circRNAs, 264 mRNAs involved in RABV infection, and 29 miRNAs. Several miRNAs and many mRNAs in the network were reported to be related to viral infection and the immune response, suggesting that circRNAs could regulate RABV infection via interacting with miRNAs and mRNAs. Taken together, this study first characterized the transcriptomic pattern of circRNAs, and signaling pathways and function that circRNAs are involved in, which may indicate directions for further research to understand mechanisms of RABV pathogenesis.
Collapse
|
25
|
Geng SC, Li XL, Fang WH. Porcine circovirus 3 capsid protein induces autophagy in HEK293T cells by inhibiting phosphorylation of the mammalian target of rapamycin. J Zhejiang Univ Sci B 2021; 21:560-570. [PMID: 32633110 DOI: 10.1631/jzus.b1900657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Porcine circovirus 3 (PCV3) has been detected in major pig-producing countries around the world since its first report in the US in 2016. Most current studies have focused on epidemiological investigations and detection methods of PCV3 because of lack of live virus strains for research on its pathogenesis in porcine cells or even in pigs. We constructed a recombinant plasmid pCMV-Cap carrying the PCV3 orf2 gene to investigate the effects of capsid (Cap) protein expression on autophagic response in human embryonic kidney cell line 293T (HEK293T). We demonstrate that PCV3 Cap protein induced complete autophagy shown as formation of autophagosomes and autophagosome-like vesicles as well as LC3-II conversion from LC3-I via inhibiting phosphorylation of the mammalian target of rapamycin (mTOR) in HEK293T cells. The ubiquitin-proteasome pathway is also involved in the autophagy process. These findings provide insight for further exploration of PCV3 pathogenetic mechanisms in porcine cells.
Collapse
Affiliation(s)
- Shi-Chao Geng
- Institute of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China.,Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou 310058, China
| | - Xiao-Liang Li
- Institute of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China.,Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou 310058, China
| | - Wei-Huan Fang
- Institute of Preventive Veterinary Medicine, Zhejiang University, Hangzhou 310058, China.,Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Hangzhou 310058, China
| |
Collapse
|
26
|
Wang Y, He H, Li J, Chen L, Luo J, Kuang Y, Lv Z, Fan R, Zhang B, Luo Y, Guo X. Rabies Virus-Induced Autophagy Is Dependent on Viral Load in BV2 Cells. Front Microbiol 2021; 12:595678. [PMID: 34113320 PMCID: PMC8186530 DOI: 10.3389/fmicb.2021.595678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
An increasing number of studies are showing that autophagy plays a vital role in viral replication and escape. Rabies virus (RABV), a typical neurotropic virus, has been proven to induce autophagy in neurons. However, there are no reports indicating that RABV can cause autophagy in other cells of the central nervous system. Thus, we aimed to explore the relationship between autophagy and RABV infection in BV2 cells in this study. Results of viral growth curves showed that the titers of microglial BV2 cells infected with RABV peaked at 12 hours post-infection (hpi) and then decreased continuously over time. However, it was found that the viral genome RNA and structural proteins can express normally in BV2 cells. In addition, Western blotting indicated that RABV infection increased LC3-II and p62 expression in BV2 cells. LC3 punctate increased with RABV infection in BV2 cells after the transfection of fluorescent protein-tagged LC3 plasmids. Moreover, autophagy cargo protein further accumulated with RABV infection in Bafilomycin A1-treated cells. Subsequently, RABV infection inhibited the fusion of autophagosomes with lysosomes by using a tandem fluorescent marker. Furthermore, a higher multiplicity of infection induced stronger autophagy. Thus, RABV can induce autophagy in BV2 cells, and the autophagy is positively associated with the viral load.
Collapse
Affiliation(s)
- Yang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongling He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiesen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Luman Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yanqi Kuang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ziyu Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ruqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
27
|
Bhutta MS, Gallo ES, Borenstein R. Multifaceted Role of AMPK in Viral Infections. Cells 2021; 10:1118. [PMID: 34066434 PMCID: PMC8148118 DOI: 10.3390/cells10051118] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023] Open
Abstract
Viral pathogens often exploit host cell regulatory and signaling pathways to ensure an optimal environment for growth and survival. Several studies have suggested that 5'-adenosine monophosphate-activated protein kinase (AMPK), an intracellular serine/threonine kinase, plays a significant role in the modulation of infection. Traditionally, AMPK is a key energy regulator of cell growth and proliferation, host autophagy, stress responses, metabolic reprogramming, mitochondrial homeostasis, fatty acid β-oxidation and host immune function. In this review, we highlight the modulation of host AMPK by various viruses under physiological conditions. These intracellular pathogens trigger metabolic changes altering AMPK signaling activity that then facilitates or inhibits viral replication. Considering the COVID-19 pandemic, understanding the regulation of AMPK signaling following infection can shed light on the development of more effective therapeutic strategies against viral infectious diseases.
Collapse
Affiliation(s)
- Maimoona Shahid Bhutta
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
| | - Elisa S. Gallo
- Board-Certified Dermatologist and Independent Researcher, Norfolk, VA 23507, USA;
| | - Ronen Borenstein
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
| |
Collapse
|
28
|
Park G, Lee JY, Han HM, An HS, Jin Z, Jeong EA, Kim KE, Shin HJ, Lee J, Kang D, Kim HJ, Bae YC, Roh GS. Ablation of dynamin-related protein 1 promotes diabetes-induced synaptic injury in the hippocampus. Cell Death Dis 2021; 12:445. [PMID: 33953167 PMCID: PMC8099876 DOI: 10.1038/s41419-021-03723-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/03/2023]
Abstract
Dynamin-related protein 1 (Drp1)-mediated mitochondrial dysfunction is associated with synaptic injury in the diabetic brain. However, the dysfunctional mitochondria by Drp1 deletion in the diabetic brain are poorly understood. Here, we investigated the effects of neuron-specific Drp1 deletion on synaptic damage and mitophagy in the hippocampus of a high-fat diet (HFD)/streptozotocin (STZ)-induced diabetic mice. HFD/STZ-induced diabetic mice exhibited metabolic disturbances and synaptic damages. Floxed Drp1 mice were crossed with Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα)-Cre mice, to generate neuron-specific Drp1 knockout (Drp1cKO) mice, which showed marked mitochondrial swelling and dendritic spine loss in hippocampal neurons. In particular, diabetic Drp1cKO mice exhibited an increase in dendritic spine loss and higher levels of oxidative stress and neuroinflammation compared with diabetic wild-type (WT) mice. Diabetic WT mice generally displayed increased Drp1-induced small mitochondrial morphology in hippocampal neurons, but large mitochondria were prominently observed in diabetic Drp1cKO mice. The levels of microtubule-associated protein 1 light-chain 3 and lysosomal-associated membrane protein 1 proteins were significantly increased in the hippocampus of diabetic Drp1cKO mice compared with diabetic WT mice. The inhibition of Drp1 adversely promotes synaptic injury and neurodegeneration in the diabetic brain. The findings suggest that the exploratory mechanisms behind Drp1-mediated mitochondrial dysfunction could provide a possible therapeutic target for diabetic brain complications.
Collapse
Affiliation(s)
- Gyeongah Park
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.267301.10000 0004 0386 9246Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Jong Youl Lee
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Hye Min Han
- grid.258803.40000 0001 0661 1556Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41944 South Korea
| | - Hyeong Seok An
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Zhen Jin
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.267301.10000 0004 0386 9246Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163 USA
| | - Eun Ae Jeong
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Kyung Eun Kim
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Hyun Joo Shin
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Jaewoong Lee
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Dawon Kang
- grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Department of Physiology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Hyun Joon Kim
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| | - Yong Chul Bae
- grid.258803.40000 0001 0661 1556Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41944 South Korea
| | - Gu Seob Roh
- grid.256681.e0000 0001 0661 1492Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea ,grid.256681.e0000 0001 0661 1492Bio Anti-Aging Medical Research Center, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam 52727 Republic of Korea
| |
Collapse
|
29
|
Rabies: Presentation, case management and therapy. J Neurol Sci 2021; 424:117413. [PMID: 33812240 DOI: 10.1016/j.jns.2021.117413] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/26/2021] [Accepted: 03/22/2021] [Indexed: 11/23/2022]
Abstract
Several Lyssaviruses are known to be a causative agent of rabies and rabies like syndrome. There are no proven effective treatment strategies for symptomatic rabies patient. Risk of infection from dog variant of rabies virus is highest with deep bite reaching muscular layer and much higher when compared to scratch. Failure of viral eradication at the central nervous system (CNS) is partly due to inadequate immune response. Favipiravir selectively inhibit viral RNA polymerase and has been shown to reduce rabies replication in neuronal cell and mouse model system. Endocannabinoid system has emerged as an important regulator for CNS integrity, cell fate and may serve as an important novel neuroprotective agent. Cannabinoid may be able to regulate the impaired homeostasis induced by rabies virus by promoting infected cell survival and promote complete autophagy in infected cell.
Collapse
|
30
|
Guo Y, Yang PT, Wang ZW, Xu K, Kou WH, Luo H. Identification of Three Autophagy-Related Long Non-Coding RNAs as a Novel Head and Neck Squamous Cell Carcinoma Prognostic Signature. Front Oncol 2021; 10:603864. [PMID: 33575215 PMCID: PMC7871905 DOI: 10.3389/fonc.2020.603864] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/09/2020] [Indexed: 01/08/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has a poor prognosis. Considerable evidence indicates that autophagy and non-coding RNA play essential roles in the biological processes involved in cancers, but associations between autophagy-related long non-coding RNAs (lncRNAs) and HNSCC remain unclear. In the present study, HNSCC RNA sequences and autophagy-related gene data were extracted from The Cancer Genome Atlas database and the Human Autophagy Database. A total of 1,153 autophagy-related lncRNAs were selected via calculating Pearson’s correlation coefficient. Three prognosis-related autophagy lncRNAs were identified via univariate Cox regression, least absolute shrinkage and selection operator analysis, and multivariate Cox regression analysis. We also constructed a prognostic model based on these autophagy-related lncRNAs and evaluated its ability to accurately and independently predict the prognosis of HNSCC patients. The area under the curve (AUC) was 0.864 (3-year) and 0.836 (5-year), and our model can independently predict the prognosis of HNSCC. The prognostic value of the three autophagy lncRNAs was confirmed via analysis of samples from five databases. To further identify the functions of the three lncRNAs, a co-expression network was constructed and pathway analysis was performed. In that analysis the lncRNAs were correlated with 189 related genes and 20 autophagy-related genes, and these lncRNAs mainly involved homologous recombination, the Fanconi anemia pathway, the autophagy-related pathway, and immune-related pathways. In addition, we validated the expression levels of three lncRNAs and autophagy markers (ATG12, BECN1, and MAP1LC3B) based on TIMER, Oncomine, and HPA database analysis. Our results indicated that TTTY15 was increased in HPV positive and HPV negative HNSCC patients, and three autophagy markers were up-regulated in all HNSCCC patients. Lastly, association between three lncRNAs and autophagy markers was performed, and our results showed that TTTY15 and MIF-AS1 were associated with autophagy markers. Collectively, these results suggested that three autophagy-related lncRNAs have prognostic value in HNSCC patients.
Collapse
Affiliation(s)
- Ya Guo
- Department of Radiation Oncology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Peng Tao Yang
- Department of Radiation Oncology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhong Wei Wang
- Department of Radiation Oncology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kun Xu
- Department of Radiation Oncology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wei Hua Kou
- Department of Radiation Oncology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Heng Luo
- Department of Radiation Oncology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
31
|
Mara K, Dai M, Brice AM, Alexander MR, Tribolet L, Layton DS, Bean AGD. Investigating the Interaction between Negative Strand RNA Viruses and Their Hosts for Enhanced Vaccine Development and Production. Vaccines (Basel) 2021; 9:vaccines9010059. [PMID: 33477334 PMCID: PMC7830660 DOI: 10.3390/vaccines9010059] [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: 12/19/2020] [Accepted: 01/13/2021] [Indexed: 11/30/2022] Open
Abstract
The current pandemic has highlighted the ever-increasing risk of human to human spread of zoonotic pathogens. A number of medically-relevant zoonotic pathogens are negative-strand RNA viruses (NSVs). NSVs are derived from different virus families. Examples like Ebola are known for causing severe symptoms and high mortality rates. Some, like influenza, are known for their ease of person-to-person transmission and lack of pre-existing immunity, enabling rapid spread across many countries around the globe. Containment of outbreaks of NSVs can be difficult owing to their unpredictability and the absence of effective control measures, such as vaccines and antiviral therapeutics. In addition, there remains a lack of essential knowledge of the host–pathogen response that are induced by NSVs, particularly of the immune responses that provide protection. Vaccines are the most effective method for preventing infectious diseases. In fact, in the event of a pandemic, appropriate vaccine design and speed of vaccine supply is the most critical factor in protecting the population, as vaccination is the only sustainable defense. Vaccines need to be safe, efficient, and cost-effective, which is influenced by our understanding of the host–pathogen interface. Additionally, some of the major challenges of vaccines are the establishment of a long-lasting immunity offering cross protection to emerging strains. Although many NSVs are controlled through immunisations, for some, vaccine design has failed or efficacy has proven unreliable. The key behind designing a successful vaccine is understanding the host–pathogen interaction and the host immune response towards NSVs. In this paper, we review the recent research in vaccine design against NSVs and explore the immune responses induced by these viruses. The generation of a robust and integrated approach to development capability and vaccine manufacture can collaboratively support the management of outbreaking NSV disease health risks.
Collapse
|
32
|
Ranganayaki S, Jamshidi N, Aiyaz M, Rashmi SK, Gayathri N, Harsha PK, Padmanabhan B, Srinivas Bharath MM. Inhibition of mitochondrial complex II in neuronal cells triggers unique pathways culminating in autophagy with implications for neurodegeneration. Sci Rep 2021; 11:1483. [PMID: 33452321 PMCID: PMC7810707 DOI: 10.1038/s41598-020-79339-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/23/2020] [Indexed: 02/08/2023] Open
Abstract
Mitochondrial dysfunction and neurodegeneration underlie movement disorders such as Parkinson’s disease, Huntington’s disease and Manganism among others. As a corollary, inhibition of mitochondrial complex I (CI) and complex II (CII) by toxins 1-methyl-4-phenylpyridinium (MPP+) and 3-nitropropionic acid (3-NPA) respectively, induced degenerative changes noted in such neurodegenerative diseases. We aimed to unravel the down-stream pathways associated with CII inhibition and compared with CI inhibition and the Manganese (Mn) neurotoxicity. Genome-wide transcriptomics of N27 neuronal cells exposed to 3-NPA, compared with MPP+ and Mn revealed varied transcriptomic profile. Along with mitochondrial and synaptic pathways, Autophagy was the predominant pathway differentially regulated in the 3-NPA model with implications for neuronal survival. This pathway was unique to 3-NPA, as substantiated by in silico modelling of the three toxins. Morphological and biochemical validation of autophagy markers in the cell model of 3-NPA revealed incomplete autophagy mediated by mechanistic Target of Rapamycin Complex 2 (mTORC2) pathway. Interestingly, Brain Derived Neurotrophic Factor (BDNF), which was elevated in the 3-NPA model could confer neuroprotection against 3-NPA. We propose that, different downstream events are activated upon neurotoxin-dependent CII inhibition compared to other neurotoxins, with implications for movement disorders and regulation of autophagy could potentially offer neuroprotection.
Collapse
Affiliation(s)
- Sathyanarayanan Ranganayaki
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, Karnataka, 560029, India
| | - Neema Jamshidi
- Department of Radiological Sciences, Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095, USA
| | - Mohamad Aiyaz
- Genotypic Technology Pvt. Ltd., 2/13, Balaji Complex, 80 feet Road, RMV 2nd Stage, Bangalore, Karnataka, 560094, India
| | - Santhosh-Kumar Rashmi
- Department of Neuropathology, NIMHANS, No. 2900, Hosur Road, Bangalore, Karnataka, 560029, India
| | - Narayanappa Gayathri
- Department of Neuropathology, NIMHANS, No. 2900, Hosur Road, Bangalore, Karnataka, 560029, India
| | - Pulleri Kandi Harsha
- Department of Neurovirology, NIMHANS, No. 2900, Hosur Road, Bangalore, Karnataka, 560029, India
| | | | - Muchukunte Mukunda Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, Karnataka, 560029, India.
| |
Collapse
|
33
|
Role of autophagy in nerve cell apoptosis in mice infected with street rabies virus. Arch Virol 2020; 165:2857-2867. [PMID: 33034763 DOI: 10.1007/s00705-020-04815-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022]
Abstract
Rabies is an important zoonotic disease in Iran. Autophagy is a process that maintains homeostasis and can be used as an innate defense mechanism against viruses. Apoptosis is the process of programmed cell death induced by physiological and pathological conditions. The crosstalk of autophagy and apoptosis plays a key role in rabies virus infection. In the current study, NMRI mice intra-cranially received 3-Methyl Adenine (3-MA), rapamycin, street rabies virus (SRABV) and drugs plus SRABV. SRABV and Map1lc3, Beclin-1, Atg5 gene expression were assayed by real-time PCR. Immunohistochemistry was carried out via LC3 protein staining as an autophagy marker, and apoptotic cell death was measured using a TUNEL assay. Map1lc3, Beclin-1 and Atg5 genes expression was significantly increased in drug-plus-SRBV-treated tissues compared to control at 24 hpi. Map1lc3 and Atg5 gene expression showed a slight change in the drugs-plus-virus group compared with the control at 72 hpi. The presence of LC3 in the tissues of the group treated with rapamycin plus SRBV confirmed induction of autophagy, but it was not present in the tissues treated with 3-MA plus SRBV. Our data revealed that apoptosis was induced only in the groups receiving the SRBV or rapamycin or both at 24 hpi. Apoptosis was observed after 72 hours, when the drugs' effect had disappeared in all but the autophagy inhibitor group. Understanding the interaction of SRABV with autophagy pathway genes and its effect on host cell apoptosis may open a new horizon for human intervention and allow a deeper understanding of rabies infections.
Collapse
|
34
|
Liu J, Liao M, Yan Y, Yang H, Wang H, Zhou J. Rabies virus phosphoprotein P5 binding to BECN1 regulates self-replication by BECN1-mediated autophagy signaling pathway. Cell Commun Signal 2020; 18:153. [PMID: 32948206 PMCID: PMC7499888 DOI: 10.1186/s12964-020-00644-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/10/2020] [Indexed: 12/25/2022] Open
Abstract
Background Rabies virus (RABV) is reported to encode five phosphoproteins (P), which are involved in viral genomic replication, axonal transport, oxidative stress, interferon antagonism, and autophagy induction. However, the functions of the different P proteins are poorly understood. Methods Immunofluorescence staining and western blot were performed to detect the autophagy activity, the form of ring-like structure, and the colocalization of BECN1 and P. Co-immunoprecipitation was performed to detect the interaction between P and BECN1. QRT-PCR and TCID50 assay were performed to detect the replication level of RABV. Small interfering RNA was used to detect the autophagy signaling pathway. Results We found that P5 attaches to N-terminal residues 1–139 of BECN1 (beclin1) on the BECN1 ring-like structure through amino acid residues 173–222 of P5. Subsequently, we found that P5-induced autophagosomes did not fuse with lysosomes. Becn1 silencing did not recover P5 overexpression-induced promotion of RABV replication. Mechanistically, RABV protein PΔN82 (P5) induced incomplete autophagy via the BECN1-mediated signaling pathway. Conclusions Our data indicate that P5 binding to the BECN1 ring benefits RABV replication by inducing BECN1 signaling pathway-dependent incomplete autophagy, which provides a potential target for antiviral drugs against RABV. Video abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Juan Liu
- MOA Key Laboratory of Animal Virology, Center for Veterinary Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Min Liao
- MOA Key Laboratory of Animal Virology, Center for Veterinary Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Yan Yan
- MOA Key Laboratory of Animal Virology, Center for Veterinary Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Hui Yang
- MOA Key Laboratory of Animal Virology, Center for Veterinary Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Hailong Wang
- MOA Key Laboratory of Animal Virology, Center for Veterinary Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Center for Veterinary Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.,Collaborative innovation center and State Key laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310058, PR China
| |
Collapse
|
35
|
Luo J, Zhang Y, Wang Y, Liu Q, Chen L, Zhang B, Luo Y, Huang S, Guo X. Rhabdovirus Infection Is Dependent on Serine/Threonine Kinase AP2-Associated Kinase 1. Life (Basel) 2020; 10:E170. [PMID: 32872567 PMCID: PMC7554979 DOI: 10.3390/life10090170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 01/01/2023] Open
Abstract
Rabies virus (RABV) causes a fatal neurological disease in both humans and animals. Understanding the mechanism of RABV infection is vital for prevention and therapy of virulent rabies infection. Our previous proteomics analysis based on isobaric tags for relative and absolute quantitation to identify factors revealed that RABV infection enhanced AP-2-associated protein kinase 1 (AAK1) in N2a cells. In this study, to further confirm the role of AAK1, we showed that RABV infection increased the transcription and expression of AAK1 in N2a cells. AAK1 knockdown significantly decreased RABV infection in both N2a and BHK-21 cells. AAK1 knockout inhibited RABV infection in N2a cells. Furthermore, inhibition of AAK1 kinase activity using sunitinib decreased RABV infection. However, AAK1 overexpression did not change RABV infection in vitro. Therapeutic administration of sunitinib did not significantly improve the survival rate of mice following lethal RABV challenge. In addition, AAK1 knockdown decreased infection in N2a cells by vesicular stomatitis virus, which is another rhabdovirus. These results indicate that rhabdovirus infection is dependent on AAK1 and inhibition of AAK1 is a potential strategy for the prevention and therapy of rabies.
Collapse
Affiliation(s)
- Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Qing Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Luman Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| |
Collapse
|
36
|
Blanco J, Cameirao C, López MC, Muñoz-Barroso I. Phosphatidylinositol-3-kinase-Akt pathway in negative-stranded RNA virus infection: a minireview. Arch Virol 2020; 165:2165-2176. [PMID: 32740830 DOI: 10.1007/s00705-020-04740-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/17/2020] [Indexed: 12/23/2022]
Abstract
The PI3K/Akt signalling pathway is a crucial signalling cascade that regulates transcription, protein translation, cell growth, proliferation, cell survival, and metabolism. During viral infection, viruses exploit a variety of cellular pathways, including the well-known PI3K/Akt signalling pathway. Conversely, cells rely on this pathway to stimulate an antiviral response. The PI3K/Akt pathway is manipulated by a number of viruses, including DNA and RNA viruses and retroviruses. The aim of this review is to provide up-to-date information about the role of the PI3K-Akt pathway in infection with members of five different families of negative-sense ssRNA viruses. This pathway is hijacked for viral entry, regulation of endocytosis, suppression of premature apoptosis, viral protein expression, and replication. Although less common, the PI3K/Akt pathway can be downregulated as an immunomodulatory strategy or as a mechanism for inducing autophagy. Moreover, the cell activates this pathway as an antiviral strategy for interferon and cytokine production, among other strategies. Here, we present new data concerning the role of this pathway in infection with the paramyxovirus Newcastle disease virus (NDV). Our data seem to indicate that NDV uses the PI3K/Akt pathway to delay cell death and increase cell survival as a means of improving its replication. The interference of negative-sense ssRNA viruses with this essential pathway might have implications for the development of antiviral therapies.
Collapse
Affiliation(s)
- Javier Blanco
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain
| | - Cristina Cameirao
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253, Bragança, Portugal
| | - María Carmen López
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.
| |
Collapse
|
37
|
Wang S, Ren X, Li J, Lin C, Zhou J, Zhou J, Gu J. NAP1L4 inhibits porcine circovirus type 2 replication via IFN-β signaling pathway. Vet Microbiol 2020; 246:108692. [PMID: 32605741 DOI: 10.1016/j.vetmic.2020.108692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/30/2022]
Abstract
Porcine circovirus type 2 (PCV2) capsid protein (Cap) was previously reported to interact with nucleosome assembly protein 1-like 4 (NAP1L4). The biological function of Cap-NAP1L4 interaction is unknown. Here, we demonstrated that PCV2 Cap could directly interact with NAP1L4, which the amino acid residues 124-279 of NAP1L4 were responsible for the interaction. Furthermore, over-expression of NAP1L4 down-regulated the expression of PCV2 Cap and Rep. DNA copies and virus titers were also decreased in NAP1L4 over-expressed PK15 cells. While, knockout of NAP1L4 through CRISPR/Cas9 technology in PK15 cells could up-regulate the mRNA and protein levels of PCV2 Cap and Rep. PCV2 genomic DNA copies and virus titers were also increased in NAP1L4-knockdown/-knockout PK15 cells compared with wild type PK15 cells. In addition, NAP1L4 depletion was demonstrated to facilitate cytosolic carboxypeptidase-like protein 5 (CCP5) and cytosolic carboxypeptidase 6 (CCP6) expression, which could activate cGAS to promote IFN-β production. Indeed, knockout of NAP1L4 was also confirmed to increase IFN-β expression. And IFN-β stimulation could promote PCV2 replication in PK15 cells. Taken together, our findings suggest that NAP1L4 interacts with PCV2 Cap and inhibits PCV2 replication through regulating IFN-β production. Our study provides theoretical basis for further study of PCV2.
Collapse
Affiliation(s)
- Shengnan Wang
- Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; MOA Key Laboratory of Animal Virology, Institute of Preventive Veterinary Science, Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Xuqian Ren
- Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiarong Li
- MOA Key Laboratory of Animal Virology, Institute of Preventive Veterinary Science, Department of Veterinary Medicine, Zhejiang University, Hangzhou, China; Veterianry Medical Research Center, Zhejiang University, Hangzhou, China
| | - Cui Lin
- MOA Key Laboratory of Animal Virology, Institute of Preventive Veterinary Science, Department of Veterinary Medicine, Zhejiang University, Hangzhou, China; Veterianry Medical Research Center, Zhejiang University, Hangzhou, China
| | - Jianwei Zhou
- MOA Key Laboratory of Animal Virology, Institute of Preventive Veterinary Science, Department of Veterinary Medicine, Zhejiang University, Hangzhou, China; Veterianry Medical Research Center, Zhejiang University, Hangzhou, China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Institute of Preventive Veterinary Science, Department of Veterinary Medicine, Zhejiang University, Hangzhou, China; Veterianry Medical Research Center, Zhejiang University, Hangzhou, China.
| | - Jinyan Gu
- Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
38
|
Kaur S, Changotra H. The beclin 1 interactome: Modification and roles in the pathology of autophagy-related disorders. Biochimie 2020; 175:34-49. [PMID: 32428566 DOI: 10.1016/j.biochi.2020.04.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/18/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022]
Abstract
Beclin 1 a yeast Atg6/VPS30 orthologue has a significant role in autophagy process (Macroautophagy) and protein sorting. The function of beclin 1 depends on the interaction with several autophagy-related genes (Atgs) and other proteins during the autophagy process. The role mediated by beclin 1 is controlled by various conditions and factors. Beclin 1 is regulated at the gene and protein levels by different factors. These regulations could subsequently alter the beclin 1 induced autophagy process. Therefore, it is important to study the components of beclin 1 interactome and factors affecting its expression. Expression of this gene is differentially regulated under different conditions in different cells or tissues. So, the regulation part is important to study as beclin 1 is one of the candidate genes involved in diseases related to autophagy dysfunction. This review focuses on the functions of beclin 1, its interacting partners, regulations at gene and protein level, and the role of beclin 1 interactome in relation to various diseases along with the recent developments in the field.
Collapse
Affiliation(s)
- Sargeet Kaur
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Solan, 173 234, Himachal Pradesh, India
| | - Harish Changotra
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Solan, 173 234, Himachal Pradesh, India.
| |
Collapse
|
39
|
Bello-Perez M, Pereiro P, Coll J, Novoa B, Perez L, Falco A. Zebrafish C-reactive protein isoforms inhibit SVCV replication by blocking autophagy through interactions with cell membrane cholesterol. Sci Rep 2020; 10:566. [PMID: 31953490 PMCID: PMC6969114 DOI: 10.1038/s41598-020-57501-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/31/2019] [Indexed: 02/06/2023] Open
Abstract
In the present work, the mechanisms involved in the recently reported antiviral activity of zebrafish C-reactive protein-like protein (CRP1-7) against the spring viraemia of carp rhabdovirus (SVCV) in fish are explored. The results neither indicate blocking of the attachment or the binding step of the viral replication cycle nor suggest the direct inhibition of G protein fusion activity or the stimulation of the host’s interferon system. However, an antiviral state in the host is induced. Further results showed that the antiviral protection conferred by CRP1-7 was mainly due to the inhibition of autophagic processes. Thus, given the high affinity of CRPs for cholesterol and the recently described influence of the cholesterol balance in lipid rafts on autophagy, both methyl-β-cyclodextrin (a cholesterol-complexing agent) and 25-hydroxycholesterol (a cholesterol molecule with antiviral properties) were used to further describe CRP activity. All the tested compounds exerted antiviral activity by affecting autophagy in a similar manner. Further assays indicate that CRP reduces autophagy activity by initially disturbing the cholesterol ratios in the host cellular membranes, which in turn negatively affects the intracellular regulation of reactive oxygen species (ROS) and increases lysosomal pH as a consequence. Ultimately, here we propose that such pH changes exert an inhibitory direct effect on SVCV replication by disrupting the pH-dependent membrane-fusogenic ability of the viral glycoprotein G, which allows the release of the virus from endosomes into cytoplasm during its entry phase.
Collapse
Affiliation(s)
- Melissa Bello-Perez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Miguel Hernández University (UMH), Elche, 03202, Spain
| | - Patricia Pereiro
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, 36208, Spain
| | - Julio Coll
- Instituto Nacional de Investigaciones y Tecnologías Agrarias y Alimentarias (INIA), Dpto. Biotecnología, Madrid, 28040, Spain
| | - Beatriz Novoa
- Instituto de Investigaciones Marinas (IIM), Consejo Superior de Investigaciones Científicas (CSIC), Vigo, 36208, Spain
| | - Luis Perez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Miguel Hernández University (UMH), Elche, 03202, Spain.
| | - Alberto Falco
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Miguel Hernández University (UMH), Elche, 03202, Spain.
| |
Collapse
|
40
|
Jespersen NE, Leyrat C, Gérard FC, Bourhis JM, Blondel D, Jamin M, Barbar E. The LC8-RavP ensemble Structure Evinces A Role for LC8 in Regulating Lyssavirus Polymerase Functionality. J Mol Biol 2019; 431:4959-4977. [PMID: 31634467 PMCID: PMC7060403 DOI: 10.1016/j.jmb.2019.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 12/25/2022]
Abstract
The rabies and Ebola viruses recruit the highly conserved host protein LC8 for their own reproductive success. In vivo knockouts of the LC8 recognition motif within the rabies virus phosphoprotein (RavP) result in completely nonlethal viral infections. In this work, we examine the molecular role LC8 plays in viral lethality. We show that RavP and LC8 colocalize in rabies infected cells, and that LC8 interactions are essential for efficient viral polymerase functionality. NMR, SAXS, and molecular modeling demonstrate that LC8 binding to a disordered linker adjacent to an endogenous dimerization domain results in restrictions in RavP domain orientations. The resulting ensemble structure of RavP-LC8 tetrameric complex is similar to that of a related virus phosphoprotein that does not bind LC8, suggesting that with RavP, LC8 binding acts as a switch to induce a more active conformation. The high conservation of the LC8 motif in Lyssavirus phosphoproteins and its presence in other analogous proteins such as the Ebola virus VP35 evinces a broader purpose for LC8 in regulating downstream phosphoprotein functions vital for viral replication.
Collapse
Affiliation(s)
- Nathan E Jespersen
- From the Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Cedric Leyrat
- Institut de Génomique Fonctionnelle, CNRS UMR-5203 INSERM U1191, University of Montpellier, Montpellier, France
| | - Francine C Gérard
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000, Grenoble, France
| | - Jean-Marie Bourhis
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000, Grenoble, France
| | - Danielle Blondel
- Institut de Biologie Intégrative de La Cellule (I2BC), CEA, CNRS, Université Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette Cedex, France
| | - Marc Jamin
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000, Grenoble, France
| | - Elisar Barbar
- From the Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
41
|
Li Y, Zhou X, Zhang Y, Yang J, Xu Y, Zhao Y, Wang X. CUL4B regulates autophagy via JNK signaling in diffuse large B-cell lymphoma. Cell Cycle 2019; 18:379-394. [PMID: 30612524 DOI: 10.1080/15384101.2018.1560718] [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: 01/09/2023] Open
Abstract
Aberrant expression of CUL4B was identified in various types of solid cancers. Cumulative evidences support the oncogenic role of CUL4B in cancers, including regulation of cell proliferation and signal transduction. However, its clinical value and potential pathogenic mechanism in diffuse large B-cell lymphoma (DLBCL) have not been described previously. Therefore, we hypothesize that overexpressed CUL4B may contribute to the pathogenesis of DLBCL. The aim of this study is to assess the expression and the biological function of CUL4B in DLBCL progression. In our study, CUL4B overexpression was observed in DLBCL tissues, and its upregulation was closely associated with poor prognosis in patients. Furthermore, the functional roles of CUL4B was detected both in vitro and in vivo. We demonstrated that silencing CUL4B could not only induce cell proliferation inhibition, cell cycle arrest, and motility attenuation of DLBCL cells in vitro, but also decrease tumor growth in DLBCL xenografts mice. In addition, we identified that CUL4B may act as a potent inductor of JNK phosphorylation in regulation of autophagy. Our findings demonstrated a significant role of CUL4B in the development and progression of DLBCL. CUL4B may act as a useful biomarker and a novel therapeutic target in DLBCL.
Collapse
Affiliation(s)
- Ying Li
- a Department of Hematology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China
| | - Xiangxiang Zhou
- a Department of Hematology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China
| | - Ya Zhang
- a Department of Hematology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China
| | - Juan Yang
- a Department of Hematology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China
| | - Yangyang Xu
- a Department of Hematology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China
| | - Yi Zhao
- a Department of Hematology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China
| | - Xin Wang
- a Department of Hematology , Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , People's Republic of China.,b School of Medicine , Shandong University , Jinan , Shandong , People's Republic of China
| |
Collapse
|
42
|
Lu ZS, Chen QS, Zheng QX, Shen JJ, Luo ZP, Fan K, Xu SH, Shen Q, Liu PP. Proteomic and Phosphoproteomic Analysis in Tobacco Mosaic Virus-Infected Tobacco (Nicotiana tabacum). Biomolecules 2019; 9:E39. [PMID: 30678100 PMCID: PMC6406717 DOI: 10.3390/biom9020039] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Tobacco mosaic virus (TMV) is a common source of biological stress that significantly affects plant growth and development. It is also useful as a model in studies designed to clarify the mechanisms involved in plant viral disease. Plant responses to abiotic stress were recently reported to be regulated by complex mechanisms at the post-translational modification (PTM) level. Protein phosphorylation is one of the most widespread and major PTMs in organisms. Using immobilized metal ion affinity chromatography (IMAC) enrichment, high-pH C18 chromatography fraction, and high-accuracy mass spectrometry (MS), a set of proteins and phosphopeptides in both TMV-infected tobacco and control tobacco were identified. A total of 4905 proteins and 3998 phosphopeptides with 3063 phosphorylation sites were identified. These 3998 phosphopeptides were assigned to 1311 phosphoproteins, as some proteins carried multiple phosphorylation sites. Among them, 530 proteins and 337 phosphopeptides corresponding to 277 phosphoproteins differed between the two groups. There were 43 upregulated phosphoproteins, including phosphoglycerate kinase, pyruvate phosphate dikinase, protein phosphatase 2C, and serine/threonine protein kinase. To the best of our knowledge, this is the first phosphoproteomic analysis of leaves from a tobacco cultivar, K326. The results of this study advance our understanding of tobacco development and TMV action at the protein phosphorylation level.
Collapse
Affiliation(s)
- Zi-Shu Lu
- Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou 450000, China.
| | - Qian-Si Chen
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450000, China.
| | - Qing-Xia Zheng
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450000, China.
| | - Juan-Juan Shen
- Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou 450000, China.
| | - Zhao-Peng Luo
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450000, China.
| | - Kai Fan
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450000, China.
| | - Sheng-Hao Xu
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Qi Shen
- Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou 450000, China.
| | - Ping-Ping Liu
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450000, China.
| |
Collapse
|
43
|
Silwal P, Kim JK, Yuk JM, Jo EK. AMP-Activated Protein Kinase and Host Defense against Infection. Int J Mol Sci 2018; 19:ijms19113495. [PMID: 30404221 PMCID: PMC6274990 DOI: 10.3390/ijms19113495] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023] Open
Abstract
5′-AMP-activated protein kinase (AMPK) plays diverse roles in various physiological and pathological conditions. AMPK is involved in energy metabolism, which is perturbed by infectious stimuli. Indeed, various pathogens modulate AMPK activity, which affects host defenses against infection. In some viral infections, including hepatitis B and C viral infections, AMPK activation is beneficial, but in others such as dengue virus, Ebola virus, and human cytomegaloviral infections, AMPK plays a detrimental role. AMPK-targeting agents or small molecules enhance the antiviral response and contribute to the control of microbial and parasitic infections. In addition, this review focuses on the double-edged role of AMPK in innate and adaptive immune responses to infection. Understanding how AMPK regulates host defenses will enable development of more effective host-directed therapeutic strategies against infectious diseases.
Collapse
Affiliation(s)
- Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015, Korea.
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015, Korea.
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015, Korea.
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015, Korea.
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.
| | - Jae-Min Yuk
- Department of Infection Biology, Chungnam National University School of Medicine, Daejeon 35015, Korea.
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015, Korea.
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015, Korea.
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea.
| |
Collapse
|
44
|
Ji S, Zhu M, Zhang J, Cai Y, Zhai X, Wang D, Li G, Su S, Zhou J. Microarray analysis of lncRNA expression in rabies virus infected human neuroblastoma cells. INFECTION GENETICS AND EVOLUTION 2018; 67:88-100. [PMID: 30391720 DOI: 10.1016/j.meegid.2018.10.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
Rabies, caused by the rabies virus (RABV), is the oldest known zoonotic infectious disease. Although the molecular mechanisms of RABV pathogenesis have been investigated extensively, the interactions between host and RABV are not clearly understood. It is now known that long non-coding RNAs (lncRNAs) participate in various physiological and pathological processes, but their possible roles in the host response to RABV infection remain to be elucidated. To better understand the pathogenesis of RABV, RNAs from RABV-infected and uninfected human neuroblastoma cells (SK-N-SH) were analyzed using human lncRNA microarrays. We identified 896 lncRNAs and 579 mRNAs that were differentially expressed after infection, indicating a potential role for lncRNAs in the immune response to RABV. Differentially expressed RNAs were examined using Gene Ontology (GO) analysis and were tentatively assigned to biological pathways using the Kyoto Encyclopedia of Genes and Genomes (KEGG). A lncRNA-mRNA-transcription factor co-expression network was constructed to relate lncRNAs to regulatory factors and pathways that may be important in virus-host interactions. The network analysis suggests that E2F4, TAF7 and several lncRNAs function as transcriptional regulators in various signaling pathways. This study is the first global analysis of lncRNA and mRNA co-expression during RABV infection, provides deeper insight into the mechanism of RABV pathogenesis, and reveals promising candidate for future investigation.
Collapse
Affiliation(s)
- Senlin Ji
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Mengyan Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Junyan Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuchen Cai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaofeng Zhai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Dong Wang
- China Institute of Veterina Drug Control, China
| | - Gairu Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shuo Su
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| | - Jiyong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University, Hangzhou 310058, China; Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
45
|
Chang H, Tian L, Chen J, Tang A, Li C, Li Z, Yang Z. Rapamycin and ZSTK474 can have differential effects at different post‑infection time‑points regarding CVB3 replication and CVB3‑induced autophagy. Mol Med Rep 2018; 18:1088-1094. [PMID: 29845290 DOI: 10.3892/mmr.2018.9037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 05/03/2018] [Indexed: 11/05/2022] Open
Abstract
Coxsackievirus B3 (CVB3) infection has been shown to stimulate autophagy. We have demonstrated that the inhibition of phosphoinositide 3‑kinase (PI3K)/protein kinase B/mammalian target of rapamycin complex (mTORC) signaling pathway could affect the autophagic reaction induced by CVB3 infection in our previous study. However, the processes associating autophagy and CVB3 replication remain to be determined. In the present study, CVB3‑induced autophagy and its impact on viral replication were investigated. Rapamycin (inhibitor of mTOR) and ZSTK474 (inhibitor of PI3K) were used to change the autophagic reaction caused by CVB3 in Hela cells at different post‑infection (p.i.) time points (6, 9, 12 and 24 h p.i.), meanwhile, we detected the CVB3 mRNA replication and CVB3 capsid protein VP1 expression following the change of autophagy. Here, it was showed that ZSTK474 and Rapamycin promoted CVB3‑induced autophagy, as well as decreasing CVB3 mRNA replication and CVB3 capsid protein VP1 expression at 6 and 9 h p.i. ZSTK474 also alleviated CVB3‑induced autophagy, and decreased CVB3 mRNA replication and VP1 expression at 12 and 24 h p.i. However, Rapamycin continued to promote CVB3‑induced autophagy and increase CVB3 mRNA replication at 12 and 24 h p.i, as well as increase VP1 expression at 12 h, but not at 24 h, p.i. In the present study, we found Rapamycin and ZSTK474 have differential effects at different p.i. time‑points regarding CVB3 replication and CVB3‑induced autophagy. This indicates that the association between CVB3‑induced autophagy and viral replication depends on the infection time. During the early course of infection, autophagy may help host cells clear the virus, thereby providing protection, whereas when the infection time increases, autophagy may be exploited for viral replication.
Collapse
Affiliation(s)
- Huan Chang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Lang Tian
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jia Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Anliu Tang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Chunyun Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhuoying Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Zuocheng Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
46
|
Tu Z, Gong W, Zhang Y, Feng Y, Liu Y, Tu C. Inhibition of Rabies Virus by 1,2,3,4,6-Penta- O-galloyl-β-d-Glucose Involves mTOR-Dependent Autophagy. Viruses 2018; 10:v10040201. [PMID: 29673174 PMCID: PMC5923495 DOI: 10.3390/v10040201] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/04/2018] [Accepted: 04/14/2018] [Indexed: 12/25/2022] Open
Abstract
The compound 1,2,3,4,6-penta-O-galloyl-β-d-glucose (PGG), a gallotannin present in various plants such as Rhus chinensis Mill and Paeonia suffruticosa, has a broad spectrum of antiviral effects. The present study investigated its potency against infection of mice with rabies virus (RABV). Results demonstrated that PGG strongly inhibited virus titers (50-fold), viral mRNA expression (up to 90%), and protein synthesis in vitro. Importantly, we found that PGG not only suppressed viral adsorption and entry, but also directly inactivated RABV through suppression of autophagy by mediating activation of the mTOR-dependent autophagy signaling pathway. In vivo, PGG (10 mg/kg) alleviated the clinical symptoms and reduced the mortality of infected mice by 27.3%. Collectively, our results indicate that PGG has potent anti-RABV effect, and merits further investigation as an anti-RABV drug.
Collapse
Affiliation(s)
- Zhongzhong Tu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Jilin 130122, China.
| | - Wenjie Gong
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Jilin 130122, China.
| | - Yan Zhang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Jilin 130122, China.
| | - Ye Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Jilin 130122, China.
| | - Yan Liu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Jilin 130122, China.
| | - Changchun Tu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Jilin 130122, China.
| |
Collapse
|
47
|
Xu Y, Cai X, Zong B, Feng R, Ji Y, Chen G, Li Z. Qianlie Xiaozheng Decoction Induces Autophagy in Human Prostate Cancer Cells via Inhibition of the Akt/mTOR Pathway. Front Pharmacol 2018; 9:234. [PMID: 29670523 PMCID: PMC5893804 DOI: 10.3389/fphar.2018.00234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
Qianlie Xiaozheng decoction (QLXZD), a traditional Chinese medicinal formula, has been used clinically to treat advanced prostate cancer (PCa) for more than 10 years. However, experimental evidence supporting its efficacy is lacking. Here, we investigated the anticancer properties and molecular mechanism of QLXZD in vitro in a human PCa cell line (PC3) and in vivo using PC3 xenografts in nude mice. We confirmed the antineoplastic activity of QLXZD by analyzing cell viability and tumor volume growth, which decreased significantly compared to that of the controls. Autophagy following QLXZD treatment was detected morphologically using transmission electron microscopy and was confirmed by measuring the expression of autophagy markers (LC3-II and p62) using fluorescence analysis, flow cytometry, and western blotting. Increasing autophagic flux induced by QLXZD was monitored via pmCherry-GFP-LC3 fluorescence analysis. QLXZD-induced autophagic cell death was alleviated by the autophagy inhibitors, 3-methyl adenine and hydroxychloroquine. We evaluated the total expression and phosphorylation levels of proteins involved in the Akt/mTOR pathway regulating autophagy. Phosphorylation of Akt, mTOR, and p70S6K, but not total protein levels, decreased following treatment. This is the first study to demonstrate the autophagy-related mechanistic pathways utilized during QLXZD-mediated antitumor activity both in vitro and in vivo. These findings support the clinical use of QLXZD for PCa treatment.
Collapse
Affiliation(s)
- Yuehua Xu
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, China
| | - Xueting Cai
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Bin Zong
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, China
| | - Rui Feng
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, China
| | - Yali Ji
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, China
| | - Gang Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhongxing Li
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, China
| |
Collapse
|
48
|
|
49
|
Immunological aspects of rabies: a literature review. Arch Virol 2017; 162:3251-3268. [PMID: 28726129 DOI: 10.1007/s00705-017-3484-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/27/2017] [Indexed: 02/08/2023]
Abstract
Rabies is a lethal disease caused by the neurotropic virus rabies virus (RABV), and it remains an important public health problem globally. It is known that the host immune response is important for control of viral infection and promoting viral clearance. In this context, it is well documented that, in addition to RABV neutralizing antibody, interferons and cell-mediated immunity also have an important role in preventing the establishment of disease. On the other hand, RABV suppresses host immunity through different mechanisms, for example, direct inhibition of host gene expression, sequestration of pathogen-associated molecular patterns, or modification of cytokine signalling pathways, which hinder the protective host immune responses to RABV infection. Here, we review the immunological aspects of rabies, highlighting innate and adaptive immunity, as well as the host evasion immune mechanisms used by the virus. Finally, we briefly discuss how this knowledge can direct new research and be harnessed for future therapeutic strategies.
Collapse
|
50
|
Forsberg J, Zhivotovsky B, Olsson M. Caspase-2: an orphan enzyme out of the shadows. Oncogene 2017; 36:5441-5444. [PMID: 28581521 DOI: 10.1038/onc.2017.169] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/18/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
Abstract
Caspase-2 has been embodied as an initiator or executioner protease in diverse apoptotic scenarios. However, accumulating evidence is challenging this view, pertaining to its true role. The enzyme's catalytic activity is currently implicated in various functions required for correct cell proliferation, such as counteracting genomic instability, as well as suppressing tumorigenesis. Here, apart from summarizing the latest observations in caspase-2-related research, we make an attempt to reconcile these findings and discuss their implications for future directions.
Collapse
Affiliation(s)
- J Forsberg
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - B Zhivotovsky
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - M Olsson
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|