1
|
Yuen NKY, Eng M, Hudson NJ, Sole-Guitart A, Coyle MP, Bielefeldt-Ohmann H. Distinct cellular and molecular responses to infection in three target cell types from horses, a species naturally susceptible to Ross River virus. Microb Pathog 2025; 202:107408. [PMID: 40010657 DOI: 10.1016/j.micpath.2025.107408] [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: 12/02/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Our current understanding of the pathogenesis of Ross River virus (RRV) infection has been derived from murine models, which do not reproduce clinical disease as experienced by infected humans and horses. This prompted us to establish more relevant host model systems to study host-virus interactions using ex vivo peripheral blood mononuclear cells (PBMCs) and in vitro primary synovial fibroblast and epidermal keratinocyte cultures. Transcriptomic analysis revealed that the expression of the transmembrane protein matrix remodelling associated 8 (mxra8), recently found to be responsible for RRV cell entry, was downregulated in all cell types when infected with RRV, compared to mock-infected controls. Potent antiviral and inflammatory responses were generated by both synovial fibroblasts and epidermal keratinocytes upon RRV infection. Upregulation of multiple genes, inducible by double-stranded RNA, together with upregulation of toll-like receptor (TLR) tlr-3, but not tlr-7, 8 and 9, suggests possible abortive replication of RRV in these cell types and potent antiviral mechanisms. This was corroborated by virus growth kinetic studies which indicated inefficient RRV replication in synovial fibroblasts and epidermal keratinocytes. Cellular metabolic flux studies on PBMCs and synovial fibroblasts showed that RRV infected cells had reduced mitochondrial function. In addition, compared to PBMCs of seronegative horses, an enhanced antiviral state and reduced inflammation related gene expression was seen in PBMCs of seropositive horses infected with RRV. Thus, despite potent antiviral and inflammatory responses via the interferon pathway exhibited in all cell types, restricting virus growth, mitochondria capacity and function of infected cells remained negatively impacted.
Collapse
Affiliation(s)
- Nicholas K Y Yuen
- School of Veterinary Science, Faculty of Science, University of Queensland, Gatton, Queensland, Australia.
| | - Melodie Eng
- School of Veterinary Science, Faculty of Science, University of Queensland, Gatton, Queensland, Australia
| | - Nicholas J Hudson
- School of Agriculture and Food Sustainability, Faculty of Science, University of Queensland, Gatton, Queensland, Australia
| | - Albert Sole-Guitart
- School of Veterinary Science, Faculty of Science, University of Queensland, Gatton, Queensland, Australia
| | - Mitchell P Coyle
- Equine Unit, Office of the Director Gatton Campus, Faculty of Science, University of Queensland, Gatton, Queensland, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, Faculty of Science, University of Queensland, St Lucia, Queensland, Australia; Australian Infectious Diseases Research Centre, University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
2
|
Parashar B, Malviya R, Sridhar SB, Wadhwa T, Talath S, Shareef J. Eastern equine encephalitis virus: Pathogenesis, immune response, and clinical manifestations. INFECTIOUS MEDICINE 2025; 4:100167. [PMID: 40026316 PMCID: PMC11869868 DOI: 10.1016/j.imj.2025.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025]
Abstract
Eastern equine encephalitis virus (EEEV) is a lethal Alphavirus transmitted by Culiseta melanura mosquitoes that primarily cycles between birds. Although rare, infections in humans and horses are associated with high mortality rates and severe neurological effects. Climate change appears to be increasing the spread of this virus. This study aims to provide a comprehensive analysis of EEEV, including its transmission dynamics, pathogenesis, induced host immune response, and long-term impacts on survivors. It also highlights the virus's unique immune evasion strategies that complicate disease management and contribute to severe clinical outcomes, such as encephalitis with fever, convulsions, and coma. Survivors often face chronic cognitive, motor, and psychosocial impairments. Despite these significant public health risks, gaps remain in understanding the molecular mechanisms underlying immune evasion and the long-term neurological sequelae in survivors. By collating current knowledge, this review underscores the urgent need for the development of targeted vaccines and therapeutic interventions to mitigate the growing threat of EEEV, particularly in the context of climate change-driven geographical expansion.
Collapse
Affiliation(s)
- Bhumika Parashar
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
- Galgotias Multi-Disciplinary Research & Development Cell (G-MRDC), Galgotias University, Greater Noida 201308, Uttar Pradesh, India
| | - Sathvik Belagodu Sridhar
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Tarun Wadhwa
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sirajunisa Talath
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Javedh Shareef
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| |
Collapse
|
3
|
Martin CK, Yin P, Kielian M. The sticky business of Alphavirus capsid-host interactions. Trends Microbiol 2025; 33:321-339. [PMID: 39665907 PMCID: PMC11916923 DOI: 10.1016/j.tim.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024]
Abstract
Alphaviruses are a serious threat to global health and can cause lethal encephalitic or arthritogenic disease in humans and animals. As there are no licensed antivirals, it is critical to improve our understanding of alphavirus interactions with the host cell. Here, we focus on the essential alphavirus protein capsid. While its roles in genome packaging and virus assembly have been well-studied, much less is known about capsid's interactions with host proteins and their functional relevance for infection. Recently, several new capsid interactor candidates were identified, collectively emphasising the complexity of capsid-host biology. In this review we summarise these novel interactor candidates, highlight capsid's emerging role in immune evasion, and discuss the challenges and opportunities arising from capsid-host interactions.
Collapse
Affiliation(s)
- Caroline K Martin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Peiqi Yin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
4
|
VanderGiessen M, Jamiu A, Heath B, Akhrymuk I, Kehn-Hall K. Cellular takeover: How new world alphaviruses impact host organelle function. Virology 2025; 603:110365. [PMID: 39733515 DOI: 10.1016/j.virol.2024.110365] [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: 10/15/2024] [Revised: 11/28/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024]
Abstract
Alphavirus replication is dependent on host cell organelles to facilitate multiple steps of the viral life cycle. New world alphaviruses (NWA) consisting of eastern, western and Venezuelan equine encephalitis viruses are a subgroup of alphaviruses associated with central nervous system disease. Despite differing morbidity and mortality amongst these viruses, all are important human pathogens due to their transmission through viral aerosolization and mosquito transmission. In this review, we summarize the utilization of host organelles for NWA replication and the subversion of the host innate immune responses. The impact of viral proteins and replication processes on organelle function is also discussed. Literature involving old world alphaviruses (OWA), such as chikungunya virus and Sindbis virus, is included to compare and contrast between OWA and NWA and highlight gaps in knowledge for NWA. Finally, potential targets for therapeutics or vaccine candidates are highlighted with a focus on host-directed therapeutics.
Collapse
Affiliation(s)
- Morgen VanderGiessen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Abdullahi Jamiu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Brittany Heath
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Ivan Akhrymuk
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Kylene Kehn-Hall
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.
| |
Collapse
|
5
|
Garzan A, Ahmed SK, Haese NN, Sulgey G, Medica S, Smith JL, Zhang S, Ahmad F, Karyakarte S, Rasmussen L, DeFilippis V, Tekwani B, Bostwick R, Suto MJ, Hirsch AJ, Morrison TE, Heise MT, Augelli-Szafran CE, Streblow DN, Pathak AK, Moukha-Chafiq O. 4-Substituted-2-Thiazole Amides as Viral Replication Inhibitors of Alphaviruses. J Med Chem 2024; 67:20858-20878. [PMID: 39621435 DOI: 10.1021/acs.jmedchem.4c01073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
2-(Methylthio)-N-(4-(naphthalen-2-yl)thiazol-2-yl)nicotinamide 1 was identified as an inhibitor against Chikungunya virus (CHIKV) with good antiviral activity [EC50 = 0.6 μM; EC90 = 0.93 μM and viral titer reduction (VTR) of 6.9 logs at 10 μM concentration] with no observed cytotoxicity (CC50 = 132 μM) in normal human dermal fibroblast (NHDF) cells. Structure-activity relationship (SAR) studies to further improve the potency, efficacy, and drug-like properties of 1 led to the discovery of a new potent inhibitor N-(4-(3-((4-cyanophenyl)amino)phenyl)thiazol-2-yl)-2-(methylthio)nicotinamide 26, which showed a VTR of 8.7 logs at 10 μM against CHIKV and an EC90 of 0.45 μM with considerably improved MLM stability (t1/2 = 74 min) as compared to 1. Mechanism of action studies show that 26 inhibits alphavirus replication by blocking subgenomic viral RNA translation and structural protein synthesis. The in vivo efficacy studies of compound 26 on CHIKV infection in mice are reported.
Collapse
Affiliation(s)
- Atefeh Garzan
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - S Kaleem Ahmed
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Nicole N Haese
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Gauthami Sulgey
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Samuel Medica
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Jessica L Smith
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Sixue Zhang
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Fahim Ahmad
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Shuklendu Karyakarte
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Lynn Rasmussen
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Victor DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Babu Tekwani
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Robert Bostwick
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Mark J Suto
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19th Avenue, Aurora, Colorado 80045, United States
| | - Mark T Heise
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Corinne E Augelli-Szafran
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Ashish K Pathak
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Omar Moukha-Chafiq
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| |
Collapse
|
6
|
Freppel W, Silva LA, Stapleford KA, Herrero LJ. Pathogenicity and virulence of chikungunya virus. Virulence 2024; 15:2396484. [PMID: 39193780 PMCID: PMC11370967 DOI: 10.1080/21505594.2024.2396484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted, RNA virus that causes an often-severe musculoskeletal illness characterized by fever, joint pain, and a range of debilitating symptoms. The virus has re-emerged as a global health threat in recent decades, spreading from its origin in Africa across Asia and the Americas, leading to widespread outbreaks impacting millions of people. Despite more than 50 years of research into the pathogenesis of CHIKV, there is still no curative treatment available. Current management of CHIKV infections primarily involves providing supportive care to alleviate symptoms and improve the patient's quality of life. Given the ongoing threat of CHIKV, there is an urgent need to better understand its pathogenesis. This understanding is crucial for deciphering the mechanisms underlying the disease and for developing effective strategies for both prevention and management. This review aims to provide a comprehensive overview of CHIKV and its pathogenesis, shedding light on the complex interactions of viral genetics, host factors, immune responses, and vector-related factors. By exploring these intricate connections, the review seeks to contribute to the knowledge base surrounding CHIKV, offering insights that may ultimately lead to more effective prevention and management strategies for this re-emerging global health threat.
Collapse
Affiliation(s)
- Wesley Freppel
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| | - Laurie A. Silva
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lara J. Herrero
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| |
Collapse
|
7
|
de Oliveira Souza R, Duarte Júnior JWB, Della Casa VS, Santoro Rosa D, Renia L, Claser C. Unraveling the complex interplay: immunopathology and immune evasion strategies of alphaviruses with emphasis on neurological implications. Front Cell Infect Microbiol 2024; 14:1421571. [PMID: 39211797 PMCID: PMC11358129 DOI: 10.3389/fcimb.2024.1421571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/09/2024] [Indexed: 09/04/2024] Open
Abstract
Arthritogenic alphaviruses pose a significant public health concern due to their ability to cause joint inflammation, with emerging evidence of potential neurological consequences. In this review, we examine the immunopathology and immune evasion strategies employed by these viruses, highlighting their complex mechanisms of pathogenesis and neurological implications. We delve into how these viruses manipulate host immune responses, modulate inflammatory pathways, and potentially establish persistent infections. Further, we explore their ability to breach the blood-brain barrier, triggering neurological complications, and how co-infections exacerbate neurological outcomes. This review synthesizes current research to provide a comprehensive overview of the immunopathological mechanisms driving arthritogenic alphavirus infections and their impact on neurological health. By highlighting knowledge gaps, it underscores the need for research to unravel the complexities of virus-host interactions. This deeper understanding is crucial for developing targeted therapies to address both joint and neurological manifestations of these infections.
Collapse
Affiliation(s)
- Raquel de Oliveira Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | | | - Victória Simões Della Casa
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Laurent Renia
- ASTAR Infectious Diseases Labs (ASTAR ID Labs), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Carla Claser
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
8
|
Jian Z, Jiang C, Zhu L, Li F, Deng L, Ai Y, Lai S, Xu Z. Infectivity and pathogenesis characterization of getah virus (GETV) strain via different inoculation routes in mice. Heliyon 2024; 10:e33432. [PMID: 39040396 PMCID: PMC11260979 DOI: 10.1016/j.heliyon.2024.e33432] [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: 10/30/2023] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
In recent years, the epidemiological profile of Getah virus (GETV) has become increasingly serious, posing a huge threat to animal and public health in China. GETV can cause multi-species infection, including horses, pigs, rats, cattle, kangaroos, reptiles and birds. However, there were few reports on the efficiency of the virus entering the host via routes of different systems. In the present study, a GETV strain (SC201807) was obtained from a piglet's blood in 2018 in Sichuan, China. First, we established a quantitative real-time polymerase chain reaction (qRT-PCR) SYBR assay specific to GETV. Then, we evaluated the infection efficiency of different routes using mouse animal model. 108 male mice were randomly divided into four groups as follows: intramuscular, intraoral and intranasal infection routes, and negative control. All mice in the experimental group were inoculated with 4 × 102.85 TCID50 GETV virus. Tissue tropism experiments show that GETV has a wide range of tissue distribution, and intramuscular infection is the first to infect all tissues of the body, and suggest that oral infection may be a new GETV transmission route. Histopathological examination results showed that intramuscular injection of GETV mainly caused different degrees of pathological damage to the tissues, and could rapidly induce a large amount of inflammatory regulatory factors such as IL-6 and TNF-α. Our data may help us to evaluate the risk of transmission of Porcine Getah virus and provide an experimental basis for the prevention and control of Porcine Getah virus.
Collapse
Affiliation(s)
- Zhijie Jian
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Chaoyuan Jiang
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- Chengdu Zhongji Agriculture and Animal Husbandry Co., Ltd, No. 37, Middle Section, Heshan Street, Pujiang County, Chengdu, Sichuan Province, China
| | - Ling Zhu
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Fengqin Li
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- College of Animal Science, Xichang University, Xichang, 615000, Sichuan, China
| | - Lishuang Deng
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Yanru Ai
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Siyuan Lai
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| | - Zhiwen Xu
- Veterinary Medicine College, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu, Sichuan Province, China
| |
Collapse
|
9
|
Bezerra WP, Moizéis RNC, Salmeron ACA, Pereira HWB, de Araújo JMG, Guedes PMM, Fernandes JV, Nascimento MSL. Innate immune response in patients with acute Chikungunya disease. Med Microbiol Immunol 2023:10.1007/s00430-023-00771-y. [PMID: 37285099 DOI: 10.1007/s00430-023-00771-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/29/2023] [Indexed: 06/08/2023]
Abstract
Chikungunya disease (CHIKD) is an arbovirose that presents with high morbidity, mainly due to arthralgia. Inflammatory mediators including IL-6, IL-1β, GM-CSF and others have been implicated in the pathogenesis of CHIKD, whilst type I interferons can be associated with better outcomes. The role of pattern recognition receptors has been studied incompletely. Here, we evaluated the expression of RNA-specific PRRs, their adaptor molecules and downstream cytokines in acute CHIKD patients. Twenty-eight patients were recruited during the 3rd-5th day after the symptoms onset for clinical examination, peripheral blood collection and qRT-PCR analysis of PBMC to compare to the healthy control group (n = 20). We observed common symptoms of acute CHIKD, with fever, arthralgia, headache and myalgia being the most frequent. Compared with uninfected controls, acute CHIKV infection upregulates the expression of the receptors TLR3, RIG-I and MDA5, and also the adaptor molecule TRIF. Regarding cytokine expression, we found an upregulation of IL-6, IL-12, IFN-α, IFN-β and IFN-γ, which are related directly to the inflammatory or antiviral response. The TLR3-TRIF axis correlated with high expression of IL-6 and IFN-α. Interestingly, greater expression of MDA5, IL-12 and IFN-α was related to lower viral loads in CHIKD acute patients. Together, these findings help to complete the picture of innate immune activation during acute CHIKD, while confirming the induction of strong antiviral responses. Drawing the next steps in the understanding of the immunopathology and virus clearance mechanisms of CHIKD should be of utter importance in the aid of the development of effective treatment to reduce the severity of this debilitating disease.
Collapse
Affiliation(s)
- Wallace Pitanga Bezerra
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte. Natal, Rio Grande do Norte, Natal, Rio Grande Do Norte, 59078-970, Brazil
| | - Raíza Nara Cunha Moizéis
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte. Natal, Rio Grande do Norte, Natal, Rio Grande Do Norte, 59078-970, Brazil
| | - Amanda Costa Ayres Salmeron
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaiba, Rio Grande do Norte, Brazil
| | - Hannaly Wana Bezerra Pereira
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte. Natal, Rio Grande do Norte, Natal, Rio Grande Do Norte, 59078-970, Brazil
| | - Josélio Maria Galvão de Araújo
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte. Natal, Rio Grande do Norte, Natal, Rio Grande Do Norte, 59078-970, Brazil
| | - Paulo Marcos Matta Guedes
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte. Natal, Rio Grande do Norte, Natal, Rio Grande Do Norte, 59078-970, Brazil
| | - José Veríssimo Fernandes
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte. Natal, Rio Grande do Norte, Natal, Rio Grande Do Norte, 59078-970, Brazil
| | - Manuela Sales Lima Nascimento
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte. Natal, Rio Grande do Norte, Natal, Rio Grande Do Norte, 59078-970, Brazil.
- Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaiba, Rio Grande do Norte, Brazil.
| |
Collapse
|
10
|
Treffers EE, Tas A, Scholte FEM, de Ru AH, Snijder EJ, van Veelen PA, van Hemert MJ. The alphavirus nonstructural protein 2 NTPase induces a host translational shut-off through phosphorylation of eEF2 via cAMP-PKA-eEF2K signaling. PLoS Pathog 2023; 19:e1011179. [PMID: 36848386 PMCID: PMC9997916 DOI: 10.1371/journal.ppat.1011179] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/09/2023] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging alphavirus. Since 2005, it has infected millions of people during outbreaks in Africa, Asia, and South/Central America. CHIKV replication depends on host cell factors at many levels and is expected to have a profound effect on cellular physiology. To obtain more insight into host responses to infection, stable isotope labeling with amino acids in cell culture and liquid chromatography-tandem mass spectrometry were used to assess temporal changes in the cellular phosphoproteome during CHIKV infection. Among the ~3,000 unique phosphorylation sites analyzed, the largest change in phosphorylation status was measured on residue T56 of eukaryotic elongation factor 2 (eEF2), which showed a >50-fold increase at 8 and 12 h p.i. Infection with other alphaviruses (Semliki Forest, Sindbis and Venezuelan equine encephalitis virus (VEEV)) triggered a similarly strong eEF2 phosphorylation. Expression of a truncated form of CHIKV or VEEV nsP2, containing only the N-terminal and NTPase/helicase domains (nsP2-NTD-Hel), sufficed to induce eEF2 phosphorylation, which could be prevented by mutating key residues in the Walker A and B motifs of the NTPase domain. Alphavirus infection or expression of nsP2-NTD-Hel resulted in decreased cellular ATP levels and increased cAMP levels. This did not occur when catalytically inactive NTPase mutants were expressed. The wild-type nsP2-NTD-Hel inhibited cellular translation independent of the C-terminal nsP2 domain, which was previously implicated in directing the virus-induced host shut-off for Old World alphaviruses. We hypothesize that the alphavirus NTPase activates a cellular adenylyl cyclase resulting in increased cAMP levels, thus activating PKA and subsequently eukaryotic elongation factor 2 kinase. This in turn triggers eEF2 phosphorylation and translational inhibition. We conclude that the nsP2-driven increase of cAMP levels contributes to the alphavirus-induced shut-off of cellular protein synthesis that is shared between Old and New World alphaviruses. MS Data are available via ProteomeXchange with identifier PXD009381.
Collapse
Affiliation(s)
- Emmely E. Treffers
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ali Tas
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Florine E. M. Scholte
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric J. Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A. van Veelen
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn J. van Hemert
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
11
|
Vasconcellos AF, Melo RM, Mandacaru SC, de Oliveira LS, de Oliveira AS, Moraes ECDS, Trugilho MRDO, Ricart CAO, Báo SN, Resende RO, Charneau S. Aedes aegypti Aag-2 Cell Proteome Modulation in Response to Chikungunya Virus Infection. Front Cell Infect Microbiol 2022; 12:920425. [PMID: 35782121 PMCID: PMC9240781 DOI: 10.3389/fcimb.2022.920425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/18/2022] [Indexed: 01/16/2023] Open
Abstract
Chikungunya virus (CHIKV) is a single-stranded positive RNA virus that belongs to the genus Alphavirus and is transmitted to humans by infected Aedes aegypti and Aedes albopictus bites. In humans, CHIKV usually causes painful symptoms during acute and chronic stages of infection. Conversely, virus–vector interaction does not disturb the mosquito’s fitness, allowing a persistent infection. Herein, we studied CHIKV infection of Ae. aegypti Aag-2 cells (multiplicity of infection (MOI) of 0.1) for 48 h through label-free quantitative proteomic analysis and transmission electron microscopy (TEM). TEM images showed a high load of intracellular viral cargo at 48 h postinfection (hpi), as well as an unusual elongated mitochondria morphology that might indicate a mitochondrial imbalance. Proteome analysis revealed 196 regulated protein groups upon infection, which are related to protein synthesis, energy metabolism, signaling pathways, and apoptosis. These Aag-2 proteins regulated during CHIKV infection might have roles in antiviral and/or proviral mechanisms and the balance between viral propagation and the survival of host cells, possibly leading to the persistent infection.
Collapse
Affiliation(s)
- Anna Fernanda Vasconcellos
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- Laboratory of Virology, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Reynaldo Magalhães Melo
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Samuel Coelho Mandacaru
- Laboratory of Toxinology and Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Lucas Silva de Oliveira
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Athos Silva de Oliveira
- Laboratory of Virology, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | | | | | - Carlos André Ornelas Ricart
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Sônia Nair Báo
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Renato Oliveira Resende
- Laboratory of Virology, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- *Correspondence: Sébastien Charneau, ; Renato Oliveira Resende,
| | - Sébastien Charneau
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- *Correspondence: Sébastien Charneau, ; Renato Oliveira Resende,
| |
Collapse
|
12
|
Rueda JC, Arcos-Burgos M, Santos AM, Martin-Arsanios D, Villota-Erazo C, Reyes V, Bernal-Macías S, Peláez-Ballestas I, Cardiel MH, Londono J. Human Genetic Host Factors and Its Role in the Pathogenesis of Chikungunya Virus Infection. Front Med (Lausanne) 2022; 9:654395. [PMID: 35252226 PMCID: PMC8888679 DOI: 10.3389/fmed.2022.654395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Chikungunya virus (CHIKV) is an alphavirus from the Togaviridae family that causes acute arthropathy in humans. It is an arthropod-borne virus transmitted initially by the Aedes (Ae) aegypti and after 2006's epidemic in La Reunion by Ae albopictus due to an adaptive mutation of alanine for valine in the position 226 of the E1 glycoprotein genome (A226V). The first isolated cases of CHIKV were reported in Tanzania, however since its arrival to the Western Hemisphere in 2013, the infection became a pandemic. After a mosquito bite from an infected viremic patient the virus replicates eliciting viremia, fever, rash, myalgia, arthralgia, and arthritis. After the acute phase, CHIKV infection can progress to a chronic stage where rheumatic symptoms can last for several months to years. Although there is a great number of studies on the pathogenesis of CHIKV infection not only in humans but also in animal models, there still gaps in the proper understanding of the disease. To this date, it is unknown why a percentage of patients do not develop clinical symptoms despite having been exposed to the virus and developing an adaptive immune response. Also, controversy stills exist on the pathogenesis of chronic joint symptoms. It is known that host immune response to an infectious disease is reflected on patient's symptoms. At the same time, it is now well-established that host genetic variation is an important component of the varied onset, severity, and outcome of infectious disease. It is essential to understand the interaction between the aetiological agent and the host to know the chronic sequelae of the disease. The present review summarizes the current findings on human host genetics and its relationship with immune response in CHIKV infection.
Collapse
Affiliation(s)
- Juan C. Rueda
- Faculty of Medicine and Engineering, Universidad de La Sabana, Chía, Colombia
- Grupo de Espondiloartropatías, Rheumatology Department, Universidad de La Sabana, Chía, Colombia
| | - Mauricio Arcos-Burgos
- Grupo de Investigación en Psiquiatría (GIPSI), Departamento de Psiquiatría, Faculty of Medicine, Instituto de Investigaciones Médicas, Universidad de Antioquia, Medellín, Colombia
| | - Ana M. Santos
- Grupo de Espondiloartropatías, Rheumatology Department, Universidad de La Sabana, Chía, Colombia
| | - Daniel Martin-Arsanios
- Grupo de Espondiloartropatías, Rheumatology Department, Universidad de La Sabana, Chía, Colombia
| | - Catalina Villota-Erazo
- Grupo de Espondiloartropatías, Rheumatology Department, Universidad de La Sabana, Chía, Colombia
- Rheumatology Department, Hospital Militar Central, Bogotá, Colombia
| | - Viviana Reyes
- Grupo de Espondiloartropatías, Rheumatology Department, Universidad de La Sabana, Chía, Colombia
- Rheumatology Department, Hospital Militar Central, Bogotá, Colombia
| | - Santiago Bernal-Macías
- Grupo de Espondiloartropatías, Rheumatology Department, Universidad de La Sabana, Chía, Colombia
- Rheumatology Department, Hospital Militar Central, Bogotá, Colombia
| | | | | | - John Londono
- Grupo de Espondiloartropatías, Rheumatology Department, Universidad de La Sabana, Chía, Colombia
- Rheumatology Department, Hospital Militar Central, Bogotá, Colombia
- *Correspondence: John Londono
| |
Collapse
|
13
|
Kafai NM, Diamond MS, Fox JM. Distinct Cellular Tropism and Immune Responses to Alphavirus Infection. Annu Rev Immunol 2022; 40:615-649. [PMID: 35134315 DOI: 10.1146/annurev-immunol-101220-014952] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Alphaviruses are emerging and reemerging viruses that cause disease syndromes ranging from incapacitating arthritis to potentially fatal encephalitis. While infection by arthritogenic and encephalitic alphaviruses results in distinct clinical manifestations, both virus groups induce robust innate and adaptive immune responses. However, differences in cellular tropism, type I interferon induction, immune cell recruitment, and B and T cell responses result in differential disease progression and outcome. In this review, we discuss aspects of immune responses that contribute to protective or pathogenic outcomes after alphavirus infection. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Natasha M Kafai
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; , .,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael S Diamond
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; , .,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Julie M Fox
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
14
|
Sharma A, Kontodimas K, Bosmann M. The MAVS Immune Recognition Pathway in Viral Infection and Sepsis. Antioxid Redox Signal 2021; 35:1376-1392. [PMID: 34348482 PMCID: PMC8817698 DOI: 10.1089/ars.2021.0167] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 02/03/2023]
Abstract
Significance: It is estimated that close to 50 million cases of sepsis result in over 11 million annual fatalities worldwide. The pathognomonic feature of sepsis is a dysregulated inflammatory response arising from viral, bacterial, or fungal infections. Immune recognition of pathogen-associated molecular patterns is a hallmark of the host immune defense to combat microbes and to prevent the progression to sepsis. Mitochondrial antiviral signaling protein (MAVS) is a ubiquitous adaptor protein located at the outer mitochondrial membrane, which is activated by the cytosolic pattern recognition receptors, retinoic acid-inducible gene I (RIG-I) and melanoma differentiation associated gene 5 (MDA5), following binding of viral RNA agonists. Recent Advances: Substantial progress has been made in deciphering the activation of the MAVS pathway with its interacting proteins, downstream signaling events (interferon [IFN] regulatory factors, nuclear factor kappa B), and context-dependent type I/III IFN response. Critical Issues: In the evolutionary race between pathogens and the host, viruses have developed immune evasion strategies for cleavage, degradation, or blockade of proteins in the MAVS pathway. For example, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) M protein and ORF9b protein antagonize MAVS signaling and a protective type I IFN response. Future Directions: The role of MAVS as a sensor for nonviral pathogens, host cell injury, and metabolic perturbations awaits better characterization in the future. New technical advances in multidimensional single-cell analysis and single-molecule methods will accelerate the rate of new discoveries. The ultimate goal is to manipulate MAVS activities in the form of immune-modulatory therapies to combat infections and sepsis. Antioxid. Redox Signal. 35, 1376-1392.
Collapse
Affiliation(s)
- Arjun Sharma
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Konstantinos Kontodimas
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
15
|
The C-Terminal Domain of Salmonid Alphavirus Nonstructural Protein 2 (nsP2) Is Essential and Sufficient To Block RIG-I Pathway Induction and Interferon-Mediated Antiviral Response. J Virol 2021; 95:e0115521. [PMID: 34523969 DOI: 10.1128/jvi.01155-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Salmonid alphavirus (SAV) is an atypical alphavirus that has a considerable impact on salmon and trout farms. Unlike other alphaviruses, such as the chikungunya virus, SAV is transmitted without an arthropod vector, and it does not cause cell shutoff during infection. The mechanisms by which SAV escapes the host immune system remain unknown. By studying the role of SAV proteins on the RIG-I signaling cascade, the first line of defense of the immune system during infection, we demonstrated that nonstructural protein 2 (nsP2) effectively blocks the induction of type I interferon (IFN). This inhibition, independent of the protease activity carried by nsP2, occurs downstream of IRF3, which is the transcription factor allowing the activation of the IFN promoter and its expression. The inhibitory effect of nsP2 on the RIG-I pathway depends on the localization of nsP2 in the host cell nucleus, which is linked to two nuclear localization sequences (NLS) located in its C-terminal part. The C-terminal domain of nsP2 by itself is sufficient and necessary to block IFN induction. Mutation of the NLS of nsP2 is deleterious to the virus. Finally, nsP2 does not interact with IRF3, indicating that its action is possible through a targeted interaction within discrete areas of chromatin, as suggested by its punctate distribution observed in the nucleus. These results therefore demonstrate a major role for nsP2 in the control by SAV of the host cell's innate immune response. IMPORTANCE The global consumption of fish continues to rise, and the future demand cannot be met by capture fisheries alone due to limited stocks of wild fish. Aquaculture is currently the world's fastest-growing food production sector, with an annual growth rate of 6 to 8%. Recurrent outbreaks of SAV result in significant economic losses with serious environmental consequences for wild stocks. While the clinical and pathological signs of SAV infection are fairly well known, the molecular mechanisms involved are poorly described. In the present study, we focus on the nonstructural protein nsP2 and characterize a specific domain containing nuclear localization sequences that are critical for the inhibition of the host innate immune response mediated by the RIG-I pathway.
Collapse
|
16
|
Guerrero-Arguero I, Tellez-Freitas CM, Weber KS, Berges BK, Robison RA, Pickett BE. Alphaviruses: Host pathogenesis, immune response, and vaccine & treatment updates. J Gen Virol 2021; 102. [PMID: 34435944 DOI: 10.1099/jgv.0.001644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human pathogens belonging to the Alphavirus genus, in the Togaviridae family, are transmitted primarily by mosquitoes. The signs and symptoms associated with these viruses include fever and polyarthralgia, defined as joint pain and inflammation, as well as encephalitis. In the last decade, our understanding of the interactions between members of the alphavirus genus and the human host has increased due to the re-appearance of the chikungunya virus (CHIKV) in Asia and Europe, as well as its emergence in the Americas. Alphaviruses affect host immunity through cytokines and the interferon response. Understanding alphavirus interactions with both the innate immune system as well as the various cells in the adaptive immune systems is critical to developing effective therapeutics. In this review, we summarize the latest research on alphavirus-host cell interactions, underlying infection mechanisms, and possible treatments.
Collapse
Affiliation(s)
- Israel Guerrero-Arguero
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA.,Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Bradford K Berges
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Richard A Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Brett E Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| |
Collapse
|
17
|
Brezgin S, Kostyusheva A, Bayurova E, Volchkova E, Gegechkori V, Gordeychuk I, Glebe D, Kostyushev D, Chulanov V. Immunity and Viral Infections: Modulating Antiviral Response via CRISPR-Cas Systems. Viruses 2021; 13:1373. [PMID: 34372578 PMCID: PMC8310348 DOI: 10.3390/v13071373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/13/2022] Open
Abstract
Viral infections cause a variety of acute and chronic human diseases, sometimes resulting in small local outbreaks, or in some cases spreading across the globe and leading to global pandemics. Understanding and exploiting virus-host interactions is instrumental for identifying host factors involved in viral replication, developing effective antiviral agents, and mitigating the severity of virus-borne infectious diseases. The diversity of CRISPR systems and CRISPR-based tools enables the specific modulation of innate immune responses and has contributed impressively to the fields of virology and immunology in a very short time. In this review, we describe the most recent advances in the use of CRISPR systems for basic and translational studies of virus-host interactions.
Collapse
Affiliation(s)
- Sergey Brezgin
- National Medical Research Center of Tuberculosis and Infectious Diseases, Ministry of Health, 127994 Moscow, Russia; (S.B.); (A.K.); (V.C.)
- Institute of Immunology, Federal Medical Biological Agency, 115522 Moscow, Russia
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasiya Kostyusheva
- National Medical Research Center of Tuberculosis and Infectious Diseases, Ministry of Health, 127994 Moscow, Russia; (S.B.); (A.K.); (V.C.)
| | - Ekaterina Bayurova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (I.G.)
| | - Elena Volchkova
- Department of Infectious Diseases, Sechenov University, 119991 Moscow, Russia;
| | - Vladimir Gegechkori
- Department of Pharmaceutical and Toxicological Chemistry, Sechenov University, 119991 Moscow, Russia;
| | - Ilya Gordeychuk
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (I.G.)
- Department of Organization and Technology of Immunobiological Drugs, Sechenov University, 119991 Moscow, Russia
| | - Dieter Glebe
- National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Institute of Medical Virology, Justus Liebig University of Giessen, 35392 Giessen, Germany;
| | - Dmitry Kostyushev
- National Medical Research Center of Tuberculosis and Infectious Diseases, Ministry of Health, 127994 Moscow, Russia; (S.B.); (A.K.); (V.C.)
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Vladimir Chulanov
- National Medical Research Center of Tuberculosis and Infectious Diseases, Ministry of Health, 127994 Moscow, Russia; (S.B.); (A.K.); (V.C.)
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
- Department of Infectious Diseases, Sechenov University, 119991 Moscow, Russia;
| |
Collapse
|
18
|
Kril V, Aïqui-Reboul-Paviet O, Briant L, Amara A. New Insights into Chikungunya Virus Infection and Pathogenesis. Annu Rev Virol 2021; 8:327-347. [PMID: 34255544 DOI: 10.1146/annurev-virology-091919-102021] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne alphavirus responsible for major outbreaks of disease since 2004 in the Indian Ocean islands, South east Asia, and the Americas. CHIKV causes debilitating musculoskeletal disorders in humans that are characterized by fever, rash, polyarthralgia, and myalgia. The disease is often self-limiting and nonlethal; however, some patients experience atypical or severe clinical manifestations, as well as a chronic rheumatic syndrome. Unfortunately, no efficient antivirals against CHIKV infection are available so far, highlighting the importance of deepening our knowledge of CHIKV host cell interactions and viral replication strategies. In this review, we discuss recent breakthroughs in the molecular mechanisms that regulate CHIKV infection and lay down the foundations to understand viral pathogenesis. We describe the role of the recently identified host factors co-opted by the virus for infection and pathogenesis, and emphasize the importance of CHIKV nonstructural proteins in both replication complex assembly and host immune response evasion. Expected final online publication date for the Annual Review of Virology, Volume 8 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Vasiliya Kril
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| | - Olivier Aïqui-Reboul-Paviet
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Laurence Briant
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Ali Amara
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| |
Collapse
|
19
|
Franz S, Pott F, Zillinger T, Schüler C, Dapa S, Fischer C, Passos V, Stenzel S, Chen F, Döhner K, Hartmann G, Sodeik B, Pessler F, Simmons G, Drexler JF, Goffinet C. Human IFITM3 restricts chikungunya virus and Mayaro virus infection and is susceptible to virus-mediated counteraction. Life Sci Alliance 2021; 4:e202000909. [PMID: 34078739 PMCID: PMC8200292 DOI: 10.26508/lsa.202000909] [Citation(s) in RCA: 11] [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: 09/16/2020] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 11/29/2022] Open
Abstract
Interferon-induced transmembrane (IFITM) proteins restrict membrane fusion and virion internalization of several enveloped viruses. The role of IFITM proteins during alphaviral infection of human cells and viral counteraction strategies are insufficiently understood. Here, we characterized the impact of human IFITMs on the entry and spread of chikungunya virus and Mayaro virus and provide first evidence for a CHIKV-mediated antagonism of IFITMs. IFITM1, 2, and 3 restricted infection at the level of alphavirus glycoprotein-mediated entry, both in the context of direct infection and cell-to-cell transmission. Relocalization of normally endosomal IFITM3 to the plasma membrane resulted in loss of antiviral activity. rs12252-C, a naturally occurring variant of IFITM3 that may associate with severe influenza in humans, restricted CHIKV, MAYV, and influenza A virus infection as efficiently as wild-type IFITM3 Antivirally active IFITM variants displayed reduced cell surface levels in CHIKV-infected cells involving a posttranscriptional process mediated by one or several nonstructural protein(s) of CHIKV. Finally, IFITM3-imposed reduction of specific infectivity of nascent particles provides a rationale for the necessity of a virus-encoded counteraction strategy against this restriction factor.
Collapse
Affiliation(s)
- Sergej Franz
- Institute of Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- Vitalant Research Institute, San Francisco, CA, USA
| | - Fabian Pott
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Zillinger
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital, Venusberg-Campus 1, Bonn, Germany
| | - Christiane Schüler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra Dapa
- Institute of Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Carlo Fischer
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Vânia Passos
- Institute of Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Saskia Stenzel
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fangfang Chen
- Research Group Biomarkers for Infectious Diseases, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hanover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hanover, Germany
| | - Katinka Döhner
- Institute of Virology, Hannover Medical School, Hanover, Germany
| | - Gunther Hartmann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hanover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Frank Pessler
- Research Group Biomarkers for Infectious Diseases, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hanover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hanover, Germany
| | | | - Jan Felix Drexler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
| | - Christine Goffinet
- Institute of Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
20
|
Ahmed SK, Haese NN, Cowan JT, Pathak V, Moukha-Chafiq O, Smith VJ, Rodzinak KJ, Ahmad F, Zhang S, Bonin KM, Streblow AD, Streblow CE, Kreklywich CN, Morrison C, Sarkar S, Moorman N, Sander W, Allen R, DeFilippis V, Tekwani BL, Wu M, Hirsch AJ, Smith JL, Tower NA, Rasmussen L, Bostwick R, Maddry JA, Ananthan S, Gerdes JM, Augelli-Szafran CE, Suto MJ, Morrison TE, Heise MT, Streblow DN, Pathak AK. Targeting Chikungunya Virus Replication by Benzoannulene Inhibitors. J Med Chem 2021; 64:4762-4786. [PMID: 33835811 PMCID: PMC9774970 DOI: 10.1021/acs.jmedchem.0c02183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A benzo[6]annulene, 4-(tert-butyl)-N-(3-methoxy-5,6,7,8-tetrahydronaphthalen-2-yl) benzamide (1a), was identified as an inhibitor against Chikungunya virus (CHIKV) with antiviral activity EC90 = 1.45 μM and viral titer reduction (VTR) of 2.5 log at 10 μM with no observed cytotoxicity (CC50 = 169 μM) in normal human dermal fibroblast cells. Chemistry efforts to improve potency, efficacy, and drug-like properties of 1a resulted in a novel lead compound 8q, which possessed excellent cellular antiviral activity (EC90 = 270 nM and VTR of 4.5 log at 10 μM) and improved liver microsomal stability. CHIKV resistance to an analog of 1a, compound 1c, tracked to a mutation in the nsP3 macrodomain. Further mechanism of action studies showed compounds working through inhibition of human dihydroorotate dehydrogenase in addition to CHIKV nsP3 macrodomain. Moderate efficacy was observed in an in vivo CHIKV challenge mouse model for compound 8q as viral replication was rescued from the pyrimidine salvage pathway.
Collapse
Affiliation(s)
| | | | - Jaden T. Cowan
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Vibha Pathak
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Omar Moukha-Chafiq
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Valerie J. Smith
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Kevin J. Rodzinak
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Fahim Ahmad
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Sixue Zhang
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Kiley M. Bonin
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Aaron D. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Cassilyn E. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Clayton Morrison
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Sanjay Sarkar
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Nathaniel Moorman
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Wes Sander
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Robbie Allen
- Oregon Translational Research and Development Institute, Portland, Oregon 97239, United States
| | - Victor DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Babu L. Tekwani
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Mousheng Wu
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Alec J. Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Jessica L. Smith
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Nichole A. Tower
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Lynn Rasmussen
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Robert Bostwick
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Joseph A. Maddry
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Subramaniam Ananthan
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - John M Gerdes
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | | | - Mark J. Suto
- Drug Discovery Division, Southern Research, Birmingham, Alabama 35205, United States
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Mark T. Heise
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Ashish K. Pathak
- Drug Discovery Division, Southern, Research, Birmingham, Alabama 35205, United States
| |
Collapse
|
21
|
Plasmacytoid Dendritic Cells Mediate Control of Ross River Virus Infection via a Type I Interferon-Dependent, MAVS-Independent Mechanism. J Virol 2021; 95:JVI.01538-20. [PMID: 33361425 DOI: 10.1128/jvi.01538-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/15/2020] [Indexed: 11/20/2022] Open
Abstract
Ross River virus (RRV) is a mosquito-borne alphavirus that causes epidemics of debilitating musculoskeletal disease. To define the innate immune mechanisms that mediate control of RRV infection, we studied a RRV strain encoding 6 nonsynonymous mutations in nsP1 (RRV-T48-nsP16M) that is attenuated in wild-type (WT) mice and Rag1 -/- mice, which are unable to mount adaptive immune responses, but not in mice that lack the capacity to respond to type I interferon (IFN) (Ifnar1 -/- mice). Utilizing this attenuated strain, our prior studies revealed that mitochondrial antiviral signaling (MAVS)-dependent production of type I IFN by Ly6Chi monocytes is critical for control of acute RRV infection. Here, we infected Mavs -/- mice with either WT RRV or RRV-T48-nsP16M to elucidate MAVS-independent protective mechanisms. Mavs -/- mice infected with WT RRV developed severe disease and succumbed to infection, whereas those infected with RRV-T48-nsP16M exhibited minimal disease signs. Mavs -/- mice infected with RRV-T48-nsP16M had higher levels of systemic type I IFN than Mavs -/- mice infected with WT virus, and treatment of Mavs -/- mice infected with the attenuated nsP1 mutant virus with an IFNAR1-blocking antibody resulted in a lethal infection. In vitro, type I IFN expression was induced in plasmacytoid dendritic cells (pDCs) cocultured with RRV-infected cells in a MAVS-independent manner, and depletion of pDCs in Mavs -/- mice resulted in increased viral burdens in joint and muscle tissues, suggesting that pDCs are a source of the protective IFN in Mavs -/- mice. These data suggest that pDC production of type I IFN through a MAVS-independent pathway contributes to control of RRV infection.IMPORTANCE Arthritogenic alphaviruses, including Ross River virus (RRV), are human pathogens that cause debilitating acute and chronic musculoskeletal disease and are a significant public health burden. Using an attenuated RRV with enhanced susceptibility to host innate immune responses has revealed key cellular and molecular mechanisms that can mediate control of attenuated RRV infection and that are evaded by more virulent RRV strains. In this study, we found that pDCs contribute to the protective type I interferon response during RRV infection through a mechanism that is independent of the mitochondrial antiviral signaling (MAVS) adaptor protein. These findings highlight a key innate immune mechanism that contributes to control of alphavirus infections.
Collapse
|
22
|
Raghuvanshi R, Bharate SB. Recent Developments in the Use of Kinase Inhibitors for Management of Viral Infections. J Med Chem 2021; 65:893-921. [PMID: 33539089 DOI: 10.1021/acs.jmedchem.0c01467] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Kinases are a group of therapeutic targets involved in the progression of numerous diseases, including cancer, rheumatoid arthritis, Alzheimer's disease, and viral infections. The majority of approved antiviral agents are inhibitors of virus-specific targets that are encoded by individual viruses. These inhibitors are narrow-spectrum agents that can cause resistance development. Viruses are dependent on host cellular proteins, including kinases, for progression of their life-cycle. Thus, targeting kinases is an important therapeutic approach to discovering broad-spectrum antiviral agents. As there are a large number of FDA approved kinase inhibitors for various indications, their repurposing for viral infections is an attractive and time-sparing strategy. Many kinase inhibitors, including baricitinib, ruxolitinib, imatinib, tofacitinib, pacritinib, zanubrutinib, and ibrutinib, are under clinical investigation for COVID-19. Herein, we discuss FDA approved kinase inhibitors, along with a repertoire of clinical/preclinical stage kinase inhibitors that possess antiviral activity or are useful in the management of viral infections.
Collapse
Affiliation(s)
- Rinky Raghuvanshi
- Medicinal Chemistry Division,CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.,Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Sandip B Bharate
- Medicinal Chemistry Division,CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.,Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
23
|
Boussier J, Levi L, Weger-Lucarelli J, Poirier EZ, Vignuzzi M, Albert ML. Chikungunya virus superinfection exclusion is mediated by a block in viral replication and does not rely on non-structural protein 2. PLoS One 2020; 15:e0241592. [PMID: 33180795 PMCID: PMC7660575 DOI: 10.1371/journal.pone.0241592] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/16/2020] [Indexed: 01/20/2023] Open
Abstract
Superinfection exclusion (SIE) is a process by which a virally infected cell is protected from subsequent infection by the same or a closely related virus. By preventing cell coinfection, SIE favors preservation of genome integrity of a viral strain and limits its recombination potential with other viral genomes, thereby impacting viral evolution. Although described in virtually all viral families, the precise step(s) impacted by SIE during the viral life cycle have not been systematically explored. Here, we describe for the first time SIE triggered by chikungunya virus (CHIKV), an alphavirus of public health importance. Using single-cell technologies, we demonstrate that CHIKV excludes subsequent infection with: CHIKV; Sindbis virus, a related alphavirus; and influenza A, an unrelated RNA virus. We further demonstrate that SIE does not depend on the action of type I interferon, nor does it rely on host cell transcription. Moreover, exclusion is not mediated by the action of a single CHIKV protein; in particular, we observed no role for non-structural protein 2 (nsP2), making CHIKV unique among characterized alphaviruses. By stepping through the viral life cycle, we show that CHIKV exclusion occurs at the level of replication, but does not directly influence virus binding, nor viral structural protein translation. In sum, we characterized co-infection during CHIKV replication, which likely influences the rate of viral diversification and evolution.
Collapse
Affiliation(s)
- Jeremy Boussier
- Immubiology of Dendritic Cells unit, Institut Pasteur, Paris, France.,Inserm U1223, Institut Pasteur, Paris, France.,École doctorale Frontières du Vivant, Université Paris Diderot, Paris, France
| | - Laura Levi
- Viral Population and Pathogenesis Unit, Institut Pasteur, Paris, France
| | - James Weger-Lucarelli
- Viral Population and Pathogenesis Unit, Institut Pasteur, Paris, France.,Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, United States of America
| | - Enzo Z Poirier
- Viral Population and Pathogenesis Unit, Institut Pasteur, Paris, France.,Immunobiology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Marco Vignuzzi
- Viral Population and Pathogenesis Unit, Institut Pasteur, Paris, France
| | - Matthew L Albert
- Immubiology of Dendritic Cells unit, Institut Pasteur, Paris, France.,Insitro, South San Francisco, CA, United States of America
| |
Collapse
|
24
|
Felipe VLJ, Paula A V, Silvio UI. Chikungunya virus infection induces differential inflammatory and antiviral responses in human monocytes and monocyte-derived macrophages. Acta Trop 2020; 211:105619. [PMID: 32634389 DOI: 10.1016/j.actatropica.2020.105619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/03/2020] [Accepted: 07/03/2020] [Indexed: 01/09/2023]
Abstract
Chikungunya virus (CHIKV) is a zoonotic arthropod-borne virus that has caused several outbreaks in tropical and subtropical areas worldwide during the last 50 years. The virus is known to target different human cell types throughout the course of infection including epithelial and endothelial cells, fibroblasts, primary monocytes and monocyte-derived macrophages (MDMs). The two latter are phagocytic cell populations of the innate immune system which are involved in some aspects of CHIKV pathogenesis. However, monocytes and macrophages also potentially contribute to the control of viral replication through the expression of different pattern recognition receptors sensing viral pathogens and subsequently, inducing an type I interferone (IFN-I)-dependent antiviral immune response. The aim of this study was to determine the modulation of the expression of Toll-like receptors (TLRs), cytokine secretion capabilities and antiviral factor production in monocytes and MDMs following infection with CHIKV. Moreover, we sought to determine the replication kinetics of CHIKV in these two cell populations. We found that the maximum peak of CHIKV replication was observed between 18- and 24-hours post-infection (hpi), while after that the is strongly reduced. Furthermore, CHIKV infection induced the pro-inflammatory cytokine production starting from the first 6 hpi in both monocytes and MDMs, with similar kinetics but different protein levels. In contrast, the kinetics of transcriptional expression of some TLRs were different between both cell types. In addition, IFN-I, 2',5'-oligoadenylate synthetase 1 (OAS1), and double-stranded RNA-activated protein kinase R (PKR) mRNA levels were detected in response to CHIKV infection of monocytes and MDMs, resulting the highest expression levels at 48 hpi. In conclusion, our data provides evidence that CHIKV infection activates the TLR pathways in primary monocytes and MDMs, which play a crucial role in CHIKV pathogenesis and/or host defense, differentially. However, additional studies are required to determine the functional role of TLRs in monocytes and MDMs.
Collapse
Affiliation(s)
- Valdés López Juan Felipe
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Velilla Paula A
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Urcuqui-Inchima Silvio
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
25
|
Webb LG, Veloz J, Pintado-Silva J, Zhu T, Rangel MV, Mutetwa T, Zhang L, Bernal-Rubio D, Figueroa D, Carrau L, Fenutria R, Potla U, Reid SP, Yount JS, Stapleford KA, Aguirre S, Fernandez-Sesma A. Chikungunya virus antagonizes cGAS-STING mediated type-I interferon responses by degrading cGAS. PLoS Pathog 2020; 16:e1008999. [PMID: 33057424 PMCID: PMC7591055 DOI: 10.1371/journal.ppat.1008999] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 10/27/2020] [Accepted: 09/21/2020] [Indexed: 12/24/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus known to cause epidemics resulting in predominantly symptomatic infections, which in rare cases cause long term debilitating arthritis and arthralgia. Significant progress has been made in understanding the roles of canonical RNA sensing pathways in the host recognition of CHIKV; however, less is known regarding antagonism of CHIKV by cytosolic DNA sensing pathways like that of cyclic GMP-AMP synthase (cGAS) and Stimulator of Interferon Genes (STING). With the use of cGAS or STING null cells we demonstrate that the pathway restricts CHIKV replication in fibroblasts and immune cells. We show that DNA accumulates in the cytoplasm of infected cells and that CHIKV blocks DNA dependent IFN-β transcription. This antagonism of DNA sensing is via an early autophagy-mediated degradation of cGAS and expression of the CHIKV capsid protein is sufficient to induce cGAS degradation. Furthermore, we identify an interaction of CHIKV nsP1 with STING and map the interaction to 23 residues in the cytosolic loop of the adaptor protein. This interaction stabilizes the viral protein and increases the level of palmitoylated nsP1 in cells. Together, this work supports previous publications highlighting the relevance of the cGAS-STING pathway in the early detection of (+)ssRNA viruses and provides direct evidence that CHIKV interacts with and antagonizes cGAS-STING signaling.
Collapse
Affiliation(s)
- L. G. Webb
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - J. Veloz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - J. Pintado-Silva
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - T. Zhu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - M. V. Rangel
- Department of Microbiology, New York University School of Medicine, New York, NY, United States of America
| | - T. Mutetwa
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - L. Zhang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States of America
| | - D. Bernal-Rubio
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - D. Figueroa
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - L. Carrau
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - R. Fenutria
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - U. Potla
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - St. P. Reid
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - J. S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States of America
| | - K. A. Stapleford
- Department of Microbiology, New York University School of Medicine, New York, NY, United States of America
| | - S. Aguirre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - A. Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
26
|
Rivera-Serrano EE, Gizzi AS, Arnold JJ, Grove TL, Almo SC, Cameron CE. Viperin Reveals Its True Function. Annu Rev Virol 2020; 7:421-446. [PMID: 32603630 DOI: 10.1146/annurev-virology-011720-095930] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Most cells respond to viral infections by activating innate immune pathways that lead to the induction of antiviral restriction factors. One such factor, viperin, was discovered almost two decades ago based on its induction during viral infection. Since then, viperin has been shown to possess activity against numerous viruses via multiple proposed mechanisms. Most recently, however, viperin was demonstrated to catalyze the conversion of cytidine triphosphate (CTP) to 3'-deoxy-3',4'-didehydro-CTP (ddhCTP), a previously unknown ribonucleotide. Incorporation of ddhCTP causes premature termination of RNA synthesis by the RNA-dependent RNA polymerase of some viruses. To date, production of ddhCTP by viperin represents the only activity of viperin that links its enzymatic activity directly to an antiviral mechanism in human cells. This review examines the multiple antiviral mechanisms and biological functions attributed to viperin.
Collapse
Affiliation(s)
- Efraín E Rivera-Serrano
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Anthony S Gizzi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, USA; , .,Department of Pharmacology, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Jamie J Arnold
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Tyler L Grove
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, USA; ,
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, USA; ,
| | - Craig E Cameron
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| |
Collapse
|
27
|
Cunha MS, Costa PAG, Correa IA, de Souza MRM, Calil PT, da Silva GPD, Costa SM, Fonseca VWP, da Costa LJ. Chikungunya Virus: An Emergent Arbovirus to the South American Continent and a Continuous Threat to the World. Front Microbiol 2020; 11:1297. [PMID: 32670231 PMCID: PMC7332961 DOI: 10.3389/fmicb.2020.01297] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/20/2020] [Indexed: 01/23/2023] Open
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne virus (arbovirus) of epidemic concern, transmitted by Aedes ssp. mosquitoes, and is the etiologic agent of a febrile and incapacitating arthritogenic illness responsible for millions of human cases worldwide. After major outbreaks starting in 2004, CHIKV spread to subtropical areas and western hemisphere coming from sub-Saharan Africa, South East Asia, and the Indian subcontinent. Even though CHIKV disease is self-limiting and non-lethal, more than 30% of the infected individuals will develop chronic disease with persistent severe joint pain, tenosynovitis, and incapacitating polyarthralgia that can last for months to years, negatively impacting an individual's quality of life and socioeconomic productivity. The lack of specific drugs or licensed vaccines to treat or prevent CHIKV disease associated with the global presence of the mosquito vector in tropical and temperate areas, representing a possibility for CHIKV to continually spread to different territories, make this virus an agent of public health burden. In South America, where Dengue virus is endemic and Zika virus was recently introduced, the impact of the expansion of CHIKV infections, and co-infection with other arboviruses, still needs to be estimated. In Brazil, the recent spread of the East/Central/South Africa (ECSA) and Asian genotypes of CHIKV was accompanied by a high morbidity rate and acute cases of abnormal disease presentation and severe neuropathies, which is an atypical outcome for this infection. In this review, we will discuss what is currently known about CHIKV epidemics, clinical manifestations of the human disease, the basic concepts and recent findings in the mechanisms underlying virus-host interaction, and CHIKV-induced chronic disease for both in vitro and in vivo models of infection. We aim to stimulate scientific debate on how the characterization of replication, host-cell interactions, and the pathogenic potential of the new epidemic viral strains can contribute as potential developments in the virology field and shed light on strategies for disease control.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Luciana J. da Costa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Srivastava P, Kumar A, Hasan A, Mehta D, Kumar R, Sharma C, Sunil S. Disease Resolution in Chikungunya-What Decides the Outcome? Front Immunol 2020; 11:695. [PMID: 32411133 PMCID: PMC7198842 DOI: 10.3389/fimmu.2020.00695] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Chikungunya disease (CHIKD) is a viral infection caused by an alphavirus, chikungunya virus (CHIKV), and triggers large outbreaks leading to epidemics. Despite the low mortality rate, it is a major public health concern owing to high morbidity in affected individuals. The complete spectrum of this disease can be divided into four phases based on its clinical presentation and immunopathology. When a susceptible individual is bitten by an infected mosquito, the bite triggers inflammatory responses attracting neutrophils and initiating a cascade of events, resulting in the entry of the virus into permissive cells. This phase is termed the pre-acute or the intrinsic incubation phase. The virus utilizes the cellular components of the innate immune system to enter into circulation and reach primary sites of infection such as the lymph nodes, spleen, and liver. Also, at this point, antigen-presenting cells (APCs) present the viral antigens to the T cells thereby activating and initiating adaptive immune responses. This phase is marked by the exhibition of clinical symptoms such as fever, rashes, arthralgia, and myalgia and is termed the acute phase of the disease. Viremia reaches its peak during this phase, thereby enhancing the antigen-specific host immune response. Simultaneously, T cell-mediated activation of B cells leads to the formation of CHIKV specific antibodies. Increase in titres of neutralizing IgG/IgM antibodies results in the clearance of virus from the bloodstream and marks the initiation of the post-acute phase. Immune responses mounted during this phase of the infection determine the degree of disease progression or its resolution. Some patients may progress to a chronic arthritic phase of the disease that may last from a few months to several years, owing to a compromised disease resolution. The present review discusses the immunopathology of CHIKD and the factors that dictate disease progression and its resolution.
Collapse
Affiliation(s)
- Priyanshu Srivastava
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Ankit Kumar
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Abdul Hasan
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Divya Mehta
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Ramesh Kumar
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Chetan Sharma
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sujatha Sunil
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| |
Collapse
|
29
|
Li M, Liao Z, Xu Z, Zou X, Wang Y, Peng H, Li Y, Ou X, Deng Y, Guo Y, Gan W, Peng T, Chen D, Cai M. The Interaction Mechanism Between Herpes Simplex Virus 1 Glycoprotein D and Host Antiviral Protein Viperin. Front Immunol 2019; 10:2810. [PMID: 31921110 PMCID: PMC6917645 DOI: 10.3389/fimmu.2019.02810] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/15/2019] [Indexed: 12/17/2022] Open
Abstract
Viperin is an interferon-inducible protein that responsible for a variety of antiviral responses to different viruses. Our previous study has shown that the ribonuclease UL41 of herpes simplex virus 1 (HSV-1) can degrade the mRNA of viperin to promote HSV-1 replication. However, it is not clear whether other HSV-1 encoded proteins can regulate the function of viperin. Here, one novel viperin associated protein, glycoprotein D (gD), was identified. To verify the interaction between gD and viperin, gD and viperin expression plasmids were firstly co-transfected into COS-7 cells, and fluorescence microscope showed they co-localized at the perinuclear region, then this potential interaction was confirmed by co-immunoprecipitation (Co-IP) assays. Moreover, confocal microscopy demonstrated that gD and viperin co-localized at the Golgi body and lipid droplets. Furthermore, dual-luciferase reporter and Co-IP assays showed gD and viperin interaction leaded to the increase of IRF7-mediated IFN-β expression through promoting viperin and IRAK1 interaction and facilitating K63-linked IRAK1 polyubiquitination. Nevertheless, gD inhibited TRAF6-induced NF-κB activity by decreasing the interaction of viperin and TRAF6. In addition, gD restrained viperin-mediated interaction between IRAK1 and TRAF6. Eventually, gD and viperin interaction was corroborated to significantly inhibit the proliferation of HSV-1. Taken together, this study would open up new avenues toward delineating the function and physiological significance of gD and viperin during HSV-1 replication cycle.
Collapse
Affiliation(s)
- Meili Li
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Zongmin Liao
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.,Department of Scientific Research and Education, Yuebei People's Hospital, Shaoguan, China
| | - Zuo Xu
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Xingmei Zou
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yuanfang Wang
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Hao Peng
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yiwen Li
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Ou
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yangxi Deng
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yingjie Guo
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Weidong Gan
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Tao Peng
- State Key Laboratory of Respiratory Diseases, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.,South China Vaccine Corporation Limited, Guangzhou, China
| | - Daixiong Chen
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Mingsheng Cai
- Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Helbig KJ, Teh MY, Crosse KM, Monson EA, Smith M, Tran EN, Standish AJ, Morona R, Beard MR. The interferon stimulated gene viperin, restricts Shigella. flexneri in vitro. Sci Rep 2019; 9:15598. [PMID: 31666594 PMCID: PMC6821890 DOI: 10.1038/s41598-019-52130-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/14/2019] [Indexed: 01/08/2023] Open
Abstract
The role of interferon and interferon stimulated genes (ISG) in limiting bacterial infection is controversial, and the role of individual ISGs in the control of the bacterial life-cycle is limited. Viperin, is a broad acting anti-viral ISGs, which restricts multiple viral pathogens with diverse mechanisms. Viperin is upregulated early in some bacterial infections, and using the intracellular bacterial pathogen, S. flexneri, we have shown for the first time that viperin inhibits the intracellular bacterial life cycle. S. flexneri replication in cultured cells induced a predominantly type I interferon response, with an early increase in viperin expression. Ectopic expression of viperin limited S. flexneri cellular numbers by as much as 80% at 5hrs post invasion, with similar results also obtained for the intracellular pathogen, Listeria monocytogenes. Analysis of viperins functional domains required for anti-bacterial activity revealed the importance of both viperin's N-terminal, and its radical SAM enzymatic function. Live imaging of S. flexneri revealed impeded entry into viperin expressing cells, which corresponded to a loss of cellular cholesterol. This data further defines viperin's multi-functional role, to include the ability to limit intracellular bacteria; and highlights the role of ISGs and the type I IFN response in the control of bacterial pathogens.
Collapse
Affiliation(s)
- K J Helbig
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia.
| | - M Y Teh
- Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia
| | - K M Crosse
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - E A Monson
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - M Smith
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - E N Tran
- Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia
| | - A J Standish
- Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia
| | - R Morona
- Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia
| | - M R Beard
- Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia
| |
Collapse
|
31
|
Nelemans T, Kikkert M. Viral Innate Immune Evasion and the Pathogenesis of Emerging RNA Virus Infections. Viruses 2019; 11:v11100961. [PMID: 31635238 PMCID: PMC6832425 DOI: 10.3390/v11100961] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Positive-sense single-stranded RNA (+ssRNA) viruses comprise many (re-)emerging human pathogens that pose a public health problem. Our innate immune system and, in particular, the interferon response form the important first line of defence against these viruses. Given their genetic flexibility, these viruses have therefore developed multiple strategies to evade the innate immune response in order to optimize their replication capacity. Already many molecular mechanisms of innate immune evasion by +ssRNA viruses have been identified. However, research addressing the effect of host innate immune evasion on the pathology caused by viral infections is less prevalent in the literature, though very relevant and interesting. Since interferons have been implicated in inflammatory diseases and immunopathology in addition to their protective role in infection, antagonizing the immune response may have an ambiguous effect on the clinical outcome of the viral disease. Therefore, this review discusses what is currently known about the role of interferons and host immune evasion in the pathogenesis of emerging coronaviruses, alphaviruses and flaviviruses.
Collapse
Affiliation(s)
- Tessa Nelemans
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands.
| | - Marjolein Kikkert
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands.
| |
Collapse
|
32
|
Valdés López JF, Velilla PA, Urcuqui-Inchima S. Chikungunya Virus and Zika Virus, Two Different Viruses Examined with a Common Aim: Role of Pattern Recognition Receptors on the Inflammatory Response. J Interferon Cytokine Res 2019; 39:507-521. [DOI: 10.1089/jir.2019.0058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
| | - Paula Andrea Velilla
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
33
|
Kang LJ, Nguyen KVA, Eom S, Choi YJ, Nguyen CN, Lee J, Kim C, Lee S, Lee SG, Lee JH. Stimulating DDX3 expression by serotonin 5-HT receptor 7 through phosphorylation of p53 via the AC-PKA-ERK signaling pathway. J Cell Biochem 2019; 120:18193-18208. [PMID: 31172579 DOI: 10.1002/jcb.29125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 01/22/2023]
Abstract
DDX3 is a host viral factor that can inhibit the hepatitis B virus-induced innate immune responses. In this study, the 20 bioactive compounds have screened the effects on DDX3 and we found that 5-HT upregulated DDX3 promoter activity via the 5-HT7 receptor on liver hepatocellular cells (HepG2 cells) by using a luciferase assay, reverse transcription-polymerase chain reaction analysis, and Western blot analysis. Furthermore, we are trying to elucidate the pathways involved in the stimulating effect of 5-HT on DDX3 expression to induce innate immune responses against hepatitis B virus infection. A knockdown of the 5-HT7 receptor by transfection si-5-HT7 receptors or si-control into HepG2 cells treated by 5-HT (or 5-HT plus agonist) confirmed the role of the 5-HT7 receptor in DDX3 expression. The IFN-β-Luc expression and level of hepatitis B virus surface Antigen (HBsAg) showed that DDX3 mediated by the 5-HT7 agonist (AS-19) increased IFN-β expression and inhibited HBV replication. Luciferase assays showed the involvement of 5-HT7 receptors in DDX3 expression via cAMP/AC/PKA pathways by using protein kinase A (PKA) and adenylyl cyclase inhibitor (MDL 12330A). AS-19 mediated DDX3 promoter activated PKA extracellular signal-regulated kinase ERK signaling the p53 phosphorylation (-1080/-1070) resulted in upregulation of DDX3 promoter transactivation via the 5-HT7 receptors agonist. Overall, 5-HT7 was found to be a new potential target to inhibit hepatitis B infection by activating AC/PKA/ERK pathways by phosphorylating p53 via the 5-HT7 agonist response by mediating DDX3 expression.
Collapse
Affiliation(s)
- Li-Jung Kang
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Khoa V A Nguyen
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Sanung Eom
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Yeo-Jin Choi
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Cam Ngoc Nguyen
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Jaeeun Lee
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Chaelin Kim
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Shinhui Lee
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Seong-Gene Lee
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| | - Jun-Ho Lee
- Department of Biotechnology, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
34
|
Src Family Kinase Inhibitors Block Translation of Alphavirus Subgenomic mRNAs. Antimicrob Agents Chemother 2019; 63:AAC.02325-18. [PMID: 30917980 PMCID: PMC6496153 DOI: 10.1128/aac.02325-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/14/2019] [Indexed: 01/01/2023] Open
Abstract
Alphaviruses are arthropod-transmitted RNA viruses that can cause arthralgia, myalgia, and encephalitis in humans. Since the role of cellular kinases in alphavirus replication is unknown, we profiled kinetic changes in host kinase abundance and phosphorylation following chikungunya virus (CHIKV) infection of fibroblasts. Alphaviruses are arthropod-transmitted RNA viruses that can cause arthralgia, myalgia, and encephalitis in humans. Since the role of cellular kinases in alphavirus replication is unknown, we profiled kinetic changes in host kinase abundance and phosphorylation following chikungunya virus (CHIKV) infection of fibroblasts. Based upon the results of this study, we treated CHIKV-infected cells with kinase inhibitors targeting the Src family kinase (SFK)–phosphatidylinositol 3-kinase (PI3K)–AKT–mTORC signaling pathways. Treatment of cells with SFK inhibitors blocked the replication of CHIKV as well as multiple other alphaviruses, including Mayaro virus, O’nyong-nyong virus, Ross River virus, and Venezuelan equine encephalitis virus. Dissecting the effect of SFK inhibition on alphavirus replication, we found that viral structural protein levels were significantly reduced, but synthesis of viral genomic and subgenomic RNAs was unaffected. By measuring the association of viral RNA with polyribosomes, we found that the SFK inhibitor dasatinib blocks alphavirus subgenomic RNA translation. Our results demonstrate a role for SFK signaling in alphavirus subgenomic RNA translation and replication. Targeting host factors involved in alphavirus replication represents an innovative, perhaps paradigm-shifting, strategy for exploring the replication of CHIKV and other alphaviruses while promoting antiviral therapeutic development.
Collapse
|
35
|
Mostafavi H, Abeyratne E, Zaid A, Taylor A. Arthritogenic Alphavirus-Induced Immunopathology and Targeting Host Inflammation as A Therapeutic Strategy for Alphaviral Disease. Viruses 2019; 11:v11030290. [PMID: 30909385 PMCID: PMC6466158 DOI: 10.3390/v11030290] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/25/2022] Open
Abstract
Arthritogenic alphaviruses are a group of medically important arboviruses that cause inflammatory musculoskeletal disease in humans with debilitating symptoms, such as arthralgia, arthritis, and myalgia. The arthritogenic, or Old World, alphaviruses are capable of causing explosive outbreaks, with some viruses of major global concern. At present, there are no specific therapeutics or commercially available vaccines available to prevent alphaviral disease. Infected patients are typically treated with analgesics and non-steroidal anti-inflammatory drugs to provide often inadequate symptomatic relief. Studies to determine the mechanisms of arthritogenic alphaviral disease have highlighted the role of the host immune system in disease pathogenesis. This review discusses the current knowledge of the innate immune response to acute alphavirus infection and alphavirus-induced immunopathology. Therapeutic strategies to treat arthritogenic alphavirus disease by targeting the host immune response are also examined.
Collapse
Affiliation(s)
- Helen Mostafavi
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia.
| | - Eranga Abeyratne
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia.
| | - Ali Zaid
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia.
| | - Adam Taylor
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, QLD 4222, Australia.
| |
Collapse
|
36
|
Hwang J, Jiang A, Fikrig E. Rev-erb Agonist Inhibits Chikungunya and O'nyong'nyong Virus Replication. Open Forum Infect Dis 2018; 5:ofy315. [PMID: 30568983 PMCID: PMC6293476 DOI: 10.1093/ofid/ofy315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/16/2018] [Indexed: 01/29/2023] Open
Abstract
Chikungunya virus (CHIKV), an alphavirus spread by Aedes spp. mosquitos, causes severe inflammation and joint pain, progressing to a chronic arthralgic state in a subset of patients. Due to recent global epidemics of CHIKV and the potential for related viruses to cause outbreaks, multiple approaches to combat these pathogens are of interest. We report that SR9009, a synthetic agonist of nuclear receptors Rev-erb α/β, inhibits replication of multiple alphaviruses (CHIKV and O'nyong'nyong virus) mainly by suppressing structural protein synthesis, although viral RNA accumulation is relatively unimpeded. Furthermore, SR9009 reduces the inflammatory response in cultured murine macrophages exposed to alphavirus-infected cells.
Collapse
Affiliation(s)
- Jesse Hwang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Alfred Jiang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut.,Howard Hughes Medical Institute, Chevy Chase, Maryland
| |
Collapse
|
37
|
Henss L, Scholz T, Grünweller A, Schnierle BS. Silvestrol Inhibits Chikungunya Virus Replication. Viruses 2018; 10:v10110592. [PMID: 30380742 PMCID: PMC6266838 DOI: 10.3390/v10110592] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/19/2018] [Accepted: 10/26/2018] [Indexed: 02/08/2023] Open
Abstract
Silvestrol, a natural compound that is isolated from plants of the genus Aglaia, is a specific inhibitor of the RNA helicase eIF4A, which unwinds RNA secondary structures in 5′-untranslated regions (UTRs) of mRNAs and allows translation. Silvestrol has a broad antiviral activity against multiple RNA virus families. Here, we show that silvestrol inhibits the replication of chikungunya virus (CHIKV), a positive single-stranded RNA virus. Silvestrol delayed the protein synthesis of non-structural (nsPs) and structural proteins, resulting in a delayed innate response to CHIKV infection. Interferon-α induced STAT1 phosphorylation was not inhibited nor did eIF2α become phosphorylated 16 h post infection in the presence of silvestrol. In addition, the host protein shut-off induced by CHIKV infection was decreased in silvestrol-treated cells. Silvestrol acts by limiting the amount of nsPs, and thereby reducing CHIKV RNA replication. From our results, we propose that inhibition of the host helicase eIF4A might have potential as a therapeutic strategy to treat CHIKV infections.
Collapse
Affiliation(s)
- Lisa Henss
- Paul-Ehrlich-Institut, Department of Virology, 63225 Langen, Germany.
| | - Tatjana Scholz
- Paul-Ehrlich-Institut, Department of Virology, 63225 Langen, Germany.
| | - Arnold Grünweller
- Institut für Pharmazeutische Chemie, Philipps-Universität Marburg, 35032 Marburg, Germany.
| | | |
Collapse
|
38
|
Gall B, Pryke K, Abraham J, Mizuno N, Botto S, Sali TM, Broeckel R, Haese N, Nilsen A, Placzek A, Morrison T, Heise M, Streblow D, DeFilippis V. Emerging Alphaviruses Are Sensitive to Cellular States Induced by a Novel Small-Molecule Agonist of the STING Pathway. J Virol 2018; 92:e01913-17. [PMID: 29263267 PMCID: PMC5827377 DOI: 10.1128/jvi.01913-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/12/2017] [Indexed: 01/23/2023] Open
Abstract
The type I interferon (IFN) system represents an essential innate immune response that renders cells resistant to virus growth via the molecular actions of IFN-induced effector proteins. IFN-mediated cellular states inhibit growth of numerous and diverse virus types, including those of known pathogenicity as well as potentially emerging agents. As such, targeted pharmacologic activation of the IFN response may represent a novel therapeutic strategy to prevent infection or spread of clinically impactful viruses. In light of this, we employed a high-throughput screen to identify small molecules capable of permeating the cell and of activating IFN-dependent signaling processes. Here we report the identification and characterization of N-(methylcarbamoyl)-2-{[5-(4-methylphenyl)-1,3,4-oxadiazol-2-yl]sulfanyl}-2-phenylacetamide (referred to as C11), a novel compound capable of inducing IFN secretion from human cells. Using reverse genetics-based loss-of-function assays, we show that C11 activates the type I IFN response in a manner that requires the adaptor protein STING but not the alternative adaptors MAVS and TRIF. Importantly, treatment of cells with C11 generated a cellular state that potently blocked replication of multiple emerging alphavirus types, including chikungunya, Ross River, Venezuelan equine encephalitis, Mayaro, and O'nyong-nyong viruses. The antiviral effects of C11 were subsequently abrogated in cells lacking STING or the type I IFN receptor, indicating that they are mediated, at least predominantly, by way of STING-mediated IFN secretion and subsequent autocrine/paracrine signaling. This work also allowed characterization of differential antiviral roles of innate immune signaling adaptors and IFN-mediated responses and identified MAVS as being crucial to cellular resistance to alphavirus infection.IMPORTANCE Due to the increase in emerging arthropod-borne viruses, such as chikungunya virus, that lack FDA-approved therapeutics and vaccines, it is important to better understand the signaling pathways that lead to clearance of virus. Here we show that C11 treatment makes human cells refractory to replication of a number of these viruses, which supports its value in increasing our understanding of the immune response and viral pathogenesis required to establish host infection. We also show that C11 depends on signaling through STING to produce antiviral type I interferon, which further supports its potential as a therapeutic drug or research tool.
Collapse
Affiliation(s)
- Bryan Gall
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Kara Pryke
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Jinu Abraham
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Nobuyo Mizuno
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Sara Botto
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Tina M Sali
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rebecca Broeckel
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Nicole Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Aaron Nilsen
- Veterans Affairs Medical Center, Portland, Oregon, USA
| | | | - Thomas Morrison
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Mark Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Daniel Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Victor DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
39
|
Chang AY, Martins KAO, Encinales L, Reid SP, Acuña M, Encinales C, Matranga CB, Pacheco N, Cure C, Shukla B, Ruiz Arteta T, Amdur R, Cazares LH, Gregory M, Ward MD, Porras A, Rico Mendoza A, Dong L, Kenny T, Brueggemann E, Downey LG, Kamalapathy P, Lichtenberger P, Falls O, Simon GL, Bethony JM, Firestein GS. Chikungunya Arthritis Mechanisms in the Americas: A Cross-Sectional Analysis of Chikungunya Arthritis Patients Twenty-Two Months After Infection Demonstrating No Detectable Viral Persistence in Synovial Fluid. Arthritis Rheumatol 2018; 70:585-593. [PMID: 29266856 DOI: 10.1002/art.40383] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/14/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To determine if chikungunya virus persists in synovial fluid after infection, potentially acting as a causative mechanism of persistent arthritis. METHODS We conducted a cross-sectional study of 38 Colombian participants with clinical chikungunya virus infection during the 2014-2015 epidemic who reported chronic arthritis and 10 location-matched controls without chikungunya virus or arthritis. Prior chikungunya virus infection status was serologically confirmed, and the presence of synovial fluid chikungunya virus, viral RNA, and viral proteins was determined by viral culture, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and mass spectrometry, respectively. Biomarkers were assessed by multiplex analysis. RESULTS Patients with serologically confirmed chikungunya arthritis (33 of 38 [87%]) were predominantly female (82%) and African Colombian (55%) or white Colombian (33%), with moderate disease activity (mean ± SD Disease Activity Score in 28 joints 4.52 ± 0.77) a median of 22 months after infection (interquartile range 21-23 months). Initial symptoms of chikungunya virus infection included joint pain (97%), swelling (97%), stiffness (91%), and fever (91%). The most commonly affected joints were the knees (87%), elbows (76%), wrists (75%), ankles (56%), fingers (56%), and toes (56%). Synovial fluid samples from all patients with chikungunya arthritis were negative for chikungunya virus on qRT-PCR, showed no viral proteins on mass spectrometry, and cultures were negative. Case and control plasma cytokine and chemokine concentrations did not differ significantly. CONCLUSION This is one of the largest observational studies involving analysis of the synovial fluid of chikungunya arthritis patients. Synovial fluid analysis revealed no detectable chikungunya virus. This finding suggests that chikungunya virus may cause arthritis through induction of potential host autoimmunity, suggesting a role for immunomodulating agents in the treatment of chikungunya arthritis, or that low-level viral persistence exists in synovial tissue only and is undetectable in synovial fluid.
Collapse
Affiliation(s)
| | - Karen A O Martins
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | | | | | | | | | | | | | | | | | | | | | - Lisa H Cazares
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Melissa Gregory
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Michael D Ward
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | | | | | - Lian Dong
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Tara Kenny
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Ernie Brueggemann
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | - Lydia G Downey
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland
| | | | | | | | - Gary L Simon
- The George Washington University, Washington, DC
| | | | | |
Collapse
|
40
|
Moore LJ, Jarungsriapisit J, Nilsen TO, Stefansson S, Taranger GL, Secombes CJ, Morton HC, Patel S. Atlantic salmon adapted to seawater for 9 weeks develop a robust immune response to salmonid alphavirus upon bath challenge. FISH & SHELLFISH IMMUNOLOGY 2018; 74:573-583. [PMID: 29353080 DOI: 10.1016/j.fsi.2017.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/09/2017] [Accepted: 12/11/2017] [Indexed: 06/07/2023]
Abstract
Pancreas disease (PD) caused by salmonid alphavirus (SAV) is the most serious viral disease in Norwegian aquaculture. Study of the immune response to SAV will aid preventative measures including vaccine development. The innate immune response was studied in Atlantic salmon infected by either bath immersion (BI) or by intra-muscular (i.m.) injection (IM) with SAV subtype 3, two and nine weeks after seawater transfer (Phases A and B respectively). Phase A results have been previously published (Moore et al., 2017) and Phase B results are presented here together with a comparison of results achieved in Phase A. There was a rapid accumulation of infected fish in the IM-B (IM Phase B) group and all fish sampled were SAV RNA positive by 7 dpi (days post infection). In contrast, only a few SAV RNA positive (infected) fish were identified at 14, 21 and 28 dpi in the BI-B (BI Phase B) group. Differences in the transcription of several immune genes were apparent when compared between the infected fish in the IM-B and BI-B groups. Transcription of the analysed genes peaked at 7 dpi in the IM-B group and at 14 dpi in the BI-B group. However, this latter finding was difficult to interpret due to the low prevalence of SAV positive fish in this group. Additionally, fish positive for SAV RNA in the BI-B group showed higher transcription of IL-1β, IFNγ and CXCL11_L1, all genes associated with the inflammatory response, compared to the IM-B group. Histopathological changes in the heart were restricted to the IM-B group, while (immune) cell filtration into the pancreas was observed in both groups. Compared to the Phase A fish that were exposed to SAV3 two weeks after seawater transfer, the Phase B fish in the current paper, showed a higher and more sustained innate immune gene transcription in response to the SAV3 infection. In addition, the basal transcription of several innate immune genes in non-infected control fish in Phase B (CT-B) was also significantly different when compared to Phase A control fish (CT-A).
Collapse
Affiliation(s)
- L J Moore
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway
| | - J Jarungsriapisit
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway; Department of Biology, University of Bergen, P.O. Box 7803, 5020 Bergen, Norway
| | - T O Nilsen
- Uni Research Environment, Uni Research, Thormøhlensgt. 49B, 5006 Bergen, Norway
| | - S Stefansson
- Department of Biology, University of Bergen, P.O. Box 7803, 5020 Bergen, Norway
| | - G L Taranger
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway
| | - C J Secombes
- Scottish Fish Immunology Research Centre, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen AB24 2TZ, Scotland, UK
| | - H C Morton
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway
| | - S Patel
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway.
| |
Collapse
|
41
|
Affiliation(s)
- Shefali Khanna Sharma
- Unit of Clinical Immunology and Rheumatology; Department of Internal Medicine; Postgraduate Institute of Medical Education and Research; Chandigarh India
| | - Sanjay Jain
- Unit of Clinical Immunology and Rheumatology; Department of Internal Medicine; Postgraduate Institute of Medical Education and Research; Chandigarh India
| |
Collapse
|
42
|
Abstract
Chikungunya virus (CHIKV) was discovered more than six decades ago, but has remained poorly investigated. However, after a recent outbreak of CHIK fever in both hemispheres and viral adaptation to new species of mosquitoes, it has attracted a lot of attention. The currently available experimental data suggest that molecular mechanisms of CHIKV replication in vertebrate and mosquito cells are similar to those of other New and Old World alphaviruses. However, this virus exhibits a number of unique characteristics that distinguish it from the other, better studied members of the alphavirus genus. This review is an attempt to summarize the data accumulated thus far regarding the molecular mechanisms of alphavirus RNA replication and interaction with host cells. Emphasis was placed on demonstrating the distinct features of CHIKV in utilizing host factors to build replication complexes and modify the intracellular environment for efficient viral replication and inhibition of the innate immune response. The available data suggest that our knowledge about alphavirus replication contains numerous gaps that potentially hamper the development of new therapeutic means against CHIKV and other pathogenic alphaviruses.
Collapse
Affiliation(s)
- I Frolov
- Department of Microbiology, University of Alabama at Birmingham, 1720 2nd Ave South, BBRB373/Box 3, 35294-2170, Birmingham, AL, USA.
| | - E I Frolova
- Department of Microbiology, University of Alabama at Birmingham, 1720 2nd Ave South, BBRB373/Box 3, 35294-2170, Birmingham, AL, USA
| |
Collapse
|
43
|
Ganesan VK, Duan B, Reid SP. Chikungunya Virus: Pathophysiology, Mechanism, and Modeling. Viruses 2017; 9:v9120368. [PMID: 29194359 PMCID: PMC5744143 DOI: 10.3390/v9120368] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022] Open
Abstract
Chikungunya virus (CHIKV), a mosquito-transmitted alphavirus, is recurring in epidemic waves. In the past decade and a half, the disease has resurged in several countries around the globe, with outbreaks becoming increasingly severe. Though CHIKV was first isolated in 1952, there remain significant gaps in knowledge of CHIKV biology, pathogenesis, transmission, and mechanism. Diagnosis is largely simplified and based on symptoms, while treatment is supportive rather than curative. Here we present an overview of the disease, the challenges that lie ahead for future research, and what directions current studies are headed towards, with emphasis on improvement of current animal models and potential use of 3D models.
Collapse
Affiliation(s)
- Vaishnavi K Ganesan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA.
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - St Patrick Reid
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
44
|
Carpentier KS, Morrison TE. Innate immune control of alphavirus infection. Curr Opin Virol 2017; 28:53-60. [PMID: 29175515 DOI: 10.1016/j.coviro.2017.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/06/2017] [Indexed: 12/29/2022]
Abstract
Alphaviruses are important human pathogens that cause diseases ranging from acute and chronic polyarthralgia to encephalitis. Transmitted by mosquito vectors, alphaviruses have high potential for emergence and have initiated several recent epidemics. The innate immune response is critical for controlling the acute phase of alphavirus disease, and the induction of type I interferon (IFN) is essential in this response. In this review, we discuss our current understanding of innate host sensors that initiate antiviral responses following alphavirus infection, and the IFN-induced effector proteins that limit alphavirus replication and dissemination.
Collapse
Affiliation(s)
- Kathryn S Carpentier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
45
|
Interferon Regulatory Factor 1 Protects against Chikungunya Virus-Induced Immunopathology by Restricting Infection in Muscle Cells. J Virol 2017; 91:JVI.01419-17. [PMID: 28835505 DOI: 10.1128/jvi.01419-17] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/20/2017] [Indexed: 01/14/2023] Open
Abstract
The innate immune system protects cells against viral pathogens in part through the autocrine and paracrine actions of alpha/beta interferon (IFN-α/β) (type I), IFN-γ (type II), and IFN-λ (type III). The transcription factor interferon regulatory factor 1 (IRF-1) has a demonstrated role in shaping innate and adaptive antiviral immunity by inducing the expression of IFN-stimulated genes (ISGs) and mediating signals downstream of IFN-γ. Although ectopic expression experiments have suggested an inhibitory function of IRF-1 against infection of alphaviruses in cell culture, its role in vivo remains unknown. Here, we infected Irf1 -/- mice with two distantly related arthritogenic alphaviruses, chikungunya virus (CHIKV) and Ross River virus (RRV), and assessed the early antiviral functions of IRF-1 prior to induction of adaptive B and T cell responses. IRF-1 expression limited CHIKV-induced foot swelling in joint-associated tissues and prevented dissemination of CHIKV and RRV at early time points. Virological and histological analyses revealed greater infection of muscle tissues in Irf1 -/- mice than in wild-type mice. The antiviral actions of IRF-1 appeared to be independent of the induction of type I IFN or the effects of type II and III IFNs but were associated with altered local proinflammatory cytokine and chemokine responses and differential infiltration of myeloid cell subsets. Collectively, our in vivo experiments suggest that IRF-1 restricts CHIKV and RRV infection in stromal cells, especially muscle cells, and that this controls local inflammation and joint-associated swelling.IMPORTANCE Interferon regulatory factor 1 (IRF-1) is a transcription factor that regulates the expression of a broad range of antiviral host defense genes. In this study, using Irf1 -/- mice, we investigated the role of IRF-1 in modulating pathogenesis of two related arthritogenic alphaviruses, chikungunya virus and Ross River virus. Our studies show that IRF-1 controlled alphavirus replication and swelling in joint-associated tissues within days of infection. Detailed histopathological and virological analyses revealed that IRF-1 preferentially restricted CHIKV infection in cells of nonhematopoietic lineage, including muscle cells. The antiviral actions of IRF-1 resulted in decreased local inflammatory responses in joint-associated tissues, which prevented immunopathology.
Collapse
|
46
|
Highly Pathogenic New World Arenavirus Infection Activates the Pattern Recognition Receptor Protein Kinase R without Attenuating Virus Replication in Human Cells. J Virol 2017; 91:JVI.01090-17. [PMID: 28794024 DOI: 10.1128/jvi.01090-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
The arenavirus family consists of several highly pathogenic viruses, including the Old World (OW) arenavirus Lassa fever virus (LASV) and the New World (NW) Junin virus (JUNV) and Machupo virus (MACV). Host response to infection by these pathogenic arenaviruses is distinct in many aspects. JUNV and MACV infections readily induce an interferon (IFN) response in human cells, while LASV infection usually triggers an undetectable or weak IFN response. JUNV induces an IFN response through RIG-I, suggesting that the host non-self RNA sensor readily detects JUNV viral RNAs (vRNAs) during infection and activates IFN response. Double-stranded-RNA (dsRNA)-activated protein kinase R (PKR) is another host non-self RNA sensor classically known for its vRNA recognition activity. Here we report that infection with NW arenaviruses JUNV and MACV, but not OW LASV, activated PKR, concomitant with elevated phosphorylation of the translation initiation factor α subunit of eukaryotic initiation factor 2 (eIF2α). Host protein synthesis was substantially suppressed in MACV- and JUNV-infected cells but was only marginally reduced in LASV-infected cells. Despite the antiviral activity known for PKR against many other viruses, the replication of JUNV and MACV was not impaired but was slightly augmented in wild-type (wt) cells compared to that in PKR-deficient cells, suggesting that PKR or PKR activation did not negatively affect JUNV and MACV infection. Additionally, we found an enhanced IFN response in JUNV- or MACV-infected PKR-deficient cells, which was inversely correlated with virus replication.IMPORTANCE The detection of viral RNA by host non-self RNA sensors, including RIG-I and MDA5, is critical to the initiation of the innate immune response to RNA virus infection. Among pathogenic arenaviruses, the OW LASV usually does not elicit an interferon response. However, the NW arenaviruses JUNV and MACV readily trigger an IFN response in a RIG-I-dependent manner. Here, we demonstrate for the first time that pathogenic NW arenaviruses JUNV and MACV, but not the OW arenavirus LASV, activated the dsRNA-dependent PKR, another host non-self RNA sensor, during infection. Interestingly, the replication of JUNV and MACV was not restricted but was rather slightly augmented in the presence of PKR. Our data provide new evidence for a distinct interplay between host non-self RNA sensors and pathogenic arenaviruses, which also provides insights into the pathogenesis of arenaviruses and may facilitate the design of vaccines and treatments against arenavirus-caused diseases.
Collapse
|
47
|
Amdekar S, Parashar D, Alagarasu K. Chikungunya Virus-Induced Arthritis: Role of Host and Viral Factors in the Pathogenesis. Viral Immunol 2017; 30:691-702. [PMID: 28910194 DOI: 10.1089/vim.2017.0052] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chikungunya virus (CHIKV), a member of Alphavirus genus, is responsible for chikungunya fever (CHIKF), which is characterized by the presence of fever, rash, myalgia, and arthralgia. Reemergence of CHIKV has become a significant public health concern in Asian and African countries and is newly emerging in the Middle East, Pacific, American, and European countries. Cytokines, innate (monocytes, natural killer cells) and adaptive immune response (role of B cells and T cells i.e. CD4+ and CD8+), and/or viral factors contribute to CHIKV-induced arthritis. Vector factors such as vector competence (that includes extrinsic and intrinsic factors) and effect of genome mutations on viral replication and fitness in mosquitoes are responsible for the spread of virus, although they are not directly responsible for CHIKV-induced arthritis. CHIKV-induced arthritis mimics arthritis by involving joints and a common pattern of leukocyte infiltrate, cytokine production, and complement activation. Successful establishment of CHIKV infection and induction of arthritis depends on its ability to manipulate host cellular processes or host factors. CHIKV-induced joint damage is due to host inflammatory response mediated by macrophages, T cells, and antibodies, as well as the possible persistence of the virus in hidden sites. This review provides insight into mechanisms of CHIKV-induced arthritis. Understanding the pathogenesis of CHIKV-induced arthritis will help in developing novel strategies to predict and prevent the disease in virus-infected subjects and combat the disease, thereby decreasing the worldwide burden of the disease.
Collapse
Affiliation(s)
- Sarika Amdekar
- Dengue/Chikungunya Group, ICMR-National Institute of Virology , Pune, India
| | - Deepti Parashar
- Dengue/Chikungunya Group, ICMR-National Institute of Virology , Pune, India
| | | |
Collapse
|
48
|
Fox JM, Diamond MS. Immune-Mediated Protection and Pathogenesis of Chikungunya Virus. THE JOURNAL OF IMMUNOLOGY 2017; 197:4210-4218. [PMID: 27864552 DOI: 10.4049/jimmunol.1601426] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/13/2016] [Indexed: 01/16/2023]
Abstract
Chikungunya virus (CHIKV) is a re-emerging alphavirus that causes debilitating acute and chronic arthritis. Infection by CHIKV induces a robust immune response that is characterized by production of type I IFNs, recruitment of innate and adaptive immune cells, and development of neutralizing Abs. Despite this response, chronic arthritis can develop in some individuals, which may be due to a failure to eliminate viral RNA and Ag and/or persistent immune responses that cause chronic joint inflammation. In this review, based primarily on advances from recent studies in mice, we discuss the innate and adaptive immune factors that control CHIKV dissemination and clearance or contribute to pathogenesis.
Collapse
Affiliation(s)
- Julie M Fox
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110; .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110; and.,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
49
|
Pryke KM, Abraham J, Sali TM, Gall BJ, Archer I, Liu A, Bambina S, Baird J, Gough M, Chakhtoura M, Haddad EK, Kirby IT, Nilsen A, Streblow DN, Hirsch AJ, Smith JL, DeFilippis VR. A Novel Agonist of the TRIF Pathway Induces a Cellular State Refractory to Replication of Zika, Chikungunya, and Dengue Viruses. mBio 2017; 8:e00452-17. [PMID: 28465426 PMCID: PMC5414005 DOI: 10.1128/mbio.00452-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/11/2017] [Indexed: 01/23/2023] Open
Abstract
The ongoing concurrent outbreaks of Zika, Chikungunya, and dengue viruses in Latin America and the Caribbean highlight the need for development of broad-spectrum antiviral treatments. The type I interferon (IFN) system has evolved in vertebrates to generate tissue responses that actively block replication of multiple known and potentially zoonotic viruses. As such, its control and activation through pharmacological agents may represent a novel therapeutic strategy for simultaneously impairing growth of multiple virus types and rendering host populations resistant to virus spread. In light of this strategy's potential, we undertook a screen to identify novel interferon-activating small molecules. Here, we describe 1-(2-fluorophenyl)-2-(5-isopropyl-1,3,4-thiadiazol-2-yl)-1,2-dihydrochromeno[2,3-c]pyrrole-3,9-dione, which we termed AV-C. Treatment of human cells with AV-C activates innate and interferon-associated responses that strongly inhibit replication of Zika, Chikungunya, and dengue viruses. By utilizing genome editing, we investigated the host proteins essential to AV-C-induced cellular states. This showed that the compound requires a TRIF-dependent signaling cascade that culminates in IFN regulatory factor 3 (IRF3)-dependent expression and secretion of type I interferon to elicit antiviral responses. The other canonical IRF3-terminal adaptor proteins STING and IPS-1/MAVS were dispensable for AV-C-induced phenotypes. However, our work revealed an important inhibitory role for IPS-1/MAVS, but not TRIF, in flavivirus replication, implying that TRIF-directed viral evasion may not occur. Additionally, we show that in response to AV-C, primary human peripheral blood mononuclear cells secrete proinflammatory cytokines that are linked with establishment of adaptive immunity to viral pathogens. Ultimately, synthetic innate immune activators such as AV-C may serve multiple therapeutic purposes, including direct antimicrobial responses and facilitation of pathogen-directed adaptive immunity.IMPORTANCE The type I interferon system is part of the innate immune response that has evolved in vertebrates as a first line of broad-spectrum immunological defense against an unknowable diversity of microbial, especially viral, pathogens. Here, we characterize a novel small molecule that artificially activates this response and in so doing generates a cellular state antagonistic to growth of currently emerging viruses: Zika virus, Chikungunya virus, and dengue virus. We also show that this molecule is capable of eliciting cellular responses that are predictive of establishment of adaptive immunity. As such, this agent may represent a powerful and multipronged therapeutic tool to combat emerging and other viral diseases.
Collapse
Affiliation(s)
- Kara M Pryke
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Jinu Abraham
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Tina M Sali
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Bryan J Gall
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Iris Archer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Andrew Liu
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, Oregon, USA
| | - Jason Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, Oregon, USA
| | - Michael Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, Oregon, USA
| | - Marita Chakhtoura
- Division of Infectious Diseases and HIV Medicine, Drexel College of Medicine, Philadelphia, Pennsylvania, USA
| | - Elias K Haddad
- Division of Infectious Diseases and HIV Medicine, Drexel College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ilsa T Kirby
- Veterans Affairs Medical Center, Portland, Oregon, USA
| | - Aaron Nilsen
- Veterans Affairs Medical Center, Portland, Oregon, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Jessica L Smith
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Victor R DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
50
|
Moore LJ, Jarungsriapisit J, Nilsen TO, Stefansson S, Taranger GL, Secombes CJ, Morton HC, Patel S. Immune gene profiles in Atlantic salmon (salmo salar L.) post-smolts infected with SAV3 by bath-challenge show a delayed response and lower levels of gene transcription compared to injected fish. FISH & SHELLFISH IMMUNOLOGY 2017; 62:320-331. [PMID: 28137651 DOI: 10.1016/j.fsi.2017.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/21/2017] [Accepted: 01/23/2017] [Indexed: 06/06/2023]
Abstract
Salmonid alphavirus (SAV) causes pancreatic disease (PD) in salmonids in Northern Europe which results in large economic losses within the aquaculture industry. In order to better understand the underlying immune mechanisms during a SAV3 infection Atlantic salmon post-smolts were infected by either i.m.-injection or bath immersion and their immune responses compared. Analysis of viral loads showed that by 14 dpi i.m.-injected and bath immersion groups had 95.6% and 100% prevalence respectively and that both groups had developed the severe pathology typical of PD. The immune response was evaluated by using RT-qPCR to measure the transcription of innate immune genes involved in the interferon (IFN) response as well as genes associated with inflammation. Our results showed that IFNa transcription was only weakly upregulated, especially in the bath immersion group. Despite this, high levels of the IFN-stimulated genes (ISGs) such as Mx and viperin were observed. The immune response in the i.m.-injected group as measured by immune gene transcription was generally faster, and more pronounced than the response in the bath immersion group, especially at earlier time-points. The response in the bath immersion group started later as expected and appeared to last longer often exceeding the response in the i.m-injected fish at later time-points. High levels of transcription of many genes indicative of an active innate immune response were present in both groups.
Collapse
Affiliation(s)
- L J Moore
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway
| | - J Jarungsriapisit
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway; Department of Biology, University of Bergen, P.O. Box 7803, 5020 Bergen, Norway
| | - T O Nilsen
- Uni Research Environment, Uni Research, Thormøhlensgt, 49B 5006 Bergen, Norway
| | - S Stefansson
- Department of Biology, University of Bergen, P.O. Box 7803, 5020 Bergen, Norway
| | - G L Taranger
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway
| | - C J Secombes
- Scottish Fish Immunology Research Centre, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen AB24 2TZ, Scotland, UK
| | - H C Morton
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway
| | - S Patel
- Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway.
| |
Collapse
|