1
|
Roa-Linares VC, Betancur-Galvis LA, González-Cardenete MA, Garcia-Blanco MA, Gallego-Gomez JC. Broad-spectrum antiviral ferruginol analog affects the viral proteins translation and actin remodeling during dengue virus infection. Antiviral Res 2025; 237:106139. [PMID: 40043781 DOI: 10.1016/j.antiviral.2025.106139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Dengue virus infection is the most important arbovirosis around the world. To date, no antiviral drugs have been approved for its treatment. Host-targeted antivirals (HTA) have emerged as a promising strategy, because of their high barrier to resistance. Using plaque-forming unit assays, molecular docking, fluorescence microscopy, image analysis, and molecular/cellular assays, it was found that 18-(phthalimide-2-yl)-ferruginol, a semi-synthetic analog of the bioactive diterpenoid ferruginol, couples with high affinity to RhoA GTPase. In addition, this molecule dramatically reduced actin filament formation and induced cellular morphological changes, when added to cell cultures before or after infection, without effect on microtubules or intermediate filaments. RhoA activation in infected cells was affected when the compound was added after 6 h.p.i. Furthermore, this compound decreased dengue virus-2 (DENV-2) E protein, NS3 protein, and dsRNA as measured by fluorescence microscopy, and changes in the distribution pattern of these viral components. 18-(phthalimide-2-yl)-ferruginol treatment at 6 and 12 h.p.i. reduces the virus yield. Western blot and RT-qPCR assays reveal that this analog decreased viral protein translation. Flow cytometry and wound-healing experiments also hint that cellular effects prompted for this compound do not relate to early apoptotic events and they could be reversible. Overall, our findings strongly suggest that 18-(phthalimide-2-yl)-ferruginol has an HTA mechanism, possibly disrupting the polyprotein translation of DENV-2 via alteration of RhoA-mediated actin remodeling and other related cellular and viral processes.
Collapse
Affiliation(s)
- Vicky C Roa-Linares
- Translational Medicine Group, Medicine Faculty, Institute for Medical Research, Universidad de Antioquia, Medellín, Colombia; Crisalida Research Group, Faculty of Medical and Health Sciences, Universidad de Santander, Cucuta, Colombia.
| | - Liliana A Betancur-Galvis
- Translational Medicine Group, Medicine Faculty, Institute for Medical Research, Universidad de Antioquia, Medellín, Colombia; Group of Investigative Dermatology, Medicine Faculty, Institute for Medical Research, Universidad de Antioquia, Medellín, Colombia
| | - Miguel A González-Cardenete
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas, Avda. de los Naranjos s/n, 46022, Valencia, Spain.
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77550, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Juan C Gallego-Gomez
- Translational Medicine Group, Medicine Faculty, Institute for Medical Research, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
2
|
Upadhyay RK, Kumar K, Vishwakarma VK, Singh N, Narang R, Parakh N, Yadav M, Yadav S, Kumar S, Goyal A, Yadav HN. Delineating the NOX-Mediated Promising Therapeutic Strategies for the Management of Various Cardiovascular Disorders: A Comprehensive Review. Curr Vasc Pharmacol 2025; 23:12-30. [PMID: 39313896 DOI: 10.2174/0115701611308870240910115023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/21/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024]
Abstract
Cardiovascular disorders (CVDs) are reported to occur with very high rates of incidence and exhibit high morbidity and mortality rates across the globe. Therefore, research is focused on searching for novel therapeutic targets involving multiple pathophysiological mechanisms. Oxidative stress plays a critical role in the development and progression of various CVDs, such as hypertension, pulmonary hypertension, heart failure, arrhythmia, atherosclerosis, ischemia- reperfusion injury, and myocardial infarction. Among multiple pathways generating reactive oxygen species (ROS), Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases of the NOX family as the major source of ROS generation and plays an intricate role in the development and progression of CVDs. Therefore, exploring the role of different NADPH oxidase isoforms in various cardiovascular pathologies has attracted attention to current cardiovascular research. Focusing on NADPH oxidases to reduce oxidative stress in managing diverse CVDs may offer unique therapeutic approaches to prevent and treat various heart conditions. The current review article highlights the role of different NADPH oxidase isoforms in the pathophysiology of various CVDs. Moreover, the focus is also to emphasize different experimental studies that utilized various NADPH oxidase isoform modulators to manage other disorders. The present review article considers new avenues for researchers/scientists working in the field of cardiovascular pharmacology utilizing NADPH oxidase isoform modulators.
Collapse
Affiliation(s)
- Rohit Kumar Upadhyay
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kuldeep Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, (Punjab)-147002-India
| | - Rajiv Narang
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neeraj Parakh
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Mayank Yadav
- Department of CTVS, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sangeeta Yadav
- Department of Pharmacology, Dr. B.R. Ambedkar Centre for Biomedical Research, New Delhi, 110085-India
| | - Sachin Kumar
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, 280406, India
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
3
|
Li XM, Xu K, Wang JY, Guo JY, Wang XH, Zeng L, Wan B, Wang J, Chu BB, Yang GY, Pan JJ, Hao WB. The actin cytoskeleton is important for pseudorabies virus infection. Virology 2024; 600:110233. [PMID: 39255726 DOI: 10.1016/j.virol.2024.110233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/06/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
Viruses are dependent on the host factors for their replication and survival. Therefore, identification of host factors that druggable for antiviral development is crucial. The actin cytoskeleton plays an important role in the virus infection. The dynamics change of actin and its function are regulated by multiple actin-associated proteins (AAPs). However, the role and mechanism of various AAPs in the life cycle of virus are still enigmatic. In this study, we analyzed the roles of actin and AAPs in the replication of pseudorabies virus (PRV). Using a library of compounds targeting AAPs, our data found that multiple AAPs, such as Rho-GTPases, Rock, Myosin and Formin were involved in PRV infection. Besides, our result demonstrated that the actin-binding protein Drebrin was also participated in PRV infection. Further studies are necessary to elucidate the molecular mechanism of AAPs in the virus life cycle, in the hope of mining host factors for antiviral developments.
Collapse
Affiliation(s)
- Xin-Man Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Kun Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Jin-Yuan Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Jie-Yuan Guo
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Xiao-Han Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Bo Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Henan University of Animal Husbandry and Economy, Zhengzhou, 450047, China
| | - Jia-Jia Pan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China.
| | - Wen-Bo Hao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
4
|
Vijayakumar P, Mishra A, Deka RP, Pinto SM, Subbannayya Y, Sood R, Prasad TSK, Raut AA. Proteomics Analysis of Duck Lung Tissues in Response to Highly Pathogenic Avian Influenza Virus. Microorganisms 2024; 12:1288. [PMID: 39065055 PMCID: PMC11278641 DOI: 10.3390/microorganisms12071288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 07/28/2024] Open
Abstract
Domestic ducks (Anas platyrhynchos domesticus) are resistant to most of the highly pathogenic avian influenza virus (HPAIV) infections. In this study, we characterized the lung proteome and phosphoproteome of ducks infected with the HPAI H5N1 virus (A/duck/India/02CA10/2011/Agartala) at 12 h, 48 h, and 5 days post-infection. A total of 2082 proteins were differentially expressed and 320 phosphorylation sites mapping to 199 phosphopeptides, corresponding to 129 proteins were identified. The functional annotation of the proteome data analysis revealed the activation of the RIG-I-like receptor and Jak-STAT signaling pathways, which led to the induction of interferon-stimulated gene (ISG) expression. The pathway analysis of the phosphoproteome datasets also confirmed the activation of RIG-I, Jak-STAT signaling, NF-kappa B signaling, and MAPK signaling pathways in the lung tissues. The induction of ISG proteins (STAT1, STAT3, STAT5B, STAT6, IFIT5, and PKR) established a protective anti-viral immune response in duck lung tissue. Further, the protein-protein interaction network analysis identified proteins like AKT1, STAT3, JAK2, RAC1, STAT1, PTPN11, RPS27A, NFKB1, and MAPK1 as the main hub proteins that might play important roles in disease progression in ducks. Together, the functional annotation of the proteome and phosphoproteome datasets revealed the molecular basis of the disease progression and disease resistance mechanism in ducks infected with the HPAI H5N1 virus.
Collapse
Affiliation(s)
- Periyasamy Vijayakumar
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
- Veterinary College and Research Institute, Tamil Nadu Veterinary and Animal Sciences University, Salem 600051, Tamil Nadu, India
| | - Anamika Mishra
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
| | - Ram Pratim Deka
- International Livestock Research Institute, National Agricultural Science Complex, Pusa 110012, New Delhi, India;
| | - Sneha M. Pinto
- Centre for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India; (S.M.P.); (Y.S.)
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Yashwanth Subbannayya
- Centre for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India; (S.M.P.); (Y.S.)
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Richa Sood
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
| | | | - Ashwin Ashok Raut
- Pathogenomics Laboratory, WOAH Reference Lab for Avian Influenza, ICAR—National Institute of High Security Animal Diseases, Bhopal 462022, Madhya Pradesh, India; (P.V.); (A.M.); (R.S.)
| |
Collapse
|
5
|
Role of Host Small GTPases in Apicomplexan Parasite Infection. Microorganisms 2022; 10:microorganisms10071370. [PMID: 35889089 PMCID: PMC9319929 DOI: 10.3390/microorganisms10071370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
The Apicomplexa are obligate intracellular parasites responsible for several important human diseases. These protozoan organisms have evolved several strategies to modify the host cell environment to create a favorable niche for their survival. The host cytoskeleton is widely manipulated during all phases of apicomplexan intracellular infection. Moreover, the localization and organization of host organelles are altered in order to scavenge nutrients from the host. Small GTPases are a class of proteins widely involved in intracellular pathways governing different processes, from cytoskeletal and organelle organization to gene transcription and intracellular trafficking. These proteins are already known to be involved in infection by several intracellular pathogens, including viruses, bacteria and protozoan parasites. In this review, we recapitulate the mechanisms by which apicomplexan parasites manipulate the host cell during infection, focusing on the role of host small GTPases. We also discuss the possibility of considering small GTPases as potential targets for the development of novel host-targeted therapies against apicomplexan infections.
Collapse
|
6
|
miR-142-3p Suppresses Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) Infection by Directly Targeting Rac1. Vet Microbiol 2022; 269:109434. [DOI: 10.1016/j.vetmic.2022.109434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/23/2022] [Accepted: 04/10/2022] [Indexed: 11/20/2022]
|
7
|
Abstract
Herpes simplex virus 1 (HSV-1) infects eye corneal tissues leading to herpetic stromal keratitis (HSK), which is one of the leading causes of blindness. Here in our study, we found that 6-thioguanine (6-TG), a once clinically approved medication for child acute myelogenous leukemia, inhibited multiple strains of HSV-1 infection in vitro and in vivo. 6-TG is more potent than acyclovir (ACV) and ganciclovir (GCV), with the 50% inhibitory concentration (IC50) of 6-TG at 0.104 μM with high stimulation index (SI) (SI = 6,475.48) compared to the IC50 of ACV at 1.253 μM and the IC50 of GCV at 1.257 μM. In addition, 6-TG at 500 μM topically applied to the eyes with HSV-1 infection significantly inhibits HSV-1 replication, alleviates virus-induced HSK pathogenesis, and improves eye conditions. More importantly, 6-TG is effective against ACV-resistant HSV-1 strains, including HSV-1/153 and HSV-1/blue. Knockdown of Rac1 with small interfering RNA (siRNA) negatively affected HSV-1 replication, suggesting that Rac1 facilitated HSV-1 replication. Following HSV-1 infection of human corneal epithelial cells (HCECs), endogenous Rac1 activity was upregulated by glutathione S-transferase (GST) pulldown assay. We further found that Rac1 was highly expressed in the corneal tissue of HSK patients compared to normal individuals. Mechanistic study showed that 6-TG inhibited HSV-1 replication by targeting Rac1 activity in HSV-1 infected cells, and the Rac1 is critical in the pathogenesis of HSK. Our results indicated that 6-TG is a promising therapeutic molecule for the treatment of HSK. IMPORTANCE We reported the discovery of 6-TG inhibition of HSV-1 infection and its inhibitory roles in HSK both in vitro and in vivo. 6-TG was shown to possess at least 10× more potent inhibitory activity against HSV-1 than ACV and GCV and, more importantly, inhibit ACV/GCV-resistant mutant viruses. Animal model studies showed that gel-formulated 6-TG topically applied to eyes locally infected with HSV-1 could significantly inhibit HSV-1 replication, alleviate virus-induced HSK pathogenesis, and improve eye conditions. Further study showed that HSV-1 infection upregulated Rac1 expression, and knockdown of Rac1 using siRNA markedly restricted HSV-1 replication, suggesting that Rac1 is required for HSV-1 replication. In addition, we also documented that Rac1 is highly expressed in corneal tissues from HSK patients, indicating that Rac1 is associated with HSK pathogenesis. In view of the high potency of 6-TG, low cytotoxicity, targeting a distinct therapeutic target, we suggest that 6-TG is a potential candidate for development as a therapeutic agent for HSK therapy.
Collapse
|
8
|
Lim ZQ, Ng QY, Oo Y, Chu JJH, Ng SY, Sze SK, Alonso S. Enterovirus-A71 exploits peripherin and Rac1 to invade the central nervous system. EMBO Rep 2021; 22:e51777. [PMID: 33871166 DOI: 10.15252/embr.202051777] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/02/2021] [Accepted: 03/10/2021] [Indexed: 12/28/2022] Open
Abstract
Enterovirus-A71 (EV-A71) has been associated with severe neurological forms of hand, foot, and mouth disease (HFMD). EV-A71 infects motor neurons at neuromuscular junctions (NMJs) to invade the central nervous system (CNS). Here, we investigate the role of peripherin (PRPH) during EV-A71 infection, a type III intermediate neurofilament involved in neurodegenerative conditions. In mice infected with EV-A71, PRPH co-localizes with viral particles in the muscles at NMJs and in the spinal cord. In motor neuron-like and neuroblastoma cell lines, surface-expressed PRPH facilitates viral entry, while intracellular PRPH influences viral genome replication through interactions with structural and non-structural viral components. Importantly, PRPH does not play a role during infection with coxsackievirus A16, another causative agent of HFMD rarely associated with neurological complications, suggesting that EV-A71 ability to exploit PRPH represents a unique attribute for successful CNS invasion. Finally, we show that EV-A71 also exploits some of the many PRPH-interacting partners. Of these, small GTP-binding protein Rac1 represents a potential druggable host target to limit neuroinvasion of EV-A71.
Collapse
Affiliation(s)
- Ze Qin Lim
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Qing Yong Ng
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Yukei Oo
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Justin Jang Hann Chu
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shi Yan Ng
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Siu Kwan Sze
- Proteomics and Mass Spectrometry Services Core Facility, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sylvie Alonso
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
den Hartog G, Butcher LD, Ablack AL, Pace LA, Ablack JNG, Xiong R, Das S, Stappenbeck TS, Eckmann L, Ernst PB, Crowe SE. Apurinic/Apyrimidinic Endonuclease 1 Restricts the Internalization of Bacteria Into Human Intestinal Epithelial Cells Through the Inhibition of Rac1. Front Immunol 2021; 11:553994. [PMID: 33603730 PMCID: PMC7884313 DOI: 10.3389/fimmu.2020.553994] [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: 09/01/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Pathogenic intestinal bacteria lead to significant disease in humans. Here we investigated the role of the multifunctional protein, Apurinic/apyrimidinic endonuclease 1 (APE1), in regulating the internalization of bacteria into the intestinal epithelium. Intestinal tumor-cell lines and primary human epithelial cells were infected with Salmonella enterica serovar Typhimurium or adherent-invasive Escherichia coli. The effects of APE1 inhibition on bacterial internalization, the regulation of Rho GTPase Rac1 as well as the epithelial cell barrier function were assessed. Increased numbers of bacteria were present in APE1-deficient colonic tumor cell lines and primary epithelial cells. Activation of Rac1 was augmented following infection but negatively regulated by APE1. Pharmacological inhibition of Rac1 reversed the increase in intracellular bacteria in APE1-deficient cells whereas overexpression of constitutively active Rac1 augmented the numbers in APE1-competent cells. Enhanced numbers of intracellular bacteria resulted in the loss of barrier function and a delay in its recovery. Our data demonstrate that APE1 inhibits the internalization of invasive bacteria into human intestinal epithelial cells through its ability to negatively regulate Rac1. This activity also protects epithelial cell barrier function.
Collapse
Affiliation(s)
- Gerco den Hartog
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Lindsay D Butcher
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Amber L Ablack
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Laura A Pace
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Jailal N G Ablack
- Department of Medicine, Division of Rheumatology, University of California San Diego, La Jolla, CA, United States
| | - Richard Xiong
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Soumita Das
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States
| | | | - Lars Eckmann
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Peter B Ernst
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Center for Mucosal Immunology, Allergy and Vaccine Development, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Department of Immunology, Chiba University, Chiba, Japan
| | - Sheila E Crowe
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States.,Division of ImmunoBiology, Washington University, St. Louis, MO, United States
| |
Collapse
|
10
|
Salmikangas S, Laiho JE, Kalander K, Laajala M, Honkimaa A, Shanina I, Oikarinen S, Horwitz MS, Hyöty H, Marjomäki V. Detection of Viral -RNA and +RNA Strands in Enterovirus-Infected Cells and Tissues. Microorganisms 2020; 8:microorganisms8121928. [PMID: 33291747 PMCID: PMC7761939 DOI: 10.3390/microorganisms8121928] [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: 10/30/2020] [Revised: 11/24/2020] [Accepted: 12/02/2020] [Indexed: 11/16/2022] Open
Abstract
The current methods to study the distribution and dynamics of viral RNA molecules inside infected cells are not ideal, as electron microscopy and immunohistochemistry can only detect mature virions, and quantitative real-time PCR does not reveal localized distribution of RNAs. We demonstrated here the branched DNA in situ hybridization (bDNA ISH) technology to study both the amount and location of the emerging -RNA and +RNA during acute and persistent enterovirus infections. According to our results, the replication of the viral RNA started 2-3 h after infection and the translation shortly after at 3-4 h post-infection. The replication hotspots with newly emerging -RNA were located quite centrally in the cell, while the +RNA production and most likely virion assembly took place in the periphery of the cell. We also discovered that the pace of replication of -RNA and +RNA strands was almost identical, and -RNA was absent during antiviral treatments. ViewRNA ISH with our custom probes also showed a good signal during acute and persistent enterovirus infections in cell and mouse models. Considering these results, along with the established bDNA FISH protocol modified by us, the effects of antiviral drugs and the emergence of enterovirus RNAs in general can be studied more effectively.
Collapse
Affiliation(s)
- Sami Salmikangas
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Survontie 9C, FI-40500 Jyväskylä, Finland; (S.S.); (K.K.); (M.L.)
| | - Jutta E. Laiho
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland; (J.E.L.); (A.H.); (S.O.); (H.H.)
| | - Kerttu Kalander
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Survontie 9C, FI-40500 Jyväskylä, Finland; (S.S.); (K.K.); (M.L.)
| | - Mira Laajala
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Survontie 9C, FI-40500 Jyväskylä, Finland; (S.S.); (K.K.); (M.L.)
| | - Anni Honkimaa
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland; (J.E.L.); (A.H.); (S.O.); (H.H.)
| | - Iryna Shanina
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T1Z3, Canada; (I.S.); (M.S.H.)
| | - Sami Oikarinen
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland; (J.E.L.); (A.H.); (S.O.); (H.H.)
| | - Marc S. Horwitz
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T1Z3, Canada; (I.S.); (M.S.H.)
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland; (J.E.L.); (A.H.); (S.O.); (H.H.)
| | - Varpu Marjomäki
- Department of Biological and Environmental Science/Nanoscience Center, University of Jyväskylä, Survontie 9C, FI-40500 Jyväskylä, Finland; (S.S.); (K.K.); (M.L.)
- Correspondence: ; Tel.: +358-405634422
| |
Collapse
|
11
|
O’Dowd K, Emam M, El Khili MR, Emad A, Ibeagha-Awemu EM, Gagnon CA, Barjesteh N. Distinct miRNA Profile of Cellular and Extracellular Vesicles Released from Chicken Tracheal Cells Following Avian Influenza Virus Infection. Vaccines (Basel) 2020; 8:vaccines8030438. [PMID: 32764349 PMCID: PMC7565416 DOI: 10.3390/vaccines8030438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023] Open
Abstract
Innate responses provide the first line of defense against viral infections, including the influenza virus at mucosal surfaces. Communication and interaction between different host cells at the early stage of viral infections determine the quality and magnitude of immune responses against the invading virus. The release of membrane-encapsulated extracellular vesicles (EVs), from host cells, is defined as a refined system of cell-to-cell communication. EVs contain a diverse array of biomolecules, including microRNAs (miRNAs). We hypothesized that the activation of the tracheal cells with different stimuli impacts the cellular and EV miRNA profiles. Chicken tracheal rings were stimulated with polyI:C and LPS from Escherichia coli 026:B6 or infected with low pathogenic avian influenza virus H4N6. Subsequently, miRNAs were isolated from chicken tracheal cells or from EVs released from chicken tracheal cells. Differentially expressed (DE) miRNAs were identified in treated groups when compared to the control group. Our results demonstrated that there were 67 up-regulated miRNAs, 157 down-regulated miRNAs across all cellular and EV samples. In the next step, several genes or pathways targeted by DE miRNAs were predicted. Overall, this study presented a global miRNA expression profile in chicken tracheas in response to avian influenza viruses (AIV) and toll-like receptor (TLR) ligands. The results presented predicted the possible roles of some DE miRNAs in the induction of antiviral responses. The DE candidate miRNAs, including miR-146a, miR-146b, miR-205a, miR-205b and miR-449, can be investigated further for functional validation studies and to be used as novel prophylactic and therapeutic targets in tailoring or enhancing antiviral responses against AIV.
Collapse
Affiliation(s)
- Kelsey O’Dowd
- Research Group on Infectious Diseases in Production Animals (GREMIP), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.O.); (C.A.G.)
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | - Mehdi Emam
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- McGill University Research Centre on Complex Traits (MRCCT), Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, QC H3G 0B1, Canada
| | - Mohamed Reda El Khili
- Department of Electrical and Computer Engineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (M.R.E.K.); (A.E.)
| | - Amin Emad
- Department of Electrical and Computer Engineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (M.R.E.K.); (A.E.)
| | - Eveline M. Ibeagha-Awemu
- Sherbrooke Research & Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC J1M 0C8, Canada;
| | - Carl A. Gagnon
- Research Group on Infectious Diseases in Production Animals (GREMIP), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.O.); (C.A.G.)
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | - Neda Barjesteh
- Research Group on Infectious Diseases in Production Animals (GREMIP), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada; (K.O.); (C.A.G.)
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- Correspondence: ; Tel.: +1-450-773-8521 (ext. 33191)
| |
Collapse
|
12
|
Gray JL, von Delft F, Brennan PE. Targeting the Small GTPase Superfamily through Their Regulatory Proteins. Angew Chem Int Ed Engl 2020; 59:6342-6366. [PMID: 30869179 PMCID: PMC7204875 DOI: 10.1002/anie.201900585] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/11/2019] [Indexed: 12/11/2022]
Abstract
The Ras superfamily of small GTPases are guanine-nucleotide-dependent switches essential for numerous cellular processes. Mutations or dysregulation of these proteins are associated with many diseases, but unsuccessful attempts to target the small GTPases directly have resulted in them being classed as "undruggable". The GTP-dependent signaling of these proteins is controlled by their regulators; guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in the Rho and Rab subfamilies, guanine nucleotide dissociation inhibitors (GDIs). This review covers the recent small molecule and biologics strategies to target the small GTPases through their regulators. It seeks to critically re-evaluate recent chemical biology practice, such as the presence of PAINs motifs and the cell-based readout using compounds that are weakly potent or of unknown specificity. It highlights the vast scope of potential approaches for targeting the small GTPases in the future through their regulatory proteins.
Collapse
Affiliation(s)
- Janine L. Gray
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOld Road CampusOxfordOX3 7FZUK
- Diamond Light SourceHarwell Science and Innovation CampusDidcotOX11 0QXUK
| | - Frank von Delft
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Diamond Light SourceHarwell Science and Innovation CampusDidcotOX11 0QXUK
- Department of BiochemistryUniversity of JohannesburgAuckland Park2006South Africa
| | - Paul E. Brennan
- Structural Genomics ConsortiumUniversity of Oxford, NDMRBOld Road CampusOxfordOX3 7DQUK
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOld Road CampusOxfordOX3 7FZUK
- Alzheimer's Research (UK) Oxford Drug Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| |
Collapse
|
13
|
Gray JL, Delft F, Brennan PE. Targeting der kleinen GTPasen über ihre regulatorischen Proteine. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201900585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Janine L. Gray
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Target Discovery InstituteNuffield Department of MedicineUniversity of Oxford Old Road Campus Oxford OX3 7FZ Großbritannien
- Diamond Light Source Harwell Science and Innovation Campus Didcot OX11 0QX Großbritannien
| | - Frank Delft
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Diamond Light Source Harwell Science and Innovation Campus Didcot OX11 0QX Großbritannien
- Department of BiochemistryUniversity of Johannesburg Auckland Park 2006 Südafrika
| | - Paul E. Brennan
- Structural Genomics ConsortiumUniversity of Oxford, NDMRB Old Road Campus Oxford OX3 7DQ Großbritannien
- Target Discovery InstituteNuffield Department of MedicineUniversity of Oxford Old Road Campus Oxford OX3 7FZ Großbritannien
- Alzheimer's Research (UK) Oxford Drug Discovery InstituteNuffield Department of MedicineUniversity of Oxford Oxford OX3 7FZ Großbritannien
| |
Collapse
|
14
|
Guo H, Chen G, Gao M, Wang R, Liu Y, Yu F. Imaging of Endogenous Hydrogen Peroxide during the Process of Cell Mitosis and Mouse Brain Development with a Near-Infrared Ratiometric Fluorescent Probe. Anal Chem 2018; 91:1203-1210. [DOI: 10.1021/acs.analchem.8b05326] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hailong Guo
- The Key Laboratory of Life-Organic Analysis, Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Guang Chen
- The Key Laboratory of Life-Organic Analysis, Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- Laboratory of Tibetan Medicine Research & Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources, Northwest Institute of Plateau Biology, Chinese Academy of Science, Xining 810001, China
| | - Min Gao
- The Key Laboratory of Life-Organic Analysis, Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Rui Wang
- The Key Laboratory of Life-Organic Analysis, Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Yuxia Liu
- The Key Laboratory of Life-Organic Analysis, Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
| | - Fabiao Yu
- The Key Laboratory of Life-Organic Analysis, Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, College of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- Institute of Functional Materials and Molecular Imaging, College of Clinical Medicine, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
15
|
Bao S, Jia L, Zhou X, Zhang ZG, Wu HWL, Yu Z, Ng G, Fan Y, Wong DSM, Huang S, Wang To KK, Yuen KY, Yeung ML, Song YQ. Integrated analysis of mRNA-seq and miRNA-seq for host susceptibilities to influenza A (H7N9) infection in inbred mouse lines. Funct Integr Genomics 2018; 18:411-424. [PMID: 29564647 DOI: 10.1007/s10142-018-0602-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/12/2018] [Indexed: 02/05/2023]
Abstract
Host genetic factors play an important role in diverse host outcomes after influenza A (H7N9) infection. Studying differential responses of inbred mouse lines with distinct genetic backgrounds to influenza virus infection could substantially increase our understanding of the contributory roles of host genetic factors to disease severity. Here, we utilized an integrated approach of mRNA-seq and miRNA-seq to investigate the transcriptome expression and regulation of host genes in C57BL/6J and DBA/2J mouse strains during influenza virus infection. The differential pathogenicity of influenza virus in C57BL/6J and DBA/2J has been fully demonstrated through immunohistochemical staining, histopathological analyses, and viral replication assessment. A transcriptional molecular signature correlates to differential host response to infection has been uncovered. With the introduction of temporal expression pattern analysis, we demonstrated that host factors responsible for influenza virus replication and host-virus interaction were significantly enriched in genes exhibiting distinct temporal dynamics between different inbred mouse lines. A combination of time-series expression analysis and temporal expression pattern analysis has provided a list of promising candidate genes for future studies. An integrated miRNA regulatory network from both mRNA-seq and miRNA-seq revealed several regulatory modules responsible for regulating host susceptibilities and disease severity. Overall, a comprehensive framework for analyzing host susceptibilities to influenza infection was established by integrating mRNA-seq and miRNA-seq data of inbred mouse lines. This work suggests novel putative molecular targets for therapeutic interventions in seasonal and pandemic influenza.
Collapse
Affiliation(s)
- Suying Bao
- Schoolof Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Lilong Jia
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Xueya Zhou
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Zhi-Gang Zhang
- Schoolof Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Hazel Wai Lan Wu
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Zhe Yu
- Schoolof Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Gordon Ng
- Schoolof Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Yanhui Fan
- Schoolof Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Dana S M Wong
- Schoolof Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Shishu Huang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kelvin Kai Wang To
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Hong Kong, China
| | - Man Lung Yeung
- Department of Microbiology, The University of Hong Kong, Hong Kong, China.
| | - You-Qiang Song
- Schoolof Biomedical Sciences, The University of Hong Kong, Hong Kong, China.
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China.
- HKU-SIRI/ZIRI, The University of Hong Kong, Hong Kong, China.
- HKU-SUSTech Joint Laboratories of Matrix Biology and Diseases, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
16
|
Yin Y, Chen S, Hakim MS, Wang W, Xu L, Dang W, Qu C, Verhaar AP, Su J, Fuhler GM, Peppelenbosch MP, Pan Q. 6-Thioguanine inhibits rotavirus replication through suppression of Rac1 GDP/GTP cycling. Antiviral Res 2018; 156:92-101. [PMID: 29920300 PMCID: PMC7113846 DOI: 10.1016/j.antiviral.2018.06.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
Rotavirus infection has emerged as an important cause of complications in organ transplantation recipients and might play a role in the pathogenesis of inflammatory bowel disease (IBD). 6-Thioguanine (6-TG) has been widely used as an immunosuppressive drug for organ recipients and treatment of IBD in the clinic. This study aims to investigate the effects and mode-of-action of 6-TG on rotavirus replication. Human intestinal Caco2 cell line, 3D model of human primary intestinal organoids, laboratory rotavirus strain (SA11) and patient-derived rotavirus isolates were used. We have demonstrated that 6-TG significantly inhibits rotavirus replication in these intestinal epithelium models. Importantly, gene knockdown or knockout of Rac1, the cellular target of 6-TG, significantly inhibited rotavirus replication, indicating the supportive role of Rac1 for rotavirus infection. We have further demonstrated that 6-TG can effectively inhibit the active form of Rac1 (GTP-Rac1), which essentially mediates the anti-rotavirus effect of 6-TG. Consistently, ectopic over-expression of GTP-Rac1 facilitates but an inactive Rac1 (N17) or a specific Rac1 inhibitor (NSC23766) inhibits rotavirus replication. In conclusion, we have identified 6-TG as an effective inhibitor of rotavirus replication via the inhibition of Rac1 activation. Thus, for transplantation patients or IBD patients infected with rotavirus or at risk of rotavirus infection, the choice of 6-TG as a treatment appears rational. 6-TG inhibits rotavirus infection. Rac1, the cellular target of 6-TG, supports rotavirus infection. 6-TG inhibits the active form of Rac1 (GTP-Rac1) to exert the anti-rotavirus effect.
Collapse
Affiliation(s)
- Yuebang Yin
- Center for Biomedical Research, Northwest Minzu University, Lanzhou, China; Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands; Institute of Subtropical Agriculture, Chinese Academic of Sciences, Changsha 410125, China
| | - Sunrui Chen
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Mohamad S Hakim
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands; Department of Microbiology, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Wenshi Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Lei Xu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Wen Dang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Changbo Qu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Auke P Verhaar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Junhong Su
- Center for Biomedical Research, Northwest Minzu University, Lanzhou, China; Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands; Medical Faculty, Kunming University of Science and Technology, Kunming, PR China
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Qiuwei Pan
- Center for Biomedical Research, Northwest Minzu University, Lanzhou, China; Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
17
|
Marei H, Malliri A. Rac1 in human diseases: The therapeutic potential of targeting Rac1 signaling regulatory mechanisms. Small GTPases 2017; 8:139-163. [PMID: 27442895 PMCID: PMC5584733 DOI: 10.1080/21541248.2016.1211398] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 12/11/2022] Open
Abstract
Abnormal Rac1 signaling is linked to a number of debilitating human diseases, including cancer, cardiovascular diseases and neurodegenerative disorders. As such, Rac1 represents an attractive therapeutic target, yet the search for effective Rac1 inhibitors is still underway. Given the adverse effects associated with Rac1 signaling perturbation, cells have evolved several mechanisms to ensure the tight regulation of Rac1 signaling. Thus, characterizing these mechanisms can provide invaluable information regarding major cellular events that lead to aberrant Rac1 signaling. Importantly, this information can be utilized to further facilitate the development of effective pharmacological modulators that can restore normal Rac1 signaling. In this review, we focus on the pathological role of Rac1 signaling, highlighting the benefits and potential drawbacks of targeting Rac1 in a clinical setting. Additionally, we provide an overview of available compounds that target key Rac1 regulatory mechanisms and discuss future therapeutic avenues arising from our understanding of these mechanisms.
Collapse
Affiliation(s)
- Hadir Marei
- Cell Signaling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Angeliki Malliri
- Cell Signaling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| |
Collapse
|
18
|
Jiang W, Sheng C, Gu X, Liu D, Yao C, Gao S, Chen S, Huang Y, Huang W, Fang M. Suppression of Rac1 Signaling by Influenza A Virus NS1 Facilitates Viral Replication. Sci Rep 2016; 6:35041. [PMID: 27869202 PMCID: PMC5116764 DOI: 10.1038/srep35041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 09/13/2016] [Indexed: 11/26/2022] Open
Abstract
Influenza A virus (IAV) is a major human pathogen with the potential to become pandemic. IAV contains only eight RNA segments; thus, the virus must fully exploit the host cellular machinery to facilitate its own replication. In an effort to comprehensively characterize the host machinery taken over by IAV in mammalian cells, we generated stable A549 cell lines with over-expression of the viral non-structural protein (NS1) to investigate the potential host factors that might be modulated by the NS1 protein. We found that the viral NS1 protein directly interacted with cellular Rac1 and facilitated viral replication. Further research revealed that NS1 down-regulated Rac1 activity via post-translational modifications. Therefore, our results demonstrated that IAV blocked Rac1-mediated host cell signal transduction through the NS1 protein to facilitate its own replication. Our findings provide a novel insight into the mechanism of IAV replication and indicate new avenues for the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Wei Jiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chunjie Sheng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiuling Gu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dong Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chen Yao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shijuan Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuai Chen
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yinghui Huang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Wenlin Huang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
- Key Laboratory of Tumor Targeted Drug in Guangdong Province, Guangzhou Double Bioproducts Co., Ltd., Guangzhou, China
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- International College, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
19
|
Cypryk W, Lorey M, Puustinen A, Nyman TA, Matikainen S. Proteomic and Bioinformatic Characterization of Extracellular Vesicles Released from Human Macrophages upon Influenza A Virus Infection. J Proteome Res 2016; 16:217-227. [PMID: 27723984 DOI: 10.1021/acs.jproteome.6b00596] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Influenza A viruses (IAVs) are aggressive pathogens that cause acute respiratory diseases and annual epidemics in humans. Host defense against IAV infection is initiated by macrophages, which are the principal effector cells of the innate immune system. We have previously shown that IAV infection of human macrophages is associated with robust secretion of proteins via conventional and unconventional protein release pathways. Here we have characterized unconventional, extracellular vesicle (EV)-mediated protein secretion in human macrophages during IAV infection using proteomics, bioinformatics, and functional studies. We demonstrate that at 9 h postinfection a robust EV-mediated protein secretion takes place. We identified 2359 human proteins from EVs of IAV-infected macrophages compared with 1448 proteins identified from EVs of control cells. Bioinformatic analysis shows that many proteins involved in translation, like components of spliceosome machinery and the ribosome, are secreted in EVs in response to IAV infection. Our data also shows that EVs derived from IAV-infected macrophages contain fatty acid-binding proteins, antiviral cytokines, copper metabolism Murr-1 domain proteins, and autophagy-related proteins. In addition, our data suggest that secretory autophagy plays a role in activating EV-mediated protein secretion during IAV infection.
Collapse
Affiliation(s)
- Wojciech Cypryk
- Institute of Biotechnology, University of Helsinki , P.O. Box 56, 00014 Helsinki, Finland
| | - Martina Lorey
- University of Helsinki and Helsinki University Hospital, Rheumatology , 00029 Helsinki, Finland
| | - Anne Puustinen
- Finnish Institute of Occupational Health , Topeliuksenkatu 41 a A, 00250 Helsinki, Finland
| | - Tuula A Nyman
- Institute of Biotechnology, University of Helsinki , P.O. Box 56, 00014 Helsinki, Finland.,Institute of Clinical Medicine , Sognsvannsveien 20, Rikshospitalet, 0372 Oslo, Norway
| | - Sampsa Matikainen
- University of Helsinki and Helsinki University Hospital, Rheumatology , 00029 Helsinki, Finland
| |
Collapse
|
20
|
Stevaert A, Naesens L. The Influenza Virus Polymerase Complex: An Update on Its Structure, Functions, and Significance for Antiviral Drug Design. Med Res Rev 2016; 36:1127-1173. [PMID: 27569399 PMCID: PMC5108440 DOI: 10.1002/med.21401] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/18/2016] [Accepted: 06/24/2016] [Indexed: 12/11/2022]
Abstract
Influenza viruses cause seasonal epidemics and pandemic outbreaks associated with significant morbidity and mortality, and a huge cost. Since resistance to the existing anti‐influenza drugs is rising, innovative inhibitors with a different mode of action are urgently needed. The influenza polymerase complex is widely recognized as a key drug target, given its critical role in virus replication and high degree of conservation among influenza A (of human or zoonotic origin) and B viruses. We here review the major progress that has been made in recent years in unravelling the structure and functions of this protein complex, enabling structure‐aided drug design toward the core regions of the PA endonuclease, PB1 polymerase, or cap‐binding PB2 subunit. Alternatively, inhibitors may target a protein–protein interaction site, a cellular factor involved in viral RNA synthesis, the viral RNA itself, or the nucleoprotein component of the viral ribonucleoprotein. The latest advances made for these diverse pharmacological targets have yielded agents in advanced (i.e., favipiravir and VX‐787) or early clinical testing, besides several experimental inhibitors in various stages of development, which are all covered here.
Collapse
Affiliation(s)
| | - Lieve Naesens
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
21
|
Kwok HH, Poon PY, Fok SP, Ying-Kit Yue P, Mak NK, Chan MCW, Peiris JSM, Wong RNS. Anti-inflammatory effects of indirubin derivatives on influenza A virus-infected human pulmonary microvascular endothelial cells. Sci Rep 2016; 6:18941. [PMID: 26732368 PMCID: PMC4702174 DOI: 10.1038/srep18941] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/01/2015] [Indexed: 01/05/2023] Open
Abstract
Influenza A virus (IAV) poses global threats to human health. Acute respiratory distress syndrome and multi-organ dysfunction are major complications in patients with severe influenza infection. This may be explained by the recent studies which highlighted the role of the pulmonary endothelium as the center of innate immune cells recruitment and excessive pro-inflammatory cytokines production. In this report, we examined the potential immunomodulatory effects of two indirubin derivatives, indirubin-3′-(2,3-dihydroxypropyl)-oximether (E804) and indirubin-3′-oxime (E231), on IAV (H9N2) infected-human pulmonary microvascular endothelial cells (HPMECs). Infection of H9N2 on HPMECs induced a high level of chemokines and cytokines production including IP-10, RANTES, IL-6, IFN-β and IFN-γ1. Post-treatment of E804 or E231 could significantly suppress the production of these cytokines. H9N2 infection rapidly triggered the activation of innate immunity through phosphorylation of signaling molecules including mitogen-activated protein kinases (MAPKs) and signal transducer and activator of transcription (STAT) proteins. Using specific inhibitors or small-interfering RNA, we confirmed that indirubin derivatives can suppress H9N2-induced cytokines production through MAPKs and STAT3 signaling pathways. These results underscore the immunomodulatory effects of indirubin derivatives on pulmonary endothelium and its therapeutic potential on IAV-infection.
Collapse
Affiliation(s)
- Hoi-Hin Kwok
- Dr. Gilbert Hung Ginseng Laboratory, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region.,Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region
| | - Po-Ying Poon
- Dr. Gilbert Hung Ginseng Laboratory, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region.,Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region
| | - Siu-Ping Fok
- Dr. Gilbert Hung Ginseng Laboratory, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region.,Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region
| | - Patrick Ying-Kit Yue
- Dr. Gilbert Hung Ginseng Laboratory, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region.,Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region
| | - Nai-Ki Mak
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region
| | - Michael Chi-Wai Chan
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Joseph Sriyal Malik Peiris
- Centre of Influenza Research and School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Ricky Ngok-Shun Wong
- Dr. Gilbert Hung Ginseng Laboratory, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region.,Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region
| |
Collapse
|
22
|
Tejada-Simon MV. Modulation of actin dynamics by Rac1 to target cognitive function. J Neurochem 2015; 133:767-79. [PMID: 25818528 DOI: 10.1111/jnc.13100] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/11/2015] [Accepted: 03/14/2015] [Indexed: 12/14/2022]
Abstract
The small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in cells. Formation of extensions at the surface of the cell is required for migration and even for cell invasion and metastases. Because an elevated level and hyperactivation of this protein has been associated with metastasis in cancer, direct regulators of Rac1 are currently envisioned as a potential strategy to treat certain cancers. Less research, however, has been done regarding the role of this small GTP-binding protein in brain development, where it has an important role in dendritic spine morphogenesis through the regulation of actin. Alteration of dendritic development and spinogenesis has been often associated with mental disorders. Rac1 is associated with and required for learning and the formation of memories in the brain. Rac1 appears to be dysregulated in certain neurodevelopmental disorders that present all these three alterations: mental retardation, atypical synaptic plasticity and aberrant spine morphology. Thus, to develop novel therapies for rescuing cognitive impairment, a reasonable approach might be to target this protein, Rac1, which plays a pivotal role in directing signals that regulate actin dynamics, which in turn might have an effect in spine cytoarchitecture and synaptic function. It is possible that novel drugs that regulate Rac1 activation and function could modulate actin cytoskeleton and spine dynamics, representing potential candidates to repair intellectual disability in disorders associated with spine abnormalities. Herein, we present a list of the current Rac1 inhibitors that might fulfill this role together with a summary of the latest findings concerning their function as they relate to neuronal studies. While the small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in different type of cells, it appears to be also required for learning and the formation of memories in the brain. Abnormal regulation of this protein has been associated with cognitive disabilities, atypical synaptic plasticity and abnormal morphology of dendritic spines in certain neurodevelopmental disorders. Thus, modulation of Rac1 activity using novel inhibitors might be a strategy to reestablish cognitive function.
Collapse
Affiliation(s)
- Maria V Tejada-Simon
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA.,Department of Biology, University of Houston, Houston, Texas, USA.,Department of Psychology, University of Houston, Houston, Texas, USA.,Biology of Behavior Institute (BoBI), University of Houston, Houston, Texas, USA
| |
Collapse
|
23
|
GASPARINI R, AMICIZIA D, LAI P, BRAGAZZI N, PANATTO D. Compounds with anti-influenza activity: present and future of strategies for the optimal treatment and management of influenza. Part II: Future compounds against influenza virus. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2014; 55:109-29. [PMID: 26137785 PMCID: PMC4718316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
In the first part of this overview, we described the life cycle of the influenza virus and the pharmacological action of the currently available drugs. This second part provides an overview of the molecular mechanisms and targets of still-experimental drugs for the treatment and management of influenza. Briefly, we can distinguish between compounds with anti-influenza activity that target influenza virus proteins or genes, and molecules that target host components that are essential for viral replication and propagation. These latter compounds have been developed quite recently. Among the first group, we will focus especially on hemagglutinin, M2 channel and neuraminidase inhibitors. The second group of compounds may pave the way for personalized treatment and influenza management. Combination therapies are also discussed. In recent decades, few antiviral molecules against influenza virus infections have been available; this has conditioned their use during human and animal outbreaks. Indeed, during seasonal and pandemic outbreaks, antiviral drugs have usually been administered in mono-therapy and, sometimes, in an uncontrolled manner to farm animals. This has led to the emergence of viral strains displaying resistance, especially to compounds of the amantadane family. For this reason, it is particularly important to develop new antiviral drugs against influenza viruses. Indeed, although vaccination is the most powerful means of mitigating the effects of influenza epidemics, antiviral drugs can be very useful, particularly in delaying the spread of new pandemic viruses, thereby enabling manufacturers to prepare large quantities of pandemic vaccine. In addition, antiviral drugs are particularly valuable in complicated cases of influenza, especially in hospitalized patients. To write this overview, we mined various databases, including Embase, PubChem, DrugBank and Chemical Abstracts Service, and patent repositories.
Collapse
Affiliation(s)
- R. GASPARINI
- Correspondence: R. Gasparini, Department of Health Sciences of Genoa University, via Pastore 1, 16132 Genoa, Italy - E-mail:
| | | | | | | | | |
Collapse
|
24
|
de Chassey B, Meyniel-Schicklin L, Vonderscher J, André P, Lotteau V. Virus-host interactomics: new insights and opportunities for antiviral drug discovery. Genome Med 2014; 6:115. [PMID: 25593595 PMCID: PMC4295275 DOI: 10.1186/s13073-014-0115-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The current therapeutic arsenal against viral infections remains limited, with often poor efficacy and incomplete coverage, and appears inadequate to face the emergence of drug resistance. Our understanding of viral biology and pathophysiology and our ability to develop a more effective antiviral arsenal would greatly benefit from a more comprehensive picture of the events that lead to viral replication and associated symptoms. Towards this goal, the construction of virus-host interactomes is instrumental, mainly relying on the assumption that a viral infection at the cellular level can be viewed as a number of perturbations introduced into the host protein network when viral proteins make new connections and disrupt existing ones. Here, we review advances in interactomic approaches for viral infections, focusing on high-throughput screening (HTS) technologies and on the generation of high-quality datasets. We show how these are already beginning to offer intriguing perspectives in terms of virus-host cell biology and the control of cellular functions, and we conclude by offering a summary of the current situation regarding the potential development of host-oriented antiviral therapeutics.
Collapse
Affiliation(s)
| | | | | | - Patrice André
- />Hospices Civils de Lyon, Lyon, France
- />CIRI, Université de Lyon, Lyon, 69365 France
- />Inserm, U1111, Lyon, 69365 France
| | - Vincent Lotteau
- />CIRI, Université de Lyon, Lyon, 69365 France
- />Inserm, U1111, Lyon, 69365 France
| |
Collapse
|
25
|
Spear M, Wu Y. Viral exploitation of actin: force-generation and scaffolding functions in viral infection. Virol Sin 2014; 29:139-47. [PMID: 24938714 DOI: 10.1007/s12250-014-3476-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 05/30/2014] [Indexed: 11/25/2022] Open
Abstract
As a fundamental component of the host cellular cytoskeleton, actin is routinely engaged by infecting viruses. Furthermore, viruses from diverse groups, and infecting diverse hosts, have convergently evolved an array of mechanisms for manipulating the actin cytoskeleton for efficacious infection. An ongoing chorus of research now indicates that the actin cytoskeleton is critical for viral replication at many stages of the viral life cycle, including binding, entry, nuclear localization, genomic transcription and reverse transcription, assembly, and egress/dissemination. Specifically, viruses subvert the force-generating and macromolecular scaffolding properties of the actin cytoskeleton to propel viral surfing, internalization, and migration within the cell. Additionally, viruses utilize the actin cytoskeleton to support and organize assembly sites, and eject budding virions for cell-to-cell transmission. It is the purpose of this review to provide an overview of current research, focusing on the various mechanisms and themes of virus-mediated actin modulation described therein.
Collapse
Affiliation(s)
- Mark Spear
- National Center for Biodefense and Infectious Diseases, Department of Molecular and Microbiology, George Mason University, Manassas, VA, 20110, USA
| | | |
Collapse
|
26
|
Hillesheim A, Nordhoff C, Boergeling Y, Ludwig S, Wixler V. β-catenin promotes the type I IFN synthesis and the IFN-dependent signaling response but is suppressed by influenza A virus-induced RIG-I/NF-κB signaling. Cell Commun Signal 2014; 12:29. [PMID: 24767605 PMCID: PMC4021428 DOI: 10.1186/1478-811x-12-29] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/15/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The replication cycle of most pathogens, including influenza viruses, is perfectly adapted to the metabolism and signal transduction pathways of host cells. After infection, influenza viruses activate several cellular signaling cascades that support their propagation but suppress those that interfere with viral replication. Accumulation of viral RNA plays thereby a central role. Its sensing by the pattern recognition receptors of the host cells leads to the activation of several signal transduction waves that result in induction of genes, responsible for the cellular innate immune response. Type I interferon (IFN) genes and interferon-stimulated genes (ISG) coding for antiviral-acting proteins, such as MxA, OAS-1 or PKR, are primary targets of these signaling cascades. β- and γ-catenin are closely related armadillo repeat-containing proteins with dual roles. At the cell membrane they serve as adapter molecules linking cell-cell contacts to microfilaments. In the cytosol and nucleus, the proteins form a transcriptional complex with the lymphoid enhancer factor/T-cell factor (LEF/TCF), regulating the transcription of many genes, thereby controlling different cellular functions such as cell cycle progression and differentiation. RESULTS In this study, we demonstrate that β- and γ-catenin are important regulators of the innate cellular immune response to influenza A virus (IAV) infections. They inhibit viral replication in lung epithelial cells by enhancing the virus-dependent induction of the IFNB1 gene and interferon-stimulated genes. Simultaneously, the prolonged infection counteracts the antiviral effect of β- and γ-catenin. Influenza viruses suppress β-catenin-dependent transcription by misusing the RIG-I/NF-κB signaling cascade that is induced in the course of infection by viral RNA. CONCLUSION We identified β- and γ-catenin as novel antiviral-acting proteins. While these factors support the induction of common target genes of the cellular innate immune response, their functional activity is suppressed by pathogen evasion.
Collapse
Affiliation(s)
- Andrea Hillesheim
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, 48149 Muenster, Germany
| | - Carolin Nordhoff
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, 48149 Muenster, Germany
| | - Yvonne Boergeling
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, 48149 Muenster, Germany
| | - Stephan Ludwig
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, 48149 Muenster, Germany
| | - Viktor Wixler
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, 48149 Muenster, Germany
| |
Collapse
|