1
|
Shin SY, Lee JH, Kim JW, Im WR, Damodar K, Woo HR, Kim WK, Lee JT, Jeon SH. Evaluation of the Antiviral Activity of Tabamide A and Its Structural Derivatives against Influenza Virus. Int J Mol Sci 2023; 24:17296. [PMID: 38139128 PMCID: PMC10744247 DOI: 10.3390/ijms242417296] [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: 11/27/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Influenza viruses cause severe endemic respiratory infections in both humans and animals worldwide. The emergence of drug-resistant viral strains requires the development of new influenza therapeutics. Tabamide A (TA0), a phenolic compound isolated from tobacco leaves, is known to have antiviral activity. We investigated whether synthetic TA0 and its derivatives exhibit anti-influenza virus activity. Analysis of structure-activity relationship revealed that two hydroxyl groups and a double bond between C7 and C8 in TA0 are crucial for maintaining its antiviral action. Among its derivatives, TA25 showed seven-fold higher activity than TA0. Administration of TA0 or TA25 effectively increased survival rate and reduced weight loss of virus-infected mice. TA25 appears to act early in the viral infection cycle by inhibiting viral mRNA synthesis on the template-negative strand. Thus, the anti-influenza virus activity of TA0 can be expanded by application of its synthetic derivatives, which may aid in the development of novel antiviral therapeutics.
Collapse
Affiliation(s)
- Soo Yong Shin
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.S.); (J.H.L.); (J.W.K.); (W.R.I.)
| | - Joo Hee Lee
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.S.); (J.H.L.); (J.W.K.); (W.R.I.)
| | - Jin Woo Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.S.); (J.H.L.); (J.W.K.); (W.R.I.)
| | - Wonkyun Ronny Im
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.S.); (J.H.L.); (J.W.K.); (W.R.I.)
| | - Kongara Damodar
- Department of Chemistry and Institute of Applied Chemistry, Hallym University, Chuncheon 24252, Republic of Korea; (K.D.); (H.R.W.)
| | - Hyung Ryeol Woo
- Department of Chemistry and Institute of Applied Chemistry, Hallym University, Chuncheon 24252, Republic of Korea; (K.D.); (H.R.W.)
| | - Won-Keun Kim
- Department of Microbiology and Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea;
| | - Jeong Tae Lee
- Department of Chemistry and Institute of Applied Chemistry, Hallym University, Chuncheon 24252, Republic of Korea; (K.D.); (H.R.W.)
| | - Sung Ho Jeon
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.S.); (J.H.L.); (J.W.K.); (W.R.I.)
| |
Collapse
|
2
|
Zhu H, Chang M, Wang Q, Chen J, Liu D, He W. Identifying the Potential of miRNAs in Houttuynia cordata-Derived Exosome-Like Nanoparticles Against Respiratory RNA Viruses. Int J Nanomedicine 2023; 18:5983-6000. [PMID: 37901360 PMCID: PMC10612503 DOI: 10.2147/ijn.s425173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Pathogenic respiratory RNA viruses, including influenza A virus (IAV), respiratory syncytial virus (RSV), and SARS-CoV-2, are major causes of causes of acute respiratory infection globally. Plant-derived exosome-like nanoparticles containing miRNAs have shown substantial cross-kingdom regulatory effects on both viral and human transcripts. Houttuynia cordata (H. cordata), a traditional Chinese medicine frequently used to treat respiratory diseases. However, the role of H. cordata-derived exosome-like nanoparticles (HELNs) and the miRNA they encapsulated are unclear. Methods HELNs were isolated from fresh underground roots (uHELNs) and above ground stems and leaves (aHELNs) using differential centrifugation. The HELNs were identified using transmission electron microscopy, nanoparticle tracking analysis, and zeta potential. Small RNA sequencing and RT-PCR were employed to determine the miRNA expression in uHELNs and aHELNs. All genomes were sourced from the NCBI database. Target prediction of viral genomes was performed using RNAhybrid, while human target prediction was conducted using both RNAhybrid and Miranda. Functional enrichment analysis was applied to the predicted human targets to explore the hub targets and their roles in antiviral effects. The accessibility of miRNA target sites was determined through the MFOLD web server, and customized dual-luciferase reporter assays were administered to validate the computational findings. Results A total of 12 highly enriched miRNAs were identified in both uHELNs and aHELNs. Upon prediction and verification, miR858a and miR858b were shown to target the NP gene in H1N1, while miR166a-3p targeted the ORF1ab in SARS-CoV-2. However, no valid miRNA targets were found for RSV. Regarding human transcripts, miR168a-3p, miR168b-3p, and miR8175 were found to inhibit MAPK3 expression, and novel_mir2 could suppress both AKT1 and MAPK3 expression. Discussion This study sheds light on the collaborative antiviral mechanism of miRNAs in HELNs across two species and explores the potential antiviral scopes of both H. cordata miRNAs and HELNs.
Collapse
Affiliation(s)
- He Zhu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Mujun Chang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
- Center for Translational Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Qiulan Wang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Jing Chen
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Wenxi He
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
3
|
Zhao Z, Li J, Feng Y, Kang X, Li Y, Chen Y, Li W, Yang W, Zhao L, Huang S, Zhang S, Jiang T. Host DNA Demethylation Induced by DNMT1 Inhibition Up-Regulates Antiviral OASL Protein during Influenza a Virus Infection. Viruses 2023; 15:1646. [PMID: 37631988 PMCID: PMC10459088 DOI: 10.3390/v15081646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Influenza A virus (IAV) is a leading cause of human respiratory infections and poses a major public health concern. IAV replication can affect the expression of DNA methyltransferases (DNMTs), and the subsequent changes in DNA methylation regulate gene expression and may lead to abnormal gene transcription and translation, yet the underlying mechanisms of virus-induced epigenetic changes from DNA methylation and its role in virus-host interactions remain elusive. Here in this paper, we showed that DNMT1 expression could be suppressed following the inhibition of miR-142-5p or the PI3K/AKT signaling pathway during IAV infection, resulting in demethylation of the promotor region of the 2'-5'-oligoadenylate synthetase-like (OASL) protein and promotion of its expression in A549 cells. OASL expression enhanced RIG-I-mediated interferon induction and then suppressed replication of IAV. Our study elucidated an innate immunity mechanism by which up-regulation of OASL contributes to host antiviral responses via epigenetic modifications in IAV infection, which could provide important insights into the understanding of viral pathogenesis and host antiviral defense.
Collapse
Affiliation(s)
- Zhiyan Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.Z.); (S.H.)
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Jing Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Ye Feng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Xiaoping Kang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Yuchang Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Yuehong Chen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Wei Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Wenguang Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Lu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Shenghai Huang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.Z.); (S.H.)
| | - Sen Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| | - Tao Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.Z.); (S.H.)
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing 100071, China; (J.L.); (Y.F.); (X.K.); (Y.L.); (Y.C.); (W.L.); (W.Y.); (L.Z.)
| |
Collapse
|
4
|
Liu YY, Bai JS, Liu CC, Zhou JF, Chen J, Cheng Y, Zhou B. The Small GTPase Rab14 Regulates the Trafficking of Ceramide from Endoplasmic Reticulum to Golgi Apparatus and Facilitates Classical Swine Fever Virus Assembly. J Virol 2023; 97:e0036423. [PMID: 37255314 PMCID: PMC10231254 DOI: 10.1128/jvi.00364-23] [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: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 06/01/2023] Open
Abstract
Classical swine fever virus (CSFV) is a highly pathogenic RNA virus belonging to the Flaviviridae family that can cause deadly classical swine fever (CSF) in pigs. However, the molecular details of virus replication in the host are still unclear. Our previous studies have reported that several Rab proteins mediate CSFV entry into host cells, but it is unknown whether CSFV hijacks other Rab proteins for effective viral infection. Here, we systematically studied the role of Rab14 protein in regulating lipid metabolism for promoting viral assembly. First, Rab14 knockdown and overexpression significantly affected CSFV replication, indicating the essential role of Rab14 in CSFV infection. Interestingly, Rab14 could significantly affect virus replication in the late stage of infection. Mechanistically, CSFV NS5A recruited Rab14 to the ER, followed by ceramide transportation to the Golgi apparatus, where sphingomyelin was synthesized. The experimental data of small molecule inhibitors, RNA interference, and replenishment assay showed that the phosphatidylinositol-3-kinase (PI3K)/AKT/AS160 signaling pathway regulated the function of Rab14 to affect the transport of ceramide. More importantly, sphingomyelin on the Golgi apparatus contributed to the assembly of viral particles. Blockage of the Rab14 regulatory pathway induced the reduction of the content of sphingomyelin on the Golgi apparatus, impairing the assembly of virus particles. Our study clarifies that Rab14 regulates lipid metabolism and promotes CSFV replication, which provides insight into a novel function of Rab14 in regulating vesicles to transport lipids to the viral assembly factory. IMPORTANCE The Rab protein family members participate in the viral replication of multiple viruses and play important roles in the virus infection cycle. Our previous research focused on Rab5/7/11, which regulated the trafficking of vesicles in the early stage of CSFV infection, especially in viral endocytosis. However, the role of other Rab proteins in CSFV replication is unclear and needs further clarification. Strikingly, we screened some Rabs and found the important role of Rab14 in CSFV infection. Virus infection mobilized Rab14 to regulate the vesicle to transport ceramide from the ER to the Golgi apparatus, further promoting the synthesis of sphingomyelin and facilitating virus assembly. The treatment of inhibitors showed that the lipid transport mediated by Rab14 was regulated by the PI3K/AKT/AS160 signaling pathway. Knockdown of Rab14 or the treatment with PI3K/AKT/AS160 inhibitors reduced the ceramide content in infected cells and hindered virus assembly. Our study is the first to explain that vesicular lipid transport regulated by Rab promotes CSFV assembly, which is conducive to the development of antiviral drugs.
Collapse
Affiliation(s)
- Ya-Yun Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ji Shan Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Chun-Chun Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiang-Fei Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jing Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yan Cheng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Bin Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
5
|
Yuk JM, Park EJ, Kim IS, Jo EK. Itaconate family-based host-directed therapeutics for infections. Front Immunol 2023; 14:1203756. [PMID: 37261340 PMCID: PMC10228716 DOI: 10.3389/fimmu.2023.1203756] [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: 04/11/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
Itaconate is a crucial anti-infective and anti-inflammatory immunometabolite that accumulates upon disruption of the Krebs cycle in effector macrophages undergoing inflammatory stress. Esterified derivatives of itaconate (4-octyl itaconate and dimethyl itaconate) and its isomers (mesaconate and citraconate) are promising candidate drugs for inflammation and infection. Several itaconate family members participate in host defense, immune and metabolic modulation, and amelioration of infection, although opposite effects have also been reported. However, the precise mechanisms by which itaconate and its family members exert its effects are not fully understood. In addition, contradictory results in different experimental settings and a lack of clinical data make it difficult to draw definitive conclusions about the therapeutic potential of itaconate. Here we review how the immune response gene 1-itaconate pathway is activated during infection and its role in host defense and pathogenesis in a context-dependent manner. Certain pathogens can use itaconate to establish infections. Finally, we briefly discuss the major mechanisms by which itaconate family members exert antimicrobial effects. To thoroughly comprehend how itaconate exerts its anti-inflammatory and antimicrobial effects, additional research on the actual mechanism of action is necessary. This review examines the current state of itaconate research in infection and identifies the key challenges and opportunities for future research in this field.
Collapse
Affiliation(s)
- Jae-Min Yuk
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Infection Biology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Jin Park
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - In Soo Kim
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
6
|
Atanasova-Panchevska N, Stojchevski R, Hadzi-Petrushev N, Mitrokhin V, Avtanski D, Mladenov M. Antibacterial and Antiviral Properties of Tetrahydrocurcumin-Based Formulations: An Overview of Their Metabolism in Different Microbiotic Compartments. Life (Basel) 2022; 12:1708. [PMID: 36362863 PMCID: PMC9696410 DOI: 10.3390/life12111708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 08/29/2023] Open
Abstract
In this review, the basic metabolic characteristics of the curcuminoid tetrahydrocurcumin (THC) at the level of the intestinal microbiota were addressed. Special attention was given to the bactericidal effects of one of the THC-phospholipid formulations, which has shown greater bioavailability and activity than pure THC. Similarly, quinoline derivatives and amino acid conjugates of THC have also shown antibacterial effects in the gut. The microbial effect of pure THC is particularly pronounced in pathophysiological conditions related to the function of the intestinal microbiota, such as type II diabetes. Furthermore, the antiviral characteristics of Cur compared to those of THC are more pronounced in preventing the influenza virus. In the case of HIV infections, the new microemulsion gel formulations of THC possess high retention during preventive application in the vagina and, at the same time, do not disturb the vaginal microbiota, which is critical in maintaining low vaginal pH. Based on the reviewed literature, finding new formulations of THC which can increase its bioavailability and activity and emphasize its antibacterial and antiviral characteristics could be very important. Applying such THC formulations in preventing and treating ailments related to the microbiotic compartments in the body would be beneficial from a medical point of view.
Collapse
Affiliation(s)
- Natalija Atanasova-Panchevska
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
| | - Radoslav Stojchevski
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
| | - Vadim Mitrokhin
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, 117997 Moscow, Russia
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, 110 E 59th Street, New York, NY 10022, USA
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, P.O. Box 162, 1000 Skopje, North Macedonia
- Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova Street, 1, 117997 Moscow, Russia
| |
Collapse
|
7
|
Jheng JR, Hsieh CF, Chang YH, Ho JY, Tang WF, Chen ZY, Liu CJ, Lin TJ, Huang LY, Chern JH, Horng JT. Rosmarinic acid interferes with influenza virus A entry and replication by decreasing GSK3β and phosphorylated AKT expression levels. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:598-610. [PMID: 35650006 DOI: 10.1016/j.jmii.2022.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/20/2022] [Accepted: 04/28/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The purpose of this study was to examine the in vivo activity of rosmarinic acid (RA) - a phytochemical with antioxidant, anti-inflammatory, and antiviral properties - against influenza virus (IAV). An antibody-based kinase array and different in vitro functional assays were also applied to identify the mechanistic underpinnings by which RA may exert its anti-IAV activity. METHODS We initially examined the potential efficacy of RA using an in vivo mouse model. A time-of-addition assay and an antibody-based kinase array were subsequently applied to investigate mechanism-of-action targets for RA. The hemagglutination inhibition assay, neuraminidase inhibition assay, and cellular entry assay were also performed. RESULTS RA increased survival and prevented body weight loss in IAV-infected mice. In vitro experiments revealed that RA inhibited different IAV viruses - including oseltamivir-resistant strains. From a mechanistic point of view, RA downregulated the GSK3β and Akt signaling pathways - which are known to facilitate IAV entry and replication into host cells. CONCLUSIONS RA has promising preclinical efficacy against IAV, primarily by interfering with the GSK3β and Akt signaling pathways.
Collapse
Affiliation(s)
- Jia-Rong Jheng
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan; Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chung-Fan Hsieh
- Research Center for Emerging Viral Infections, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Yu-Hsiu Chang
- National Defense Medical Center, Institute of Preventive Medicine, Taipei 104, Taiwan
| | - Jin-Yuan Ho
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Wen-Fang Tang
- Research Center for Emerging Viral Infections, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Zi-Yi Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Chien-Jou Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Ta-Jen Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Li-Yu Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Jyh-Haur Chern
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 350, Taiwan
| | - Jim-Tong Horng
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333, Taiwan; Research Center for Emerging Viral Infections, Chang Gung University, Kweishan, Taoyuan 333, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan; Research Center for Food and Cosmetic Safety, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan.
| |
Collapse
|
8
|
Lignans from Mosla scabra Ameliorated Influenza A Virus-Induced Pneumonia via Inhibiting Macrophage Activation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1688826. [PMID: 35942373 PMCID: PMC9356792 DOI: 10.1155/2022/1688826] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022]
Abstract
The lower respiratory tract infection, induced by influenza virus, coronaviruses, and respiratory syncytial virus, remains a serious threat to human health that can cause a global pandemic. Thus, finding effective chemicals and therapeutic measures to advance the functional restoration of the respiratory tract after infection has been the emphasis of the studies on the subjects. Mosla scabra is a natural medicinal plant used for treating various lung and gastrointestinal diseases, including viral infection, cough, chronic obstructive pulmonary disease, acute gastroenteritis, and diarrhoea. In this study, the antiviral and anti-inflammatory effects of total lignans (MSTL) extracted from the plant were investigated in influenza A virus (IAV)-infected mice and RAW 264.7 macrophages. MSTL could not only protect the macrophages against IAV-induced pyroptosis but also could lighten the lung inflammation induced by IAV in vivo and in vitro. The network pharmacology analysis revealed that differentially expressed genes, mainly involving in EGFR tyrosine kinase inhibitor resistance, endocrine resistance, HIF-1 signaling pathway, C-type lectin receptor signaling pathway, and FOXO signaling pathway, contributed to the IAV-induced alveolar macrophage dysfunction. It indicated that MSTL enhanced the function of alveolar macrophages and improved IAV-induced lung injury in mice.
Collapse
|
9
|
Peiter GC, de Souza CDBT, de Oliveira LM, Pagliarin LG, dos Anjos VNF, da Silva FAF, de Melo FF, Teixeira KN. COVID-19 liver and gastroenterology findings: An in silico analysis of SARS-CoV-2 interactions with liver molecules. World J Hepatol 2022; 14:1131-1141. [PMID: 35978663 PMCID: PMC9258260 DOI: 10.4254/wjh.v14.i6.1131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/22/2022] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Coronavirus disease 19 (COVID-19) has not only been shown to affect the respiratory system, but has also demonstrated variable clinical presentations including gastrointestinal tract disorders. In addition, abnormalities in liver enzymes have been reported indicating hepatic injury. It is known that severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) might infect cells via the viral receptor angiotensin-converting enzyme 2 (ACE2) which is expressed in several organs including the liver. The viral Spike glycoprotein binds to ACE2 and must be cleaved by Furin and Type 2 Serine Protease to enter the cells. After that, the Akt/mTOR signaling pathway is activated and several COVID-19 changes are triggered.
AIM To analyze liver and gastrointestinal symptoms and cell signaling pathways triggered by SARS-CoV-2 infection due to virus-liver interactions in silico.
METHODS In this in silico study, the three-dimensional structures of the Akt, mTORC1 and Furin (receptors) were selected from the Protein Data Bank (PDB) and the structures of inhibitors (ligands) MK-2206, CC-223 and Naphthofluorescein were selected from PubChem and ZINC databases. Ligand files were downloaded as 2D structures and converted to optimized 3D structures using ViewerLite 4.2 software. Marvin Sketch® software was used to calculate prediction of the protonated form of inhibitors in a physiological environment (pH 7.4). AutoDock Tools (ADT) software was used to calculate and delimit the Grid box used in the molecular docking of each structure selected in the PDB. In addition, protonated ligands were prepared for molecular docking using ADT software. Molecular docking was performed using ADT software tools connected to Vina software. Analysis of the amino acid residues involved in ligand interactions, as well as ligand twists, the atoms involved in interactions, bond type and strength of interactions were performed using PyMol® and Discovery Studio® (BIOVIA) software.
RESULTS Molecular docking analysis showed that the mTORC1/CC-223 complex had affinity energy between the receptor and ligand of -7.7 kcal/moL with interactions ranging from 2.7 to 4.99 Å. There were four significant chemical bonds which involved two of five polypeptide chains that formed the FKBP12–Rapamycin-Binding (FRB) domain. The strongest was a hydrogen bond, the only polar interaction, and Van der Waals interactions shown to be present in 12 residues of mTORC1’s FRB domain. With regard to the Akt/MK-2206 complex there were three Van der Waals interactions and 12 chemical bonds in which seven residues of Akt were involved with all five rings of the MK-2206 structure. In this way, both ASP 388 and GLN 391 bind to the same MK-2206 ring, the smaller one. However, LYS 386 had four chemical bonds with the inhibitor, one with each structure ring, while LYS 387 binds two distinct rings. One of the MK-2206 inhibitor's rings which binds to LYS 387 also binds simultaneously to ILE 367 and LEU 385 residues, and the fifth ring of the structure was involved in a bond with the ALA 382 residue. The hydrogen bonds were the shortest bonds in the complex (2.61 and 3.08 Å) and all interactions had an affinity energy of -8.8 kcal/moL. The affinity energy in the Furin/Naphhofluorescein complex was -9.8 kcal/moL and involved six interactions ranging from 2.57 to 4.98 Å. Among them, two were polar and the others were non-polar, in addition to twelve more Van der Waals interactions. Two distinct hydrogen bonds were formed between Furin and its inhibitor involving GLN 388 and ALA 532 residues. ALA 532 also binds to two distinct rings of Naphthofluorescein, while TRP 531 residue has two simultaneous bonds with the inhibitor.
CONCLUSION Liver infection and signaling pathways altered by SARS-CoV-2 can be modulated by inhibitors that demonstrate significant interaction affinity with human proteins, which could prevent the development of infection and symptoms.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fabrício Freire de Melo
- Universidade Federal da Bahia, Campus Anísio Teixeira, Vitória da Conquista 45029-094, Bahia, Brazil
| | | |
Collapse
|
10
|
Gao S, Esparza M, Dehghan I, Aksenova V, Zhang K, Batten K, Ferretti MB, Begg BE, Cagatay T, Shay JW, García-Sastre A, Goldsmith EJ, Chen ZJ, Dasso M, Lynch KW, Cobb MH, Fontoura BMA. Nuclear speckle integrity and function require TAO2 kinase. Proc Natl Acad Sci U S A 2022; 119:e2206046119. [PMID: 35704758 PMCID: PMC9231605 DOI: 10.1073/pnas.2206046119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
Nuclear speckles are non-membrane-bound organelles known as storage sites for messenger RNA (mRNA) processing and splicing factors. More recently, nuclear speckles have also been implicated in splicing and export of a subset of mRNAs, including the influenza virus M mRNA that encodes proteins required for viral entry, trafficking, and budding. However, little is known about how nuclear speckles are assembled or regulated. Here, we uncovered a role for the cellular protein kinase TAO2 as a constituent of nuclear speckles and as a factor required for the integrity of these nuclear bodies and for their functions in pre-mRNA splicing and trafficking. We found that a nuclear pool of TAO2 is localized at nuclear speckles and interacts with nuclear speckle factors involved in RNA splicing and nuclear export, including SRSF1 and Aly/Ref. Depletion of TAO2 or inhibition of its kinase activity disrupts nuclear speckle structure, decreasing the levels of several proteins involved in nuclear speckle assembly and splicing, including SC35 and SON. Consequently, splicing and nuclear export of influenza virus M mRNA were severely compromised and caused a disruption in the virus life cycle. In fact, low levels of TAO2 led to a decrease in viral protein levels and inhibited viral replication. Additionally, depletion or inhibition of TAO2 resulted in abnormal expression of a subset of mRNAs with key roles in viral replication and immunity. Together, these findings uncovered a function of TAO2 in nuclear speckle formation and function and revealed host requirements and vulnerabilities for influenza infection.
Collapse
Affiliation(s)
- Shengyan Gao
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Matthew Esparza
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ishmael Dehghan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- HHMI, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Vasilisa Aksenova
- Division of Molecular and Cellular Biology, National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892
| | - Ke Zhang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kimberly Batten
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Max B. Ferretti
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104
| | - Bridget E. Begg
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104
| | - Tolga Cagatay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jerry W. Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Elizabeth J. Goldsmith
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Zhijian J. Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- HHMI, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Mary Dasso
- Division of Molecular and Cellular Biology, National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892
| | - Kristen W. Lynch
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104
| | - Melanie H. Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Beatriz M. A. Fontoura
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
11
|
Huo Y, Zhao G, Ruan L, Xu P, Fang G, Zhang F, Bao Z, Li X. Detect the early-warning signals of diseases based on signaling pathway perturbations on a single sample. BMC Bioinformatics 2022; 22:367. [PMID: 35045824 PMCID: PMC8772045 DOI: 10.1186/s12859-021-04286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND During the pathogenesisof complex diseases, a sudden health deterioration will occur as results of the cumulative effect of various internal or external factors. The prediction of an early warning signal (pre-disease state) before such deterioration is very important in clinical practice, especially for a single sample. The single-sample landscape entropy (SLE) was proposed to tackle this issue. However, the PPI used in SLE was lack of definite biological meanings. Besides, the calculation of multiple correlations based on limited reference samples in SLE is time-consuming and suspect. RESULTS Abnormal signals generally exert their effect through the static definite biological functions in signaling pathways across the development of diseases. Thus, it is a natural way to study the propagation of the early-warning signals based on the signaling pathways in the KEGG database. In this paper, we propose a signaling perturbation method named SSP, to study the early-warning signal in signaling pathways for single dynamic time-series data. Results in three real datasets including the influenza virus infection, lung adenocarcinoma, and acute lung injury show that the proposed SSP outperformed the SLE. Moreover, the early-warning signal can be detected by one important signaling pathway PI3K-Akt. CONCLUSIONS These results all indicate that the static model in pathways could simplify the detection of the early-warning signals.
Collapse
Affiliation(s)
- Yanhao Huo
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Geng Zhao
- Netease Youdao Information Technology (Hangzhou) Co., Ltd., Hangzhou, 310000, Zhejiang, China
| | - Luoshan Ruan
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430000, Hubei, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, 510006, Guangdong, China.,School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, 558000, Guizhou, China
| | - Gang Fang
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, 510006, Guangdong, China
| | - Fengyue Zhang
- Department of Biomedical Engineering, School of Life Science and Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhenshen Bao
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, 510006, Guangdong, China.
| | - Xin Li
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430000, Hubei, China.
| |
Collapse
|
12
|
Zeng X, Yu J, Liu P, Liu Y, Zeng T, Li B. Asiaticoside alleviates cardiomyocyte apoptosis and oxidative stress in myocardial ischemia/reperfusion injury via activating the PI3K-AKT-GSK3β pathway in vivo and in vitro. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:69. [PMID: 35282120 PMCID: PMC8848378 DOI: 10.21037/atm-21-6667] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/11/2022] [Indexed: 12/16/2022]
Abstract
Background Myocardial ischemia/reperfusion (MI/R) is one of the most important links in myocardial injury, causing damage to cardiac tissues including cell apoptosis, oxidative stress, and other serious consequences. Asiaticoside (AS), a new compound synthesized from genistein, is cardioprotective. This paper presents new evidence for the protective role of AS against MI/R injury in vitro and in vivo. Methods First, BALB/c mice underwent surgical ligation of the left anterior descending (LAD) artery to establish an MI/R animal model, and HL-1 cells were subjected to oxygen-glucose deprivation/reperfusion (OGD/R) to establish an in vitro model. Myocardial infarct size was examined by triphenyl tetrazolium chloride (TTC) staining, histopathological changes detected in heart tissues were observed using hematoxylin and eosin (H&E) and Masson staining, heart tissue apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Enzyme-linked immunosorbent assay (ELISA) kits were used to analyze cardiac troponin I (CTnI), creatine kinase-muscle and brain (CK-MB), lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA), and reduced glutathione (GSH). Cell viability was evaluated using Cell Counting Kit-8 (CCK-8) and live/dead assay. Cell apoptosis, reactive oxygen species (ROS), mitochondrial membrane potential, and mitochondrial superoxide were detected by flow cytometry and fluorescence microscopy. Both the protein expression in myocardial tissues and cardiomyocytes were examined by western blot. Results In the in vivo MI/R experiments,pretreatment of AS reduced myocardial infarct size, decrease leakage of myocardial enzyme, suppressed myocardial apoptosis, myocardial collagen deposition, and oxidative stress. In the in vitro OGD/R experiments, HL-1 cells pretreated with AS had increased cell viability, decreased apoptosis rates and depolarization of mitochondrial membrane potential, and attenuated intracellular ROS and mitochondrial superoxide. Moreover, AS downregulated the expression of apoptotic protein, and promoted phosphorylation of PI3K, AKT, and GSK3β, which was reversed by PI3K inhibitor LY294002. Conclusions The AS compound protects against MI/R injury by attenuating oxidative stress and apoptosis via activating the PI3K/AKT/GSK3β pathway in vivo and vitro.
Collapse
Affiliation(s)
- Xueliang Zeng
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junjian Yu
- Cardiovascular and Thoracic Surgery Department 2, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Peipei Liu
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yuan Liu
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Taohui Zeng
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Bei Li
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
13
|
Sohail A, Iqbal AA, Sahini N, Chen F, Tantawy M, Waqas SF, Winterhoff M, Ebensen T, Schultz K, Geffers R, Schughart K, Preusse M, Shehata M, Bähre H, Pils MC, Guzman CA, Mostafa A, Pleschka S, Falk C, Michelucci A, Pessler F. Itaconate and derivatives reduce interferon responses and inflammation in influenza A virus infection. PLoS Pathog 2022; 18:e1010219. [PMID: 35025971 PMCID: PMC8846506 DOI: 10.1371/journal.ppat.1010219] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/15/2022] [Accepted: 12/17/2021] [Indexed: 02/06/2023] Open
Abstract
Excessive inflammation is a major cause of morbidity and mortality in many viral infections including influenza. Therefore, there is a need for therapeutic interventions that dampen and redirect inflammatory responses and, ideally, exert antiviral effects. Itaconate is an immunomodulatory metabolite which also reprograms cell metabolism and inflammatory responses when applied exogenously. We evaluated effects of endogenous itaconate and exogenous application of itaconate and its variants dimethyl- and 4-octyl-itaconate (DI, 4OI) on host responses to influenza A virus (IAV). Infection induced expression of ACOD1, the enzyme catalyzing itaconate synthesis, in monocytes and macrophages, which correlated with viral replication and was abrogated by DI and 4OI treatment. In IAV-infected mice, pulmonary inflammation and weight loss were greater in Acod1-/- than in wild-type mice, and DI treatment reduced pulmonary inflammation and mortality. The compounds reversed infection-triggered interferon responses and modulated inflammation in human cells supporting non-productive and productive infection, in peripheral blood mononuclear cells, and in human lung tissue. All three itaconates reduced ROS levels and STAT1 phosphorylation, whereas AKT phosphorylation was reduced by 4OI and DI but increased by itaconate. Single-cell RNA sequencing identified monocytes as the main target of infection and the exclusive source of ACOD1 mRNA in peripheral blood. DI treatment silenced IFN-responses predominantly in monocytes, but also in lymphocytes and natural killer cells. Ectopic synthesis of itaconate in A549 cells, which do not physiologically express ACOD1, reduced infection-driven inflammation, and DI reduced IAV- and IFNγ-induced CXCL10 expression in murine macrophages independent of the presence of endogenous ACOD1. The compounds differed greatly in their effects on cellular gene homeostasis and released cytokines/chemokines, but all three markedly reduced release of the pro-inflammatory chemokines CXCL10 (IP-10) and CCL2 (MCP-1). Viral replication did not increase under treatment despite the dramatically repressed IFN responses. In fact, 4OI strongly inhibited viral transcription in peripheral blood mononuclear cells, and the compounds reduced viral titers (4OI>Ita>DI) in A549 cells whereas viral transcription was unaffected. Taken together, these results reveal itaconates as immunomodulatory and antiviral interventions for influenza virus infection. Interferon responses are part of the primary host defenses against infections. However, excessive inflammation is often a major factor in severe disease or even death in respiratory infections such as influenza, as it can lead to acute respiratory distress syndrome and sepsis-like multiorgan involvement. We applied itaconate and chemically modified versions of it (which enter cells more efficiently and can be applied at lower doses) to influenza A virus-infected human cells and lung tissue and found that these compounds markedly repress interferon responses and some pro-inflammatory processes without increasing viral replication. In fact, 4-octyl itaconate greatly decreased viral RNA replication in peripheral blood, and itaconate and 4-octyl itaconate reduced production of infectious virus in a human lung cell line. By analyzing gene expression patterns of single mononuclear cells in peripheral blood, we found that the virus infects predominantly monocytes and that these cells are the only source of ACOD1, the enzyme that synthesizes itaconate in humans. In a mouse model of influenza A virus infection, dimethyl-itaconate prevented lung inflammation and improved survival. Thus, our results suggest that novel medications based on itaconate promise to be effective treatments for influenza because they reduce deleterious inflammation and potentially also limit viral spread in the patient.
Collapse
Affiliation(s)
- Aaqib Sohail
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Azeem A. Iqbal
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Nishika Sahini
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Fangfang Chen
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Mohamed Tantawy
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Center, Dokki, Giza, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Center, Dokki, Giza, Egypt
| | - Syed F.H. Waqas
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Moritz Winterhoff
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Thomas Ebensen
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kristin Schultz
- Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klaus Schughart
- Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Matthias Preusse
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Mahmoud Shehata
- Institute for Medical Virology, Justus-Liebig-University, Giessen, Germany
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Heike Bähre
- Research Core Unit Metabolomics, Hannover Medical School, Hannover, Germany
| | - Marina C. Pils
- Mouse Pathology Platform, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A. Guzman
- Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ahmed Mostafa
- Institute for Medical Virology, Justus-Liebig-University, Giessen, Germany
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, Egypt
| | - Stephan Pleschka
- Institute for Medical Virology, Justus-Liebig-University, Giessen, Germany
- German Center for Infection Research (DZIF) partner site Giessen, Germany
| | - Christine Falk
- Department of Transplantation Immunology, Hannover Medical School, Hannover, Germany
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Frank Pessler
- Biomarkers for Infectious Diseases, Helmholtz Centre for Infection Research, Braunschweig, Germany
- TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
- Centre for Individualised Infection Medicine, Hannover, Germany
- * E-mail: , frank.pesslerwincore.de
| |
Collapse
|
14
|
Boergeling Y, Brunotte L, Ludwig S. Dynamic phospho-modification of viral proteins as a crucial regulatory layer of influenza A virus replication and innate immune responses. Biol Chem 2021; 402:1493-1504. [PMID: 34062629 DOI: 10.1515/hsz-2021-0241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Influenza viruses are small RNA viruses with a genome of about 13 kb. Because of this limited coding capacity, viral proteins have evolved to fulfil multiple functions in the infected cell. This implies that there must be mechanisms allowing to dynamically direct protein action to a distinct activity in a spatio-temporal manner. Furthermore, viruses exploit many cellular processes, which also have to be dynamically regulated during the viral replication cycle. Phosphorylation and dephosphorylation of proteins are fundamental for the control of many cellular responses. There is accumulating evidence that this mechanism represents a so far underestimated level of regulation in influenza virus replication. Here, we focus on the current knowledge of dynamics of phospho-modifications in influenza virus replication and show recent examples of findings underlining the crucial role of phosphorylation in viral transport processes as well as activation and counteraction of the innate immune response.
Collapse
Affiliation(s)
- Yvonne Boergeling
- Institute of Virology and Interdisciplinary Center for Clinical Research (IZKF), Medical Faculty, University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany
| | - Linda Brunotte
- Institute of Virology and Interdisciplinary Center for Clinical Research (IZKF), Medical Faculty, University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany
| | - Stephan Ludwig
- Institute of Virology and Interdisciplinary Center for Clinical Research (IZKF), Medical Faculty, University of Münster, Von-Esmarch-Str. 56, D-48149 Münster, Germany
| |
Collapse
|
15
|
How Influenza A Virus NS1 Deals with the Ubiquitin System to Evade Innate Immunity. Viruses 2021; 13:v13112309. [PMID: 34835115 PMCID: PMC8619935 DOI: 10.3390/v13112309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
Ubiquitination is a post-translational modification regulating critical cellular processes such as protein degradation, trafficking and signaling pathways, including activation of the innate immune response. Therefore, viruses, and particularly influenza A virus (IAV), have evolved different mechanisms to counteract this system to perform proper infection. Among IAV proteins, the non-structural protein NS1 is shown to be one of the main virulence factors involved in these viral hijackings. NS1 is notably able to inhibit the host's antiviral response through the perturbation of ubiquitination in different ways, as discussed in this review.
Collapse
|
16
|
Kleinehr J, Wilden JJ, Boergeling Y, Ludwig S, Hrincius ER. Metabolic Modifications by Common Respiratory Viruses and Their Potential as New Antiviral Targets. Viruses 2021; 13:2068. [PMID: 34696497 PMCID: PMC8540840 DOI: 10.3390/v13102068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/22/2021] [Accepted: 10/09/2021] [Indexed: 12/11/2022] Open
Abstract
Respiratory viruses are known to be the most frequent causative mediators of lung infections in humans, bearing significant impact on the host cell signaling machinery due to their host-dependency for efficient replication. Certain cellular functions are actively induced by respiratory viruses for their own benefit. This includes metabolic pathways such as glycolysis, fatty acid synthesis (FAS) and the tricarboxylic acid (TCA) cycle, among others, which are modified during viral infections. Here, we summarize the current knowledge of metabolic pathway modifications mediated by the acute respiratory viruses respiratory syncytial virus (RSV), rhinovirus (RV), influenza virus (IV), parainfluenza virus (PIV), coronavirus (CoV) and adenovirus (AdV), and highlight potential targets and compounds for therapeutic approaches.
Collapse
Affiliation(s)
- Jens Kleinehr
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| | - Janine J. Wilden
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| | - Yvonne Boergeling
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| | - Stephan Ludwig
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
- Cells in Motion Interfaculty Centre (CiMIC), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Eike R. Hrincius
- Institute of Virology Muenster (IVM), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.K.); (J.J.W.); (Y.B.); (S.L.)
| |
Collapse
|
17
|
Almutairy BK, Alshetaili A, Anwer MK, Ali N. In silico identification of MicroRNAs targeting the key nucleator of stress granules, G3BP: Promising therapeutics for SARS-CoV-2 infection. Saudi J Biol Sci 2021; 28:7499-7504. [PMID: 34456603 PMCID: PMC8381622 DOI: 10.1016/j.sjbs.2021.08.056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 11/05/2022] Open
Abstract
Stress granules (SGs) are non-membrane ribonucleoprotein condensates formed in response to environmental stress conditions via liquid–liquid phase separation (LLPS). SGs are involved in the pathogenesis of aging and aging-associated diseases, cancers, viral infection, and several other diseases. GTPase-activating protein (SH3 domain)-binding protein 1 and 2 (G3BP1/2) is a key component and commonly used marker of SGs. Recent studies have shown that SARS-CoV-2 nucleocapsid protein via sequestration of G3BPs inhibits SGs formation in the host cells. In this study, we have identified putative miRNAs targeting G3BP in search of modulators of the G3BP expression. These miRNAs could be considered as new therapeutic targets against COVID-19 infection via the regulation of SG assembly and dynamics.
Collapse
Affiliation(s)
- Bjad K Almutairy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Abdullah Alshetaili
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
18
|
Yang J, He Z, Chen C, Li S, Qian J, Zhao J, Fang R. Toxoplasma gondii Infection Inhibits Histone Crotonylation to Regulate Immune Response of Porcine Alveolar Macrophages. Front Immunol 2021; 12:696061. [PMID: 34322124 PMCID: PMC8312545 DOI: 10.3389/fimmu.2021.696061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/18/2021] [Indexed: 01/01/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular parasite that can infect almost all warm-blooded animals, causing serious public health problems. Lysine crotonylation (Kcr) is a newly discovered posttranslational modification (PTM), which is first identified on histones and has been proved relevant to procreation regulation, transcription activation, and cell signaling pathway. However, the biological functions of histone crotonylation have not yet been reported in macrophages infected with T. gondii. As a result, a total of 1,286 Kcr sites distributed in 414 proteins were identified and quantified, demonstrating the existence of crotonylation in porcine alveolar macrophages. According to our results, identified histones were overall downregulated. HDAC2, a histone decrotonylase, was found to be significantly increased, which might be the executor of histone Kcr after parasite infection. In addition, T. gondii infection inhibited the crotonylation of H2B on K12, contributing on the suppression of epigenetic regulation and NF-κB activation. Nevertheless, the reduction of histone crotonylation induced by parasite infection could promote macrophage proliferation via activating PI3K/Akt signaling pathway. The present findings point to a comprehensive understanding of the biological functions of histone crotonylation in porcine alveolar macrophages, thereby providing a certain research basis for the mechanism research on the immune response of host cells against T. gondii infection.
Collapse
Affiliation(s)
- Jing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhengming He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengjie Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Senyang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jiahui Qian
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
19
|
Deinhardt-Emmer S, Jäckel L, Häring C, Böttcher S, Wilden JJ, Glück B, Heller R, Schmidtke M, Koch M, Löffler B, Ludwig S, Ehrhardt C. Inhibition of Phosphatidylinositol 3-Kinase by Pictilisib Blocks Influenza Virus Propagation in Cells and in Lungs of Infected Mice. Biomolecules 2021; 11:biom11060808. [PMID: 34072389 PMCID: PMC8228449 DOI: 10.3390/biom11060808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Influenza virus (IV) infections are considered to cause severe diseases of the respiratory tract. Beyond mild symptoms, the infection can lead to respiratory distress syndrome and multiple organ failure. Occurrence of resistant seasonal and pandemic strains against the currently licensed antiviral medications points to the urgent need for new and amply available anti-influenza drugs. Interestingly, the virus-supportive function of the cellular phosphatidylinositol 3-kinase (PI3K) suggests that this signaling module may be a potential target for antiviral intervention. In the sense of repurposing existing drugs for new indications, we used Pictilisib, a known PI3K inhibitor to investigate its effect on IV infection, in mono-cell-culture studies as well as in a human chip model. Our results indicate that Pictilisib is a potent inhibitor of IV propagation already at early stages of infection. In a murine model of IV pneumonia, the in vitro key findings were verified, showing reduced viral titers as well as inflammatory response in the lung after delivery of Pictilisib. Our data identified Pictilisib as a promising drug candidate for anti-IV therapies that warrant further studying. These results further led to the conclusion that the repurposing of previously approved substances represents a cost-effective and efficient way for development of novel antiviral strategies.
Collapse
Affiliation(s)
- Stefanie Deinhardt-Emmer
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, D-07747 Jena, Germany; (M.K.); (B.L.)
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knoell-Str. 2, D-07745 Jena, Germany; (C.H.); (S.B.); (B.G.); (M.S.)
- Correspondence: (S.D.-E.); (C.E.); Tel.: +49-(0)3641-9393640 (S.D.-E.); +49-(0)3641-9395700 (C.E.)
| | - Laura Jäckel
- Institute of Virology Muenster, Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University, D-48149 Muenster, Germany; (L.J.); (J.J.W.); (S.L.)
| | - Clio Häring
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knoell-Str. 2, D-07745 Jena, Germany; (C.H.); (S.B.); (B.G.); (M.S.)
| | - Sarah Böttcher
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knoell-Str. 2, D-07745 Jena, Germany; (C.H.); (S.B.); (B.G.); (M.S.)
| | - Janine J. Wilden
- Institute of Virology Muenster, Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University, D-48149 Muenster, Germany; (L.J.); (J.J.W.); (S.L.)
| | - Brigitte Glück
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knoell-Str. 2, D-07745 Jena, Germany; (C.H.); (S.B.); (B.G.); (M.S.)
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knoell-Str. 2, D-07745 Jena, Germany;
| | - Michaela Schmidtke
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knoell-Str. 2, D-07745 Jena, Germany; (C.H.); (S.B.); (B.G.); (M.S.)
| | - Mirijam Koch
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, D-07747 Jena, Germany; (M.K.); (B.L.)
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, D-07747 Jena, Germany; (M.K.); (B.L.)
| | - Stephan Ludwig
- Institute of Virology Muenster, Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University, D-48149 Muenster, Germany; (L.J.); (J.J.W.); (S.L.)
| | - Christina Ehrhardt
- Section of Experimental Virology, Institute of Medical Microbiology, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knoell-Str. 2, D-07745 Jena, Germany; (C.H.); (S.B.); (B.G.); (M.S.)
- Correspondence: (S.D.-E.); (C.E.); Tel.: +49-(0)3641-9393640 (S.D.-E.); +49-(0)3641-9395700 (C.E.)
| |
Collapse
|
20
|
Ludwig S, Hrincius ER, Boergeling Y. The Two Sides of the Same Coin-Influenza Virus and Intracellular Signal Transduction. Cold Spring Harb Perspect Med 2021; 11:a038513. [PMID: 31871235 PMCID: PMC7778220 DOI: 10.1101/cshperspect.a038513] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cells respond to extracellular agents by activation of intracellular signaling pathways. Viruses can be regarded as such agents, leading to a firework of signaling inside the cell, primarily induced by pathogen-associated molecular patterns (PAMPs) that provoke safeguard mechanisms to defend from the invader. In the constant arms race between pathogen and cellular defense, viruses not only have evolved mechanisms to suppress or misuse supposedly antiviral signaling processes for their own benefit but also actively induce signaling to promote replication. This creates viral dependencies that may be exploited for novel strategies of antiviral intervention. Here, we will summarize the current knowledge of activation and function of influenza virus-induced signaling pathways with a focus on nuclear factor (NF)-κB signaling, mitogen-activated protein kinase cascades, and the phosphatidylinositol-3-kinase pathway. We will discuss the opportunities and drawbacks of targeting these signaling pathways for antiviral intervention.
Collapse
Affiliation(s)
- Stephan Ludwig
- Institute of Virology Muenster, University of Muenster, 48149 Muenster, Germany
| | - Eike R Hrincius
- Institute of Virology Muenster, University of Muenster, 48149 Muenster, Germany
| | - Yvonne Boergeling
- Institute of Virology Muenster, University of Muenster, 48149 Muenster, Germany
| |
Collapse
|
21
|
Host factors involved in influenza virus infection. Emerg Top Life Sci 2020; 4:389-398. [PMID: 33210707 DOI: 10.1042/etls20200232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022]
Abstract
Influenza virus causes an acute febrile respiratory disease in humans that is commonly known as 'flu'. Influenza virus has been around for centuries and is one of the most successful, and consequently most studied human viruses. This has generated tremendous amount of data and information, thus it is pertinent to summarise these for, particularly interdisciplinary readers. Viruses are acellular organisms and exist at the interface of living and non-living. Due to this unique characteristic, viruses require another organism, i.e. host to survive. Viruses multiply inside the host cell and are obligate intracellular pathogens, because their relationship with the host is almost always harmful to host. In mammalian cells, the life cycle of a virus, including influenza is divided into five main steps: attachment, entry, synthesis, assembly and release. To complete these steps, some viruses, e.g. influenza utilise all three parts - plasma membrane, cytoplasm and nucleus, of the cell; whereas others, e.g. SARS-CoV-2 utilise only plasma membrane and cytoplasm. Hence, viruses interact with numerous host factors to complete their life cycle, and these interactions are either exploitative or antagonistic in nature. The host factors involved in the life cycle of a virus could be divided in two broad categories - proviral and antiviral. This perspective has endeavoured to assimilate the information about the host factors which promote and suppress influenza virus infection. Furthermore, an insight into host factors that play a dual role during infection or contribute to influenza virus-host adaptation and disease severity has also been provided.
Collapse
|
22
|
Merk R, Heßelbach K, Osipova A, Popadić D, Schmidt-Heck W, Kim GJ, Günther S, Piñeres AG, Merfort I, Humar M. Particulate Matter (PM 2.5) from Biomass Combustion Induces an Anti-Oxidative Response and Cancer Drug Resistance in Human Bronchial Epithelial BEAS-2B Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E8193. [PMID: 33171923 PMCID: PMC7664250 DOI: 10.3390/ijerph17218193] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Nearly half of the world's population relies on combustion of solid biofuels to cover fundamental energy demands. Epidemiologic data demonstrate that particularly long-term emissions adversely affect human health. However, pathological molecular mechanisms are insufficiently characterized. Here we demonstrate that long-term exposure to fine particulate matter (PM2.5) from biomass combustion had no impact on cellular viability and proliferation but increased intracellular reactive oxygen species (ROS) levels in bronchial epithelial BEAS-2B cells. Exposure to PM2.5 induced the nuclear factor erythroid 2-related factor 2 (Nrf2) and mediated an anti-oxidative response, including enhanced levels of intracellular glutathione (GSH) and nuclear accumulation of heme oxygenase-1 (HO-1). Activation of Nrf2 was promoted by the c-Jun N-terminal kinase JNK1/2, but not p38 or Akt, which were also induced by PM2.5. Furthermore, cells exposed to PM2.5 acquired chemoresistance to doxorubicin, which was associated with inhibition of apoptosis and elevated levels of GSH in these cells. Our findings propose that exposure to PM2.5 induces molecular defense mechanisms, which prevent cellular damage and may thus explain the initially relative rare complications associated with PM2.5. However, consistent induction of pro-survival pathways may also promote the progression of diseases. Environmental conditions inducing anti-oxidative responses may have the potential to promote a chemoresistant cellular phenotype.
Collapse
Affiliation(s)
- Regina Merk
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Katharina Heßelbach
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Anastasiya Osipova
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Désirée Popadić
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Wolfgang Schmidt-Heck
- Department of Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll Institute (HKI), 07745 Jena, Germany;
| | - Gwang-Jin Kim
- Department of Pharmaceutical Bioinformatics, Institute of Pharmaceutical Sciences, Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany; (G.-J.K.); (S.G.)
| | - Stefan Günther
- Department of Pharmaceutical Bioinformatics, Institute of Pharmaceutical Sciences, Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany; (G.-J.K.); (S.G.)
| | - Alfonso García Piñeres
- Centro de Investigación en Biología Celular y Molecular (CIBCM), Universidad de Costa Rica, 11501-2060 San José, Costa Rica;
- Escuela de Química, Universidad de Costa Rica, 11501-2060 San José, Costa Rica
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany
| | - Matjaz Humar
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| |
Collapse
|
23
|
Appelberg S, Gupta S, Svensson Akusjärvi S, Ambikan AT, Mikaeloff F, Saccon E, Végvári Á, Benfeitas R, Sperk M, Ståhlberg M, Krishnan S, Singh K, Penninger JM, Mirazimi A, Neogi U. Dysregulation in Akt/mTOR/HIF-1 signaling identified by proteo-transcriptomics of SARS-CoV-2 infected cells. Emerg Microbes Infect 2020; 9:1748-1760. [PMID: 32691695 PMCID: PMC7473213 DOI: 10.1080/22221751.2020.1799723] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
How severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections engage cellular host pathways and innate immunity in infected cells remains largely elusive. We performed an integrative proteo-transcriptomics analysis in SARS-CoV-2 infected Huh7 cells to map the cellular response to the invading virus over time. We identified four pathways, ErbB, HIF-1, mTOR and TNF signaling, among others that were markedly modulated during the course of the SARS-CoV-2 infection in vitro. Western blot validation of the downstream effector molecules of these pathways revealed a dose-dependent activation of Akt, mTOR, S6K1 and 4E-BP1 at 24 hours post infection (hpi). However, we found a significant inhibition of HIF-1α through 24hpi and 48hpi of the infection, suggesting a crosstalk between the SARS-CoV-2 and the Akt/mTOR/HIF-1 signaling pathways. Inhibition of the mTOR signaling pathway using Akt inhibitor MK-2206 showed a significant reduction in virus production. Further investigations are required to better understand the molecular sequelae in order to guide potential therapy in the management of severe coronavirus disease 2019 (COVID-19) patients.
Collapse
Affiliation(s)
| | - Soham Gupta
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Sara Svensson Akusjärvi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anoop T Ambikan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Flora Mikaeloff
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Elisa Saccon
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University Stockholm, Sweden
| | - Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Marie Ståhlberg
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Shuba Krishnan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Kamal Singh
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Veterinary Pathobiology and the Bond Life Science Center, University of Missouri, Columbia, MO, USA
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, Canada
| | - Ali Mirazimi
- Public Health Agency of Sweden, Solna, Sweden.,Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,National Veterinary Institute, Uppsala, Sweden
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Veterinary Pathobiology and the Bond Life Science Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
24
|
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: 2.4] [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
|
25
|
Casalino-Matsuda SM, Chen F, Gonzalez-Gonzalez FJ, Nair A, Dib S, Yemelyanov A, Gates KL, Budinger GRS, Beitel GJ, Sporn PHS. Hypercapnia Suppresses Macrophage Antiviral Activity and Increases Mortality of Influenza A Infection via Akt1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:489-501. [PMID: 32540997 PMCID: PMC7343622 DOI: 10.4049/jimmunol.2000085] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
Hypercapnia (HC), elevation of the partial pressure of CO2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that HC inhibits multiple macrophage and neutrophil antimicrobial functions and increases the mortality of bacterial pneumonia in mice. In this study, we show that normoxic HC increases viral replication, lung injury, and mortality in mice infected with influenza A virus (IAV). Elevated CO2 increased IAV replication and inhibited antiviral gene and protein expression in macrophages in vivo and in vitro. HC potentiated IAV-induced activation of Akt, whereas specific pharmacologic inhibition or short hairpin RNA knockdown of Akt1 in alveolar macrophages blocked HC's effects on IAV growth and the macrophage antiviral response. Our findings suggest that targeting Akt1 or the downstream pathways through which elevated CO2 signals could enhance macrophage antiviral host defense and improve clinical outcomes in hypercapnic patients with advanced lung disease.
Collapse
Affiliation(s)
- S Marina Casalino-Matsuda
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611;
| | - Fei Chen
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Francisco J Gonzalez-Gonzalez
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Aisha Nair
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Sandra Dib
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Alex Yemelyanov
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Khalilah L Gates
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612; and
| | - Greg J Beitel
- Department of Molecular Biosciences, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208
| | - Peter H S Sporn
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612; and
| |
Collapse
|
26
|
Human Type I Interferon Antiviral Effects in Respiratory and Reemerging Viral Infections. J Immunol Res 2020; 2020:1372494. [PMID: 32455136 PMCID: PMC7231083 DOI: 10.1155/2020/1372494] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/17/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Type I interferons (IFN-I) are a group of related proteins that help regulate the activity of the immune system and play a key role in host defense against viral infections. Upon infection, the IFN-I are rapidly secreted and induce a wide range of effects that not only act upon innate immune cells but also modulate the adaptive immune system. While IFN-I and many IFN stimulated genes are well-known for their protective antiviral role, recent studies have associated them with potential pathogenic functions. In this review, we summarize the current knowledge regarding the complex effects of human IFN-I responses in respiratory as well as reemerging flavivirus infections of public health significance and the molecular mechanisms by which viral proteins antagonize the establishment of an antiviral host defense. Antiviral effects and immune modulation of IFN-stimulated genes is discussed in resisting and controlling pathogens. Understanding the mechanisms of these processes will be crucial in determining how viral replication can be effectively controlled and in developing safe and effective vaccines and novel therapeutic strategies.
Collapse
|
27
|
Yu Y, Zhang Y, Wang S, Liu W, Hao C, Wang W. Inhibition effects of patchouli alcohol against influenza a virus through targeting cellular PI3K/Akt and ERK/MAPK signaling pathways. Virol J 2019; 16:163. [PMID: 31870450 PMCID: PMC6929483 DOI: 10.1186/s12985-019-1266-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/05/2019] [Indexed: 01/30/2023] Open
Abstract
Background Patchouli alcohol (PA) is a tricyclic sesquiterpene extracted from Pogostemonis Herba, which is a traditional Chinese medicine used for therapy of inflammatory diseases. Recent studies have shown that PA has various pharmacological activities, including anti-bacterial and anti-viral effects. Methods In this study, the anti-influenza virus (IAV) activities and mechanisms were investigated both in vitro and in vivo. The inhibitory effects of PA against IAV in vitro were evaluated by plaque assay and immunofluorescence assay. The neuraminidase inhibition assay, hemagglutination inhibition (HI) assay, and western blot assay were used to explore the anti-viral mechanisms. The anti-IAV activities in vivo were determined by mice pneumonia model and HE staining. Results The results showed that PA significantly inhibited different IAV strains multiplication in vitro, and may block IAV infection through inactivating virus particles directly and interfering with some early stages after virus adsorption. Cellular PI3K/Akt and ERK/MAPK signaling pathways may be involved in the anti-IAV actions of PA. Intranasal administration of PA markedly improved mice survival and attenuated pneumonia symptoms in IAV infected mice, comparable to the effects of Oseltamivir. Conclusions Therefore, Patchouli alcohol has the potential to be developed into a novel anti-IAV agent in the future.
Collapse
Affiliation(s)
- Yunjia Yu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Yang Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Shuyao Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Wei Liu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Cui Hao
- Systems Biology & Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, 266003, People's Republic of China.
| | - Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, People's Republic of China. .,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, People's Republic of China.
| |
Collapse
|
28
|
Zhou T, Sun Y, Wang Y, Chen X, Zhuo L, Bu L, Xu S, Han J, Li X, Shi J. Umbilical Cord Blood Mesenchymal Stem Cells Enhance Lipopolysaccharide-Induced IL-10 and IL-37 Production in THP-1 Cells. Inflammation 2019; 42:987-993. [PMID: 30707387 DOI: 10.1007/s10753-019-00960-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Umbilical cord blood mesenchymal stem cells (UCB-MSCs) have been shown to be a source of stem cells for use in cellular therapies and have immunomodulatory effects on several immune cells in an inflammatory environment. However, whether UCB-MSCs have immunomodulatory effects against lipopolysaccharide (LPS)-induced inflammatory cytokine secretion in macrophages and whether it is involved in phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway remain unclear. After co-culture of UCB-MSCs and phorbol 12-myristate 13-acetate (PMA)-activated human THP-1 cells using a transwell system, it showed that LPS significantly induced increases in the expression levels of interleukin 10 (IL-10), interleukin 37 (IL-37), phospho-PI3K (p-PI3K), and phospho-Akt (p-Akt) in macrophages. UCB-MSCs upregulated the expression of IL-10, IL-37, p-PI3K, and p-Akt, while it had no obvious effect on PI3K and Akt levels. Inhibitors of PI3K (LY294002) significantly suppressed the expression of IL-10, IL-37, p-PI3K, and p-Akt; however, it had no effect on the expression levels of PI3K and Akt. The present study demonstrated that UCB-MSCs increased the LPS-stimulated expression of IL-10 and IL-37 in macrophages through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China
| | - Yan Sun
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China
| | - Yanli Wang
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China
| | - Xiaobing Chen
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China
| | - Luo Zhuo
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China
| | - Lin Bu
- Department of Critical Care Medicine, Xuzhou Medical University Affiliated Hospital, No.99 West Huaihai Road, Xuzhou, 221000, China
| | - Suo Xu
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China
| | - Jiayan Han
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China
| | - Xiaomin Li
- Department of Emergency Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China.
| | - Jiaxin Shi
- Department of Respiratory Medicine, Xuzhou Medical University Affiliated Hospital of Lianyungang, No.182 North Tongguan Road, Lianyungang, 222002, China.
| |
Collapse
|
29
|
New therapeutic targets for the prevention of infectious acute exacerbations of COPD: role of epithelial adhesion molecules and inflammatory pathways. Clin Sci (Lond) 2019; 133:1663-1703. [PMID: 31346069 DOI: 10.1042/cs20181009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022]
Abstract
Chronic respiratory diseases are among the leading causes of mortality worldwide, with the major contributor, chronic obstructive pulmonary disease (COPD) accounting for approximately 3 million deaths annually. Frequent acute exacerbations (AEs) of COPD (AECOPD) drive clinical and functional decline in COPD and are associated with accelerated loss of lung function, increased mortality, decreased health-related quality of life and significant economic costs. Infections with a small subgroup of pathogens precipitate the majority of AEs and consequently constitute a significant comorbidity in COPD. However, current pharmacological interventions are ineffective in preventing infectious exacerbations and their treatment is compromised by the rapid development of antibiotic resistance. Thus, alternative preventative therapies need to be considered. Pathogen adherence to the pulmonary epithelium through host receptors is the prerequisite step for invasion and subsequent infection of surrounding structures. Thus, disruption of bacterial-host cell interactions with receptor antagonists or modulation of the ensuing inflammatory profile present attractive avenues for therapeutic development. This review explores key mediators of pathogen-host interactions that may offer new therapeutic targets with the potential to prevent viral/bacterial-mediated AECOPD. There are several conceptual and methodological hurdles hampering the development of new therapies that require further research and resolution.
Collapse
|
30
|
Meineke R, Rimmelzwaan GF, Elbahesh H. Influenza Virus Infections and Cellular Kinases. Viruses 2019; 11:E171. [PMID: 30791550 PMCID: PMC6410056 DOI: 10.3390/v11020171] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/24/2022] Open
Abstract
Influenza A viruses (IAVs) are a major cause of respiratory illness and are responsible for yearly epidemics associated with more than 500,000 annual deaths globally. Novel IAVs may cause pandemic outbreaks and zoonotic infections with, for example, highly pathogenic avian influenza virus (HPAIV) of the H5N1 and H7N9 subtypes, which pose a threat to public health. Treatment options are limited and emergence of strains resistant to antiviral drugs jeopardize this even further. Like all viruses, IAVs depend on host factors for every step of the virus replication cycle. Host kinases link multiple signaling pathways in respond to a myriad of stimuli, including viral infections. Their regulation of multiple response networks has justified actively targeting cellular kinases for anti-cancer therapies and immune modulators for decades. There is a growing volume of research highlighting the significant role of cellular kinases in regulating IAV infections. Their functional role is illustrated by the required phosphorylation of several IAV proteins necessary for replication and/or evasion/suppression of the innate immune response. Identified in the majority of host factor screens, functional studies further support the important role of kinases and their potential as host restriction factors. PKC, ERK, PI3K and FAK, to name a few, are kinases that regulate viral entry and replication. Additionally, kinases such as IKK, JNK and p38 MAPK are essential in mediating viral sensor signaling cascades that regulate expression of antiviral chemokines and cytokines. The feasibility of targeting kinases is steadily moving from bench to clinic and already-approved cancer drugs could potentially be repurposed for treatments of severe IAV infections. In this review, we will focus on the contribution of cellular kinases to IAV infections and their value as potential therapeutic targets.
Collapse
Affiliation(s)
- Robert Meineke
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), Bünteweg 17, 30559 Hannover, Germany.
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), Bünteweg 17, 30559 Hannover, Germany.
| | - Husni Elbahesh
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
31
|
Zhang J, Ruan T, Sheng T, Wang J, Sun J, Wang J, Prinz RA, Peng D, Liu X, Xu X. Role of c-Jun terminal kinase (JNK) activation in influenza A virus-induced autophagy and replication. Virology 2019; 526:1-12. [PMID: 30316042 PMCID: PMC6424123 DOI: 10.1016/j.virol.2018.09.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 01/12/2023]
Abstract
The non-structural protein 1 (NS1) of different influenza A virus (IAV) strains can differentially regulate the activity of c-Jun terminal kinase (JNK) and PI-3 kinase (PI3K). Whether varying JNK and PI3K activation impacts autophagy and IAV replication differently remains uncertain. Here we report that H5N1 (A/mallard/Huadong/S/2005) influenza A virus induced functional autophagy, as evidenced by increased LC3 lipidation and decreased p62 levels, and the presence of autolysosomes in chicken fibroblast cells. H9N2 (A/chicken/Shanghai/F/98) virus weakly induced autophagy, whereas H1N1 virus (A/PR/8/34, PR8) blocked autophagic flux. H5N1 virus activated JNK but inhibited the PI-3 kinase pathway. In contrast, N9N2 virus infection led to modest JNK activation and strong PI-3 kinase activation; whereas H1N1 virus activated the PI-3 kinase pathway but did not activate JNK. SP600125, a JNK inhibitor, inhibited H5N1 virus-induced autophagy and virus replication in a DF-1 chicken fibroblast cell line. Our study uncovered a previously unrecognized role of JNK in IAV replication and autophagy.
Collapse
Affiliation(s)
- Jingting Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Tao Ruan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Tianyu Sheng
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Jiongjiong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Jing Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Jin Wang
- Center for Immunological Research, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Richard A Prinz
- Department of Surgery, NorthShore University Health System, Evanston IL60201, USA
| | - Daxin Peng
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China
| | - Xiulong Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, Jiangsu Province, PR China.
| |
Collapse
|
32
|
Chang C, Xi L, Zhang J, Zhao W, Liu Z, Guo J, Xu C. Roles of Cyclin A, Myc, Jun and Ppm1l in tumourigenic transformation of NIH3T3 cell. J Genet 2018; 97:1155-1168. [PMID: 30555065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To analyse the mechanism of tumourigenic transformation of NIH3T3 cells at the transcriptional level, we used cancerogen 3-methylcholanthrene (3-MCA) and cancerogenic substance phorbol-12-myristate-13-acetate (PMA) to transform NIH3T3 cells and the assessment of transformation was performed using Giemsa staining and methylcellulose colony formation assay. Changes in gene expression profile were detected by Mouse Genome 430 2.0 microarray; and quantitative real-time polymerase chain reaction and Western blotting were used to verify the expression changes of mRNAs and proteins, respectively. With the aid of bioinformatics method, five signalling pathways were identified to participate in different stages of NIH3T3 cell transformation. Further, our study suggested that oncogenes Cyclin A, Myc, Jun and the tumour suppressor gene Ppm1l may play important roles in these pathways.
Collapse
Affiliation(s)
- Cuifang Chang
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation, Henan Normal University, Xinxiang 453007, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Transcriptome Analysis of Infected and Bystander Type 2 Alveolar Epithelial Cells during Influenza A Virus Infection Reveals In Vivo Wnt Pathway Downregulation. J Virol 2018; 92:JVI.01325-18. [PMID: 30111569 DOI: 10.1128/jvi.01325-18] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/12/2018] [Indexed: 12/29/2022] Open
Abstract
Influenza virus outbreaks remain a serious threat to public health. A greater understanding of how cells targeted by the virus respond to the infection can provide insight into the pathogenesis of disease. Here we examined the transcriptional profile of in vivo-infected and uninfected type 2 alveolar epithelial cells (AEC) in the lungs of influenza virus-infected mice. We show for the first time the unique gene expression profiles induced by the in vivo infection of AEC as well as the transcriptional response of uninfected bystander cells. This work allows us to distinguish the direct and indirect effects of infection at the cellular level. Transcriptome analysis revealed that although directly infected and bystander AEC from infected animals shared many transcriptome changes compared to AEC from uninfected animals, directly infected cells produce more interferon and express lower levels of Wnt signaling-associated transcripts, while concurrently expressing more transcripts associated with cell death pathways, than bystander uninfected AEC. The Wnt signaling pathway was downregulated in both in vivo-infected AEC and in vitro-infected human lung epithelial A549 cells. Wnt signaling did not affect type I and III interferon production by infected A549 cells. Our results reveal unique transcriptional changes that occur within infected AEC and show that influenza virus downregulates Wnt signaling. In light of recent findings that Wnt signaling is essential for lung epithelial stem cells, our findings reveal a mechanism by which influenza virus may affect host lung repair.IMPORTANCE Influenza virus infection remains a major public health problem. Utilizing a recombinant green fluorescent protein-expressing influenza virus, we compared the in vivo transcriptomes of directly infected and uninfected bystander cells from infected mouse lungs and discovered many pathways uniquely regulated in each population. The Wnt signaling pathway was downregulated in directly infected cells and was shown to affect virus but not interferon production. Our study is the first to discern the in vivo transcriptome changes induced by direct viral infection compared to mere exposure to the lung inflammatory milieu and highlight the downregulation of Wnt signaling. This downregulation has important implications for understanding influenza virus pathogenesis, as Wnt signaling is critical for lung epithelial stem cells and lung epithelial cell differentiation. Our findings reveal a mechanism by which influenza virus may affect host lung repair and suggest interventions that prevent damage or accelerate recovery of the lung.
Collapse
|
35
|
Cuartas-López AM, Hernández-Cuellar CE, Gallego-Gómez JC. Disentangling the role of PI3K/Akt, Rho GTPase and the actin cytoskeleton on dengue virus infection. Virus Res 2018; 256:153-165. [DOI: 10.1016/j.virusres.2018.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 12/22/2022]
|
36
|
Fu C, Luo J, Ye S, Yuan Z, Li S. Integrated Lung and Tracheal mRNA-Seq and miRNA-Seq Analysis of Dogs with an Avian-Like H5N1 Canine Influenza Virus Infection. Front Microbiol 2018; 9:303. [PMID: 29556219 PMCID: PMC5844969 DOI: 10.3389/fmicb.2018.00303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/09/2018] [Indexed: 12/12/2022] Open
Abstract
Avian-like H5N1 canine influenza virus (CIV) causes severe respiratory infections in dogs. However, the mechanism underlying H5N1 CIV infection in dogs is unknown. The present study aimed to identify differentially expressed miRNAs and mRNAs in the lungs and trachea in H5N1 CIV-infected dogs through a next-generation sequencing-based method. Eighteen 40-day-old beagles were inoculated intranasally with CIV, A/canine/01/Guangdong/2013 (H5N1) at a tissue culture infectious dose 50 (TCID50) of 106, and lung and tracheal tissues were harvested at 3 and 7 d post-inoculation. The tissues were processed for miRNA and mRNA analysis. By means of miRNA-gene expression integrative negative analysis, we found miRNA–mRNA pairs. Lung and trachea tissues showed 138 and 135 negative miRNA–mRNA pairs, respectively. One hundred and twenty negative miRNA–mRNA pairs were found between the different tissues. In particular, pathways including the influenza A pathway, chemokine signaling pathways, and the PI3K-Akt signaling pathway were significantly enriched in all groups in responses to virus infection. Furthermore, dysregulation of miRNA and mRNA expression was observed in the respiratory tract of H5N1 CIV-infected dogs and notably, TLR4 (miR-146), NF-κB (miR-34c) and CCL5 (miR-335), CCL10 (miR-8908-5p), and GNGT2 (miR-122) were found to play important roles in regulating pathways that resist virus infection. To our knowledge, the present study is the first to analyze miRNA and mRNA expression in H5N1 CIV-infected dogs; furthermore, the present findings provide insights into the molecular mechanisms underlying influenza virus infection.
Collapse
Affiliation(s)
- Cheng Fu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou, China.,Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Jie Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou, China.,Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Shaotang Ye
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou, China.,Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| | - Ziguo Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou, China
| | - Shoujun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou, China.,Guangdong Technological Engineering Research Center for Pet, Guangzhou, China
| |
Collapse
|
37
|
Pizzorno A, Dubois J, Machado D, Cartet G, Traversier A, Julien T, Lina B, Bourdon JC, Rosa-Calatrava M, Terrier O. Influenza A viruses alter the stability and antiviral contribution of host E3-ubiquitin ligase Mdm2 during the time-course of infection. Sci Rep 2018; 8:3746. [PMID: 29487367 PMCID: PMC5829072 DOI: 10.1038/s41598-018-22139-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/19/2018] [Indexed: 11/09/2022] Open
Abstract
The interplay between influenza A viruses (IAV) and the p53 pathway has been reported in several studies, highlighting the antiviral contribution of p53. Here, we investigated the impact of IAV on the E3-ubiquitin ligase Mdm2, a major regulator of p53, and observed that IAV targets Mdm2, notably via its non-structural protein (NS1), therefore altering Mdm2 stability, p53/Mdm2 interaction and regulatory loop during the time-course of infection. This study also highlights a new antiviral facet of Mdm2 possibly increasing the list of its many p53-independent functions. Altogether, our work contributes to better understand the mechanisms underlining the complex interactions between IAV and the p53 pathway, for which both NS1 and Mdm2 arise as key players.
Collapse
Affiliation(s)
- Andrés Pizzorno
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
| | - Julia Dubois
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
| | - Daniela Machado
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
- Laboratoire des Pathogènes Emergents, Fondation Mérieux. CIRI, UCBL1- INSERM U1111, ENS Lyon, CNRS UMR5308, Lyon, France
| | - Gaëlle Cartet
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
| | - Aurelien Traversier
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
| | - Thomas Julien
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
| | - Bruno Lina
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
- Laboratoire de Virologie, Centre National de Référence des virus Influenza, Institut des Agents Infectieux, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Jean-Christophe Bourdon
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France
| | - Olivier Terrier
- Virologie et Pathologie Humaine-VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, CNRS UMR5308, France.
| |
Collapse
|
38
|
Vasin AV, Petrova-Brodskaya AV, Plotnikova MA, Tsvetkov VB, Klotchenko SA. EVOLUTIONARY DYNAMICS OF STRUCTURAL AND FUNCTIONAL DOMAINS OF INFLUENZA A VIRUS NS1 PROTEIN. Vopr Virusol 2017; 62:246-258. [PMID: 36494956 DOI: 10.18821/0507-4088-2017-62-6-246-258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Indexed: 12/13/2022]
Abstract
Influenza A virus (IAV) NS1 protein is one of the key viral factors responsible for virus-host interactions. NS1 counteracts host antiviral defense, participates in the processing and export of cellular mRNAs, regulates the activity of viral RNA polymerase and the expression of viral genes, and influences the cellular signaling systems. Multiple NS1 functions are carried out due to the interactions with cellular factors, the number of which exceeds one hundred. It is noteworthy that only two segments of IAV genome - NS and NP - did not undergo reassortment and evolved in the course of genetic drift, beginning with the pandemic of 1918 to the present. This fact may indicate the importance of NS1 and its numerous interactions with cellular factors in the interspecific adaptation of the virus. The review presents data on the evolution of the human IAV NS1 protein and analysis of the amino acid substitutions in the main structural and functional domains of NS1 protein during evolution.
Collapse
Affiliation(s)
- A V Vasin
- Research Institute of Influenza.,Peter the Great St. Petersburg Polytechnic University
| | - A V Petrova-Brodskaya
- Research Institute of Influenza.,Peter the Great St. Petersburg Polytechnic University
| | | | - V B Tsvetkov
- Research Institute of Influenza.,A.V. Topchiev Institute of Petrochemical Synthesis, Russian Academy of Sciences.,Federal Research and Clinical Center of Physical-Chemical Medicine
| | | |
Collapse
|
39
|
Holzberg M, Boergeling Y, Schräder T, Ludwig S, Ehrhardt C. Vemurafenib Limits Influenza A Virus Propagation by Targeting Multiple Signaling Pathways. Front Microbiol 2017; 8:2426. [PMID: 29312159 PMCID: PMC5735105 DOI: 10.3389/fmicb.2017.02426] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/23/2017] [Indexed: 12/12/2022] Open
Abstract
Influenza A viruses (IAV) can cause severe global pandemic outbreaks. The currently licensed antiviral drugs are not very effective and prone to viral resistance. Thus, novel effective and broadly active drugs are urgently needed. We have identified the cellular Raf/MEK/ERK signaling cascade as crucial for IAV replication and suitable target for an antiviral intervention. Since this signaling cascade is aberrantly activated in many human cancers, several clinically approved inhibitors of Raf and MEK are now available. Here we explored the anti-IAV action of the licensed B-RafV600E inhibitor Vemurafenib. Treatment of B-RafWT cells with Vemurafenib induced a hyperactivation of the Raf/MEK/ERK cascade rather than inhibiting its activation upon IAV infection. Despite this hyperactivation, which has also been confirmed by others, Vemurafenib still strongly limited IAV-induced activation of other signaling cascades especially of p38 and JNK mitogen-activated protein kinase (MAPK) pathways. Most interestingly, Vemurafenib inhibited virus-induced apoptosis via impaired expression of apoptosis-inducing cytokines and led to hampered viral protein expression most likely due to the decreased activation of p38 and JNK MAPK. These multiple actions resulted in a profound and broadly active inhibition of viral replication, up to a titer reduction of three orders of a magnitude. Thus, while Vemurafenib did not act similar to MEK inhibitors, it displays strong antiviral properties via a distinct and multi-target mode of action.
Collapse
Affiliation(s)
- Magdalena Holzberg
- Institute of Virology Muenster, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Yvonne Boergeling
- Institute of Virology Muenster, Westfaelische Wilhelms-University Muenster, Muenster, Germany
- Cluster of Excellence Cells in Motion, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Tobias Schräder
- Institute of Virology Muenster, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Stephan Ludwig
- Institute of Virology Muenster, Westfaelische Wilhelms-University Muenster, Muenster, Germany
- Cluster of Excellence Cells in Motion, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| | - Christina Ehrhardt
- Institute of Virology Muenster, Westfaelische Wilhelms-University Muenster, Muenster, Germany
- Cluster of Excellence Cells in Motion, Westfaelische Wilhelms-University Muenster, Muenster, Germany
| |
Collapse
|
40
|
Cheng Y, Ma J, Liu Y, Gao Q, Yan Y, Wang H, Ding C, Sun J. Chicken TBK1 interacts with STING and is involved in IFN-β signaling regulation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 77:200-209. [PMID: 28837824 DOI: 10.1016/j.dci.2017.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/19/2017] [Accepted: 08/19/2017] [Indexed: 06/07/2023]
Abstract
TANK-binding kinase 1 (TBK1) is an essential serine/threonine-protein kinase required for the Toll-like receptor (TLR)- and retinoic acid-inducible gene I (RIG-I) -mediated induction of type I IFN. Through endogenous Co-IP and LC-MS/MS, we identified chicken TBK1 (chTBK1) as a chSTING-interactive protein. Through exogenous Co-IP assay in transfected cells, we confirmed the interaction between chSTING and chTBK1. To better understand the biological role of chTBK1 in the chSTING-mediated IFN pathway, we cloned the chTBK1 and investigated its biological functions. Quantitative RT-PCR showed that chTBK1 mRNA was widely expressed in different tissues. The overexpression of chTBK1 in DF-1 cells induced the expression of IFN-β and ISGs and inhibited AIV viral replication. We identified indispensable domains of chTBK1 on IFN-β production via the generation of various chTBK1 mutant forms. Together, we identified the chTBK1 as a chSTING interactive protein and concluded that chTBK1 is involved in chSTING-triggered IFN-β signaling in chicken cells.
Collapse
Affiliation(s)
- Yuqiang Cheng
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jingjiao Ma
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yunxia Liu
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Quanxin Gao
- The Center for Disease Prevention and Control of Baoshan, Shanghai 201901, China
| | - Yaxian Yan
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hengan Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Jianhe Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
41
|
Transient activation of the PI3K/Akt pathway promotes Newcastle disease virus replication and enhances anti-apoptotic signaling responses. Oncotarget 2017; 8:23551-23563. [PMID: 28423596 PMCID: PMC5410326 DOI: 10.18632/oncotarget.15796] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/06/2017] [Indexed: 01/22/2023] Open
Abstract
Viral infection activates a host's cellular phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, which is involved in cell differentiation, growth, survival, and apoptosis. To elucidate molecular mechanisms in the pathogenesis of Newcastle disease virus (NDV), we demonstrated that NDV transiently activates the PI3K/Akt pathway in chicken cells at an early phase of infection. Its activation was observed as early as 15 min post-infection and gradually weakened after 24 h. Incubating cells with a PI3K inhibitor, LY294002 or wortmannin, prior to NDV infection decreased NDV progeny yields and suppressed Akt phosphorylation at early times post-infection. Akt activation is triggered by NDV-GM or NDV-F48E9 and is abolished by methyl β-cyclodextrin and chlorpromazine. Treatment following NDV-La Sota infection had no obvious effect. However, inhibiting PI3K activation promoted apoptotic responses during an early stage of NDV infection. The pan caspase inhibitor ZVAD-FMK mitigated the reduction in Akt phosphorylation by inhibiting PI3K activation, which indicates the signaling pathway promotes cell survival and, in turn, facilitates viral replication. By suppressing premature apoptosis upon NDV infection, the PI3K/Akt pathway enhances the anti-apoptotic response.
Collapse
|
42
|
Alsuwaidi AR, George JA, Almarzooqi S, Hartwig SM, Varga SM, Souid AK. Sirolimus alters lung pathology and viral load following influenza A virus infection. Respir Res 2017; 18:136. [PMID: 28693498 PMCID: PMC5504865 DOI: 10.1186/s12931-017-0618-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022] Open
Abstract
Background Inhibitors of mTOR, such as sirolimus, have been shown to induce thymus involution and inflammatory lung disease in mice. The latter effect supports the role of this serine/threonine kinase in ameliorating lung inflammation. Other studies have shown sirolimus reduces/delays lung disease associated with various strains of influenza A virus (IAV). Thus, the effects of mTOR inhibitors on influenza infection deserve further studies. Methods Here, we examined the changes in lung viral copies, pathology and pulmonary function associated with IAV (A/PR/8/34) infection in mice treated with sirolimus. Results Body weight loss peaked between days 6–11 post-infection and was more severe in IAV-infected mice that were administered sirolimus as compared to mice that received IAV alone (p = 0.030). Natural log viral gene copies, mean ± SD per mg lung tissue, in IAV-infected mice that were administered sirolimus were 17.31 ± 1.27 on day 4, 19.31 ± 7.46 on day 10, and 0 on day 25. The corresponding number of copies in mice that received IAV alone were 18.56 ± 0.95 on day 4 (p = 0.132), 1.52 ± 1.39 on day 10 (p = 0.008), and 0 on day 25. Lung pathology was evident on days 4, 10, and 25 post infection, with mean ± SD inflammatory score of 9.0 ± 4.5 in IAV-infected mice that were administered sirolimus, as compared to 11.5 ± 4.5 (p = 0.335) in mice received IAV alone (maximum score, 26.0). Impaired lung function was evident in IAV-infected mice on days 4 and 10, as demonstrated by increased airway resistance and decreased compliance. Conclusions In this model, the effects of sirolimus on influenza infection included severe weight loss and modified viral replication, respiratory function and lung inflammation. The adverse events associated with sirolimus treatment are consistent with its potent immunosuppressive activity and, thus, preclude its use in IAV infection.
Collapse
Affiliation(s)
- Ahmed R Alsuwaidi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Junu A George
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Saeeda Almarzooqi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Stacey M Hartwig
- Department of Microbiology & Immunology, Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa, USA
| | - Steven M Varga
- Department of Microbiology & Immunology, Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa, USA
| | - Abdul-Kader Souid
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
43
|
Sprouty-Related Ena/Vasodilator-Stimulated Phosphoprotein Homology 1-Domain-Containing Protein-2 Critically Regulates Influenza A Virus-Induced Pneumonia. Crit Care Med 2017; 44:e530-43. [PMID: 26757161 DOI: 10.1097/ccm.0000000000001562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Influenza A virus causes acute respiratory infections that induce annual epidemics and occasional pandemics. Although a number of studies indicated that the virus-induced intracellular signaling events are important in combating influenza virus infection, the mechanism how specific molecule plays a critical role among various intracellular signaling events remains unknown. Raf/MEK/extracellular signal-regulated kinase cascade is one of the key signaling pathways during influenza virus infection, and the Sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein has recently been identified as a negative regulator of Raf-dependent extracellular signal-regulated kinase activation. Here, we examined the role of Raf/MEK/extracellular signal-regulated kinase cascade through sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein in influenza A viral infection because the expression of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein was significantly enhanced in human influenza viral-induced pneumonia autopsy samples. DESIGN Prospective animal trial. SETTING Research laboratory. SUBJECTS Wild-type and sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 knockout mice inoculated with influenza A. INTERVENTIONS Wild-type or sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 knockout mice were infected by intranasal inoculation of influenza A (A/PR/8). An equal volume of phosphate-buffered saline was inoculated intranasally into mock-infected mice. MEASUREMENTS AND MAIN RESULTS Influenza A infection of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 knockout mice led to higher mortality with greater viral load, excessive inflammation, and enhanced cytokine production than wild-type mice. Administration of MEK inhibitor, U0126, improved mortality and reduced both viral load and cytokine levels. Furthermore, bone marrow chimeras indicated that influenza A-induced lung pathology was most severe when sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 expression was lacking in nonimmune cell populations. Furthermore, microarray analysis revealed knockdown of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 led to enhanced phosphatidylinositol 3-kinase signaling pathway, resulting that viral clearance was regulated by sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 expression through the phosphatidylinositol 3-kinase signaling pathway in murine lung epithelial cells. CONCLUSIONS These data support an important function of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 in controlling influenza virus-induced pneumonia and viral replication. Sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 may be a novel therapeutic target for controlling the immune response against influenza influenza A virus infection.
Collapse
|
44
|
Kedzierski L, Tate MD, Hsu AC, Kolesnik TB, Linossi EM, Dagley L, Dong Z, Freeman S, Infusini G, Starkey MR, Bird NL, Chatfield SM, Babon JJ, Huntington N, Belz G, Webb A, Wark PA, Nicola NA, Xu J, Kedzierska K, Hansbro PM, Nicholson SE. Suppressor of cytokine signaling (SOCS)5 ameliorates influenza infection via inhibition of EGFR signaling. eLife 2017; 6. [PMID: 28195529 PMCID: PMC5354519 DOI: 10.7554/elife.20444] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/06/2017] [Indexed: 12/11/2022] Open
Abstract
Influenza virus infections have a significant impact on global human health. Individuals with suppressed immunity, or suffering from chronic inflammatory conditions such as COPD, are particularly susceptible to influenza. Here we show that suppressor of cytokine signaling (SOCS) five has a pivotal role in restricting influenza A virus in the airway epithelium, through the regulation of epidermal growth factor receptor (EGFR). Socs5-deficient mice exhibit heightened disease severity, with increased viral titres and weight loss. Socs5 levels were differentially regulated in response to distinct influenza viruses (H1N1, H3N2, H5N1 and H11N9) and were reduced in primary epithelial cells from COPD patients, again correlating with increased susceptibility to influenza. Importantly, restoration of SOCS5 levels restricted influenza virus infection, suggesting that manipulating SOCS5 expression and/or SOCS5 targets might be a novel therapeutic approach to influenza.
Collapse
Affiliation(s)
- Lukasz Kedzierski
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia.,Monash University, Clayton, Australia
| | - Alan C Hsu
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Tatiana B Kolesnik
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Edmond M Linossi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Laura Dagley
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Zhaoguang Dong
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Sarah Freeman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Giuseppe Infusini
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Nicola L Bird
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Australia
| | - Simon M Chatfield
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Nicholas Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Gabrielle Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Andrew Webb
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Peter Ab Wark
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Nicos A Nicola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Jianqing Xu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Australia
| | - Philip M Hansbro
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| |
Collapse
|
45
|
Samir M, Vaas LAI, Pessler F. MicroRNAs in the Host Response to Viral Infections of Veterinary Importance. Front Vet Sci 2016; 3:86. [PMID: 27800484 PMCID: PMC5065965 DOI: 10.3389/fvets.2016.00086] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/12/2016] [Indexed: 12/13/2022] Open
Abstract
The discovery of small regulatory non-coding RNAs has been an exciting advance in the field of genomics. MicroRNAs (miRNAs) are endogenous RNA molecules, approximately 22 nucleotides in length, that regulate gene expression, mostly at the posttranscriptional level. MiRNA profiling technologies have made it possible to identify and quantify novel miRNAs and to study their regulation and potential roles in disease pathogenesis. Although miRNAs have been extensively investigated in viral infections of humans, their implications in viral diseases affecting animals of veterinary importance are much less understood. The number of annotated miRNAs in different animal species is growing continuously, and novel roles in regulating host–pathogen interactions are being discovered, for instance, miRNA-mediated augmentation of viral transcription and replication. In this review, we present an overview of synthesis and function of miRNAs and an update on the current state of research on host-encoded miRNAs in the genesis of viral infectious diseases in their natural animal host as well as in selected in vivo and in vitro laboratory models.
Collapse
Affiliation(s)
- Mohamed Samir
- TWINCORE, Center for Experimental and Clinical Infection Research, Hannover, Germany; Department of Zoonoses, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Lea A I Vaas
- TWINCORE, Center for Experimental and Clinical Infection Research , Hannover , Germany
| | - Frank Pessler
- TWINCORE, Center for Experimental and Clinical Infection Research, Hannover, Germany; Helmholtz Center for Infection Research, Braunschweig, Germany
| |
Collapse
|
46
|
Meta- and Orthogonal Integration of Influenza "OMICs" Data Defines a Role for UBR4 in Virus Budding. Cell Host Microbe 2016; 18:723-35. [PMID: 26651948 DOI: 10.1016/j.chom.2015.11.002] [Citation(s) in RCA: 686] [Impact Index Per Article: 76.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/06/2015] [Accepted: 11/10/2015] [Indexed: 12/24/2022]
Abstract
Several systems-level datasets designed to dissect host-pathogen interactions during influenza A infection have been reported. However, apparent discordance among these data has hampered their full utility toward advancing mechanistic and therapeutic knowledge. To collectively reconcile these datasets, we performed a meta-analysis of data from eight published RNAi screens and integrated these data with three protein interaction datasets, including one generated within the context of this study. Further integration of these data with global virus-host interaction analyses revealed a functionally validated biochemical landscape of the influenza-host interface, which can be queried through a simplified and customizable web portal (http://www.metascape.org/IAV). Follow-up studies revealed that the putative ubiquitin ligase UBR4 associates with the viral M2 protein and promotes apical transport of viral proteins. Taken together, the integrative analysis of influenza OMICs datasets illuminates a viral-host network of high-confidence human proteins that are essential for influenza A virus replication.
Collapse
|
47
|
Hui KPY, Li HS, Cheung MC, Chan RWY, Yuen KM, Mok CKP, Nicholls JM, Peiris JSM, Chan MCW. Highly pathogenic avian influenza H5N1 virus delays apoptotic responses via activation of STAT3. Sci Rep 2016; 6:28593. [PMID: 27344974 PMCID: PMC4921847 DOI: 10.1038/srep28593] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/06/2016] [Indexed: 12/25/2022] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5N1 virus continues to pose pandemic threat, but there is a lack of understanding of its pathogenesis. We compared the apoptotic responses triggered by HPAI H5N1 and low pathogenic H1N1 viruses using physiologically relevant respiratory epithelial cells. We demonstrated that H5N1 viruses delayed apoptosis in primary human bronchial and alveolar epithelial cells (AECs) compared to H1N1 virus. Both caspase-8 and -9 were activated by H5N1 and H1N1 viruses in AECs, while H5N1 differentially up-regulated TRAIL. H5N1-induced apoptosis was reduced by TRAIL receptor silencing. More importantly, STAT3 knock-down increased apoptosis by H5N1 infection suggesting that H5N1 virus delays apoptosis through activation of STAT3. Taken together, we demonstrate that STAT3 is involved in H5N1-delayed apoptosis compared to H1N1. Since delay in apoptosis prolongs the duration of virus replication and production of pro-inflammatory cytokines and TRAIL from H5N1-infected cells, which contribute to orchestrate cytokine storm and tissue damage, our results suggest that STAT3 may play a previously unsuspected role in H5N1 pathogenesis.
Collapse
Affiliation(s)
- Kenrie P. Y. Hui
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hung Sing Li
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Man Chun Cheung
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Renee W. Y. Chan
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kit M. Yuen
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chris K. P. Mok
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The HKU-Pasteur Research Pole, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - John M. Nicholls
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - J. S. Malik Peiris
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michael C. W. Chan
- Centre of Influenza Research and School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
48
|
Role of Host Type IA Phosphoinositide 3-Kinase Pathway Components in Invasin-Mediated Internalization of Yersinia enterocolitica. Infect Immun 2016; 84:1826-1841. [PMID: 27068087 DOI: 10.1128/iai.00142-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/03/2016] [Indexed: 02/07/2023] Open
Abstract
Many bacterial pathogens subvert mammalian type IA phosphoinositide 3-kinase (PI3K) in order to induce their internalization into host cells. How PI3K promotes internalization is not well understood. Also unclear is whether type IA PI3K affects different pathogens through similar or distinct mechanisms. Here, we performed an RNA interference (RNAi)-based screen to identify components of the type IA PI3K pathway involved in invasin-mediated entry of Yersinia enterocolitica, an enteropathogen that causes enteritis and lymphadenitis. The 69 genes targeted encode known upstream regulators or downstream effectors of PI3K. A similar RNAi screen was previously performed with the food-borne bacterium Listeria monocytogenes The results of the screen with Y. enterocolitica indicate that at least nine members of the PI3K pathway are needed for invasin-mediated entry. Several of these proteins, including centaurin-α1, Dock180, focal adhesion kinase (FAK), Grp1, LL5α, LL5β, and PLD2 (phospholipase D2), were recruited to sites of entry. In addition, centaurin-α1, FAK, PLD2, and mTOR were required for remodeling of the actin cytoskeleton during entry. Six of the human proteins affecting invasin-dependent internalization also promote InlB-mediated entry of L. monocytogenes Our results identify several host proteins that mediate invasin-induced effects on the actin cytoskeleton and indicate that a subset of PI3K pathway components promote internalization of both Y. enterocolitica and L. monocytogenes.
Collapse
|
49
|
Thulasi Raman SN, Zhou Y. Networks of Host Factors that Interact with NS1 Protein of Influenza A Virus. Front Microbiol 2016; 7:654. [PMID: 27199973 PMCID: PMC4855030 DOI: 10.3389/fmicb.2016.00654] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/19/2016] [Indexed: 11/13/2022] Open
Abstract
Pigs are an important host of influenza A viruses due to their ability to generate reassortant viruses with pandemic potential. NS1 protein of influenza A viruses is a key virulence factor and a major antagonist of innate immune responses. It is also involved in enhancing viral mRNA translation and regulation of virus replication. Being a protein with pleiotropic functions, NS1 has a variety of cellular interaction partners. Hence, studies on swine influenza viruses (SIV) and identification of swine influenza NS1-interacting host proteins is of great interest. Here, we constructed a recombinant SIV carrying a Strep-tag in the NS1 protein and infected primary swine respiratory epithelial cells (SRECs) with this virus. The Strep-tag sequence in the NS1 protein enabled us to purify intact, the NS1 protein and its interacting protein complex specifically. We identified cellular proteins present in the purified complex by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and generated a dataset of these proteins. 445 proteins were identified by LC-MS/MS and among them 192 proteins were selected by setting up a threshold based on MS parameters. The selected proteins were analyzed by bioinformatics and were categorized as belonging to different functional groups including translation, RNA processing, cytoskeleton, innate immunity, and apoptosis. Protein interaction networks were derived using these data and the NS1 interactions with some of the specific host factors were verified by immunoprecipitation. The novel proteins and the networks revealed in our study will be the potential candidates for targeted study of the molecular interaction of NS1 with host proteins, which will provide insights into the identification of new therapeutic targets to control influenza infection and disease pathogenesis.
Collapse
Affiliation(s)
- Sathya N Thulasi Raman
- Vaccine and Infectious Disease Organization - International Vaccine Centre, University of Saskatchewan, SaskatoonSK, Canada; Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, SaskatoonSK, Canada
| | - Yan Zhou
- Vaccine and Infectious Disease Organization - International Vaccine Centre, University of Saskatchewan, SaskatoonSK, Canada; Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, SaskatoonSK, Canada
| |
Collapse
|
50
|
Chemical Genomics Identifies the PERK-Mediated Unfolded Protein Stress Response as a Cellular Target for Influenza Virus Inhibition. mBio 2016; 7:e00085-16. [PMID: 27094326 PMCID: PMC4850254 DOI: 10.1128/mbio.00085-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Influenza A viruses generate annual epidemics and occasional pandemics of respiratory disease with important consequences for human health and the economy. Therefore, a large effort has been devoted to the development of new anti-influenza virus drugs directed to viral targets, as well as to the identification of cellular targets amenable to anti-influenza virus therapy. Here we have addressed the identification of such potential cellular targets by screening collections of drugs approved for human use. We reasoned that screening with a green fluorescent protein-based recombinant replicon system would identify cellular targets involved in virus transcription/replication and/or gene expression and hence address an early stage of virus infection. By using such a strategy, we identified Montelukast (MK) as an inhibitor of virus multiplication. MK inhibited virus gene expression but did not alter viral RNA synthesis in vitro or viral RNA accumulation in vivo. The low selectivity index of MK prevented its use as an antiviral, but it was sufficient to identify a new cellular pathway suitable for anti-influenza virus intervention. By deep sequencing of RNA isolated from mock- and virus-infected human cells, treated with MK or left untreated, we showed that it stimulates the PERK-mediated unfolded protein stress response. The phosphorylation of PERK was partly inhibited in virus-infected cells but stimulated in MK-treated cells. Accordingly, pharmacological inhibition of PERK phosphorylation led to increased viral gene expression, while inhibition of PERK phosphatase reduced viral protein synthesis. These results suggest the PERK-mediated unfolded protein response as a potential cellular target to modulate influenza virus infection. Influenza A viruses are responsible for annual epidemics and occasional pandemics with important consequences for human health and the economy. The unfolded protein response is a defense mechanism fired by cells when the demand of protein synthesis and folding is excessive, for instance, during an acute virus infection. In this report, we show that influenza virus downregulates the unfolded protein response mediated by the PERK sensor, while Montelukast, a drug used to treat asthma in humans, specifically stimulated this response and downregulated viral protein synthesis and multiplication. Accordingly, we show that PERK phosphorylation was reduced in virus-infected cells and increased in cells treated with Montelukast. Hence, our studies suggest that modulation of the PERK-mediated unfolded protein response is a target for influenza virus inhibition.
Collapse
|