1
|
Morrey JD, Siddharthan V. Adjusting susceptibilities of C57BL/6 mice to orthoflaviviruses for evaluation of antiviral drugs by altering the levels of interferon alpha/beta receptor function. J Virol Methods 2024; 331:115053. [PMID: 39426414 DOI: 10.1016/j.jviromet.2024.115053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/16/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
The purpose of this study was to optimize the infectivity of four different orthoflaviviruses in mice for evaluating antiviral drugs by using wild-type mice with intact interferon responses, type 1 interferon alpha/beta receptor knockout mice, or by injecting wild type C57BL/6 mice with varying doses of anti-type 1 interferon receptor antibody (MAR1-5A3) to optimize the infectivity and lethality. West Nile virus productively infected wild-type C57BL/6 mice to cause lethality, whereas Usutu virus required a complete absence of type 1 interferon receptor function. Deer tick virus (lineage 2 Powassan virus) and Japanese encephalitis viruses required a dampening of type 1 interferon responses by adjusting the doses of MAR1-5A3 antibody injections. Challenge dose-responsive mortality, weight loss, and viral titers of these two viruses were observed if the type 1 interferon responses were dampened with MAR1-5A3. Conversely, without MAR1-5A3 injections, these disease phenotypes were not viral challenge dose-responsive. From these different interferon-responsive models, the appropriate lethality was identified to determine that 7-deaza-2'-C-methyladenosine has high efficacy for West Nile and Usutu viruses, and low efficacy for deer tick and Japanese encephalitis viruses.
Collapse
Affiliation(s)
- John D Morrey
- Institute for Antiviral Research, Utah State University, 5600 Old Main Hill, Logan, UT 84321-5600, USA.
| | - Venkatraman Siddharthan
- Institute for Antiviral Research, Utah State University, 5600 Old Main Hill, Logan, UT 84321-5600, USA
| |
Collapse
|
2
|
Nogueira CDO, Lopes da Silva MO, de Lima EV, Christoff RR, Gavino-Leopoldino D, Lemos FS, da Silva NE, Da Poian AT, Assunção-Miranda I, Figueiredo CP, Clarke JR. Immunosuppression-induced Zika virus reactivation causes brain inflammation and behavioral deficits in mice. iScience 2024; 27:110178. [PMID: 38993676 PMCID: PMC11237861 DOI: 10.1016/j.isci.2024.110178] [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: 11/27/2023] [Revised: 02/27/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Zika virus (ZIKV) is a neurotropic flavivirus that can persist in several tissues. The late consequences of ZIKV persistence and whether new rounds of active replication can occur, remain unaddressed. Here, we investigated whether neonatally ZIKV-infected mice are susceptible to viral reactivation in adulthood. We found that when ZIKV-infected mice are treated with immunosuppressant drugs, they present increased susceptibility to chemically induced seizures. Levels of subgenomic flavivirus RNAs (sfRNAs) were increased, relative to the amounts of genomic RNAs, in the brains of mice following immunosuppression and were associated with changes in cytokine expression. We investigated the impact of immunosuppression on the testicles and found that ZIKV genomic RNA levels are increased in mice following immunosuppression, which also caused significant testicular damage. These findings suggest that ZIKV can establish new rounds of active replication long after acute stages of disease, so exposed patients should be monitored to ensure complete viral eradication.
Collapse
Affiliation(s)
- Clara de O Nogueira
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | | | - Emanuelle V de Lima
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Raíssa Rilo Christoff
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Daniel Gavino-Leopoldino
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Felipe S Lemos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Nicolas E da Silva
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Iranaia Assunção-Miranda
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Claudia P Figueiredo
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Julia R Clarke
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| |
Collapse
|
3
|
Balint E, Feng E, Giles EC, Ritchie TM, Qian AS, Vahedi F, Montemarano A, Portillo AL, Monteiro JK, Trigatti BL, Ashkar AA. Bystander activated CD8 + T cells mediate neuropathology during viral infection via antigen-independent cytotoxicity. Nat Commun 2024; 15:896. [PMID: 38316762 PMCID: PMC10844499 DOI: 10.1038/s41467-023-44667-0] [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: 02/09/2022] [Accepted: 12/21/2023] [Indexed: 02/07/2024] Open
Abstract
Although many viral infections are linked to the development of neurological disorders, the mechanism governing virus-induced neuropathology remains poorly understood, particularly when the virus is not directly neuropathic. Using a mouse model of Zika virus (ZIKV) infection, we found that the severity of neurological disease did not correlate with brain ZIKV titers, but rather with infiltration of bystander activated NKG2D+CD8+ T cells. Antibody depletion of CD8 or blockade of NKG2D prevented ZIKV-associated paralysis, suggesting that CD8+ T cells induce neurological disease independent of TCR signaling. Furthermore, spleen and brain CD8+ T cells exhibited antigen-independent cytotoxicity that correlated with NKG2D expression. Finally, viral infection and inflammation in the brain was necessary but not sufficient to induce neurological damage. We demonstrate that CD8+ T cells mediate virus-induced neuropathology via antigen-independent, NKG2D-mediated cytotoxicity, which may serve as a therapeutic target for treatment of virus-induced neurological disease.
Collapse
Affiliation(s)
- Elizabeth Balint
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Emily Feng
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Elizabeth C Giles
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Tyrah M Ritchie
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexander S Qian
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Fatemeh Vahedi
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amelia Montemarano
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ana L Portillo
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan K Monteiro
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Bernardo L Trigatti
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
4
|
Yao M, Cheng Z, Li X, Li Y, Ye W, Zhang H, Liu H, Zhang L, Lei Y, Zhang F, Lv X. N6-methyladenosine modification positively regulate Japanese encephalitis virus replication. Virol J 2024; 21:23. [PMID: 38243270 PMCID: PMC10799421 DOI: 10.1186/s12985-023-02275-w] [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: 10/19/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024] Open
Abstract
N6-methyladenosine (m6A) is present in diverse viral RNA and plays important regulatory roles in virus replication and host antiviral innate immunity. However, the role of m6A in regulating JEV replication has not been investigated. Here, we show that the JEV genome contains m6A modification upon infection of mouse neuroblast cells (neuro2a). JEV infection results in a decrease in the expression of m6A writer METTL3 in mouse brain tissue. METTL3 knockdown by siRNA leads to a substantial decrease in JEV replication and the production of progeny viruses at 48 hpi. Mechanically, JEV triggered a considerable increase in the innate immune response of METTL3 knockdown neuro2a cells compared to the control cells. Our study has revealed the distinctive m6A signatures of both the virus and host in neuro2a cells infected with JEV, illustrating the positive role of m6A modification in JEV infection. Our study further enhances understanding of the role of m6A modification in Flaviviridae viruses.
Collapse
Affiliation(s)
- Min Yao
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhirong Cheng
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
- College of Life Science, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Xueyun Li
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
- College of Basic Medicine, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Yuexiang Li
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Wei Ye
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Hui Zhang
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - He Liu
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Liang Zhang
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Yingfeng Lei
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Fanglin Zhang
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China.
| | - Xin Lv
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
5
|
Tatara JM, Rosa RL, Varela APM, Teixeira TF, Sesterheim P, Gris A, Driemeier D, Moraes ANS, Berger M, Peña RD, Roehe PM, Souza DOG, Guimarães JA, Campos AR, Santi L, Beys-da-Silva WO. Differential proteomics of Zika virus (ZIKV) infection reveals molecular changes potentially involved in immune system evasion by a Brazilian strain of ZIKV. Arch Virol 2023; 168:70. [PMID: 36658439 DOI: 10.1007/s00705-022-05629-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/01/2022] [Indexed: 01/21/2023]
Abstract
Zika virus (ZIKV) is an arbovirus that was responsible for multiple outbreaks from 2007 to 2015. It has been linked to cases of microcephaly in Brazil in 2015, among other neurological disorders. Differences among strains might be the reason for different clinical outcomes of infection. To evaluate this hypothesis, we performed a comparative proteomic analysis of Vero cells infected with the African strain MR766 (ZIKVAFR) and the Brazilian strain 17 SM (ZIKVBR). A total of 550 proteins were identified as differentially expressed in ZIKVAFR- or ZIKVBR-infected cells compared to the control. The main findings included upregulation of immune system pathways (neutrophil degranulation and adaptive/innate immune system) and potential activation of immune-system-related pathways by ZIKVAFR (mTOR, JAK-STAT, NF-κB, and others) compared with the ZIKVBR/control. In addition, phagocytosis by macrophages and engulfment of leukocytes were activated in ZIKVAFR infection. An in vivo analysis using an immunocompetent C57BL/6N mouse model identified interstitial pneumonia with neutrophil infiltration in the lungs only in mice infected with ZIKVBR at 48 hours postinfection, with a significant amount of virus detected. Likewise, only animals infected with ZIKVBR had viral material in the cytoplasm of lung macrophages. These results suggest that activation of the immune system by ZIKVAFR infection may lead to faster viral clearance by immune cells.
Collapse
Affiliation(s)
- Juliana M Tatara
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil
| | - Rafael L Rosa
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil
| | - Ana Paula M Varela
- Department of Microbiology, Immunology and Parasitology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Tais F Teixeira
- Center for Experimental Cardiology, Institute of Cardiology, Porto Alegre, Brazil
| | - Patrícia Sesterheim
- Center for Experimental Cardiology, Institute of Cardiology, Porto Alegre, Brazil
| | - Anderson Gris
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - David Driemeier
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Amanda N S Moraes
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil
| | - Markus Berger
- Experimental Research Center, Clinical Hospital of Porto Alegre, Porto Alegre, Brazil
| | - Ramon D Peña
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Paulo M Roehe
- Department of Microbiology, Immunology and Parasitology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Diogo O G Souza
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jorge A Guimarães
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil.,Center for Experimental Cardiology, Institute of Cardiology, Porto Alegre, Brazil
| | | | - Lucélia Santi
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil.,Experimental Research Center, Clinical Hospital of Porto Alegre, Porto Alegre, Brazil.,Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Walter O Beys-da-Silva
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil. .,Experimental Research Center, Clinical Hospital of Porto Alegre, Porto Alegre, Brazil. .,Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
6
|
Wang X, Wang G, Yang H, Fu S, He Y, Li F, Wang H, Wang Z. A mouse model of peripheral nerve injury induced by Japanese encephalitis virus. PLoS Negl Trop Dis 2022; 16:e0010961. [PMID: 36441775 PMCID: PMC9731479 DOI: 10.1371/journal.pntd.0010961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 12/08/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
Japanese encephalitis virus (JEV) is the most important cause of acute encephalitis in Eastern/Southern Asia. Infection with this virus also induces peripheral nerve injury. However, the disease pathogenesis is still not completely understood. Reliable animal models are needed to investigate the molecular pathogenesis of this condition. We studied the effect of Japanese encephalitis virus infection in C57BL/6 mice after a subcutaneous challenge. Limb paralysis was determined in mice using behavioral tests, including a viral paralysis scale and the hanging wire test, as well as by changes in body weight. Nerve conduction velocity and electromyography testing indicated the presence of demyelinating neuropathy of the sciatic nerve. Pathological changes in neural tissues were examined by immunofluorescence and transmission electron microscopy, which confirmed that the predominant pathologic change was demyelination. Although Western blots confirmed the presence of the virus in neural tissue, additional studies demonstrated that an immune-induced inflammatory response resulted in severe never injury. Immunofluorescence confirmed the presence of Japanese encephalitis virus in the brains of infected mice, and an inflammatory reaction was observed with hematoxylin-eosin staining as well. However, these observations were inconsistent at the time of paralysis onset. In summary, our results demonstrated that Japanese encephalitis virus infection could cause inflammatory demyelination of the peripheral nervous system in C57BL/6 mice.
Collapse
Affiliation(s)
- Xiaoli Wang
- The NO.1 People’s Hospital of Shizuishan, Shizuishan, China
- Ningxia Medical University, Yinchuan, China
| | | | - Huan Yang
- Ningxia Medical University, Yinchuan, China
- General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shihong Fu
- Department of Arbovirus, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ying He
- Department of Arbovirus, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Fan Li
- Department of Arbovirus, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Huanyu Wang
- Department of Arbovirus, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing, China
- * E-mail: (HYW); (ZHW)
| | - Zhenhai Wang
- Neurology Center, General Hospital of Ningxia Medical University, Yinchuan, China
- Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, Yinchuan, China
- * E-mail: (HYW); (ZHW)
| |
Collapse
|
7
|
Sherer ML, Lemanski EA, Patel RT, Wheeler SR, Parcells MS, Schwarz JM. A Rat Model of Prenatal Zika Virus Infection and Associated Long-Term Outcomes. Viruses 2021; 13:v13112298. [PMID: 34835104 PMCID: PMC8624604 DOI: 10.3390/v13112298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that became widely recognized due to the epidemic in Brazil in 2015. Since then, there has been nearly a 20-fold increase in the incidence of microcephaly and birth defects seen among women giving birth in Brazil, leading the Centers for Disease Control and Prevention (CDC) to officially declare a causal link between prenatal ZIKV infection and the serious brain abnormalities seen in affected infants. Here, we used a unique rat model of prenatal ZIKV infection to study three possible long-term outcomes of congenital ZIKV infection: (1) behavior, (2) cell proliferation, survival, and differentiation in the brain, and (3) immune responses later in life. Adult offspring that were prenatally infected with ZIKV exhibited motor deficits in a sex-specific manner, and failed to mount a normal interferon response to a viral immune challenge later in life. Despite undetectable levels of ZIKV in the brain and serum in these offspring at P2, P24, or P60, these results suggest that prenatal exposure to ZIKV results in lasting consequences that could significantly impact the health of the offspring. To help individuals already exposed to ZIKV, as well as be prepared for future outbreaks, we need to understand the full spectrum of neurological and immunological consequences that could arise following prenatal ZIKV infection.
Collapse
Affiliation(s)
- Morgan L. Sherer
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA; (R.T.P.); (S.R.W.); (J.M.S.)
- Correspondence: (M.L.S.); (E.A.L.)
| | - Elise A. Lemanski
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA; (R.T.P.); (S.R.W.); (J.M.S.)
- Correspondence: (M.L.S.); (E.A.L.)
| | - Rita T. Patel
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA; (R.T.P.); (S.R.W.); (J.M.S.)
| | - Shannon R. Wheeler
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA; (R.T.P.); (S.R.W.); (J.M.S.)
| | - Mark S. Parcells
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Jaclyn M. Schwarz
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA; (R.T.P.); (S.R.W.); (J.M.S.)
| |
Collapse
|
8
|
Balint E, Montemarano A, Feng E, Ashkar AA. From Mosquito Bites to Sexual Transmission: Evaluating Mouse Models of Zika Virus Infection. Viruses 2021; 13:v13112244. [PMID: 34835050 PMCID: PMC8625727 DOI: 10.3390/v13112244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/02/2021] [Indexed: 01/04/2023] Open
Abstract
Following the recent outbreak of Zika virus (ZIKV) infections in Latin America, ZIKV has emerged as a global health threat due to its ability to induce neurological disease in both adults and the developing fetus. ZIKV is largely mosquito-borne and is now endemic in many parts of Africa, Asia, and South America. However, several reports have demonstrated persistent ZIKV infection of the male reproductive tract and evidence of male-to-female sexual transmission of ZIKV. Sexual transmission may broaden the reach of ZIKV infections beyond its current geographical limits, presenting a significant threat worldwide. Several mouse models of ZIKV infection have been developed to investigate ZIKV pathogenesis and develop effective vaccines and therapeutics. However, the majority of these models focus on mosquito-borne infection, while few have considered the impact of sexual transmission on immunity and pathogenesis. This review will examine the advantages and disadvantages of current models of mosquito-borne and sexually transmitted ZIKV and provide recommendations for the effective use of ZIKV mouse models.
Collapse
|
9
|
Siddharthan V, Wang H, de Oliveira AL, Dai X, Morrey JD. Memantine treatment reduces the incidence of flaccid paralysis in a zika virus mouse model of temporary paralysis with similarities to Guillain-Barré syndrome. Antivir Chem Chemother 2021; 28:2040206620950143. [PMID: 34161179 PMCID: PMC7432970 DOI: 10.1177/2040206620950143] [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] [Indexed: 11/15/2022] Open
Abstract
Clinical evidence suggests that Zika virus contributes to Guillain-Barré syndrome that
causes temporary paralysis. We utilized a recently described Zika virus mouse model of
temporary flaccid paralysis to address the hypothesis that treatment with an
N-methyl-D-aspartate receptor antagonist, memantine, can reduce the
incidence of paralysis. Aged interferon alpha/beta-receptor knockout mice were used
because of their sublethal susceptibility to Zika virus infection. Fifteen to twenty-five
percent of mice infected with a Puerto Rico strain of Zika virus develop acute flaccid
paralysis beginning at days 8–9 and peaked at days 10–12. Mice recover from paralysis
within a week of onset. In two independent studies, twice daily oral administration of
memantine at 60 mg/kg/day on days 4 through 9 after viral challenge significantly reduced
the incidence of paralysis. No efficacy was observed with treatments from days 9 through
12. Memantine treatment in cell culture or mice did not affect viral titers. These data
indicate that early treatment of memantine before onset of paralysis is efficacious, but
treatments beyond the onset of paralysis were not efficacious. The effect of this
N-methyl-D-aspartate receptor antagonist on the incidence of Zika virus-induced paralysis
may provide guidance for investigations on the mechanism of paralysis.
Collapse
Affiliation(s)
- Venkatraman Siddharthan
- Department of Animal, Dairy, and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| | - Hong Wang
- Department of Animal, Dairy, and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| | | | - Xin Dai
- Utah Agricultural Experiment Station, College of Agriculture, Utah State University, Logan, UT, USA
| | - John D Morrey
- Department of Animal, Dairy, and Veterinary Sciences, Institute for Antiviral Research, Utah State University, Logan, UT, USA
| |
Collapse
|
10
|
Complex Roles of Neutrophils during Arboviral Infections. Cells 2021; 10:cells10061324. [PMID: 34073501 PMCID: PMC8227388 DOI: 10.3390/cells10061324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Arboviruses are known to cause large-scale epidemics in many parts of the world. These arthropod-borne viruses are a large group consisting of viruses from a wide range of families. The ability of their vector to enhance viral pathogenesis and transmission makes the development of treatments against these viruses challenging. Neutrophils are generally the first leukocytes to be recruited to a site of infection, playing a major role in regulating inflammation and, as a result, viral replication and dissemination. However, the underlying mechanisms through which neutrophils control the progression of inflammation and disease remain to be fully understood. In this review, we highlight the major findings from recent years regarding the role of neutrophils during arboviral infections. We discuss the complex nature of neutrophils in mediating not only protection, but also augmenting disease pathology. Better understanding of neutrophil pathways involved in effective protection against arboviral infections can help identify potential targets for therapeutics.
Collapse
|
11
|
Stegelmeier AA, Darzianiazizi M, Hanada K, Sharif S, Wootton SK, Bridle BW, Karimi K. Type I Interferon-Mediated Regulation of Antiviral Capabilities of Neutrophils. Int J Mol Sci 2021; 22:4726. [PMID: 33946935 PMCID: PMC8125486 DOI: 10.3390/ijms22094726] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Interferons (IFNs) are induced by viruses and are the main regulators of the host antiviral response. They balance tissue tolerance and immune resistance against viral challenges. Like all cells in the human body, neutrophils possess the receptors for IFNs and contribute to antiviral host defense. To combat viruses, neutrophils utilize various mechanisms, such as viral sensing, neutrophil extracellular trap formation, and antigen presentation. These mechanisms have also been linked to tissue damage during viral infection and inflammation. In this review, we presented evidence that a complex cross-regulatory talk between IFNs and neutrophils initiates appropriate antiviral immune responses and regulates them to minimize tissue damage. We also explored recent exciting research elucidating the interactions between IFNs, neutrophils, and severe acute respiratory syndrome-coronavirus-2, as an example of neutrophil and IFN cross-regulatory talk. Dissecting the IFN-neutrophil paradigm is needed for well-balanced antiviral therapeutics and development of novel treatments against many major epidemic or pandemic viral infections, including the ongoing pandemic of the coronavirus disease that emerged in 2019.
Collapse
Affiliation(s)
| | | | | | | | | | - Byram W. Bridle
- Correspondence: (B.W.B.); (K.K.); Tel.: +1-(519)-824-4120 (ext. 54657) (B.W.B.); +1-(519)-824-4120 (ext. 54668) (K.K.)
| | - Khalil Karimi
- Correspondence: (B.W.B.); (K.K.); Tel.: +1-(519)-824-4120 (ext. 54657) (B.W.B.); +1-(519)-824-4120 (ext. 54668) (K.K.)
| |
Collapse
|
12
|
de Mendonça SF, Rocha MN, Ferreira FV, Leite THJF, Amadou SCG, Sucupira PHF, Marques JT, Ferreira AGA, Moreira LA. Evaluation of Aedes aegypti, Aedes albopictus, and Culex quinquefasciatus Mosquitoes Competence to Oropouche virus Infection. Viruses 2021; 13:v13050755. [PMID: 33923055 PMCID: PMC8145018 DOI: 10.3390/v13050755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
The emergence of new human viral pathogens and re-emergence of several diseases are of particular concern in the last decades. Oropouche orthobunyavirus (OROV) is an arbovirus endemic to South and Central America tropical regions, responsible to several epidemic events in the last decades. There is little information regarding the ability of OROV to be transmitted by urban/peri-urban mosquitoes, which has limited the predictability of the emergence of permanent urban transmission cycles. Here, we evaluated the ability of OROV to infect, replicate, and be transmitted by three anthropophilic and urban species of mosquitoes, Aedes aegypti, Aedes albopictus, and Culex quinquefasciatus. We show that OROV is able to infect and efficiently replicate when systemically injected in all three species tested, but not when orally ingested. Moreover, we find that, once OROV replication has occurred in the mosquito body, all three species were able to transmit the virus to immunocompromised mice during blood feeding. These data provide evidence that OROV is restricted by the midgut barrier of three major urban mosquito species, but, if this restriction is overcome, could be efficiently transmitted to vertebrate hosts. This poses a great risk for the emergence of permanent urban cycles and geographic expansion of OROV to other continents.
Collapse
Affiliation(s)
- Silvana F. de Mendonça
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
| | - Marcele N. Rocha
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Flávia V. Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Thiago H. J. F Leite
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Siad C. G. Amadou
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Pedro H. F. Sucupira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
| | - João T. Marques
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
- Faculté des Sciences de laVie, Université de Strasbourg, CNRS UPR9022, Inserm U1257, 67084 Strasbourg, France
| | - Alvaro G. A. Ferreira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
| | - Luciano A. Moreira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
- Correspondence:
| |
Collapse
|
13
|
Diagnostic differentiation of Zika and dengue virus exposure by analyzing T cell receptor sequences from peripheral blood of infected HLA-A2 transgenic mice. PLoS Negl Trop Dis 2020; 14:e0008896. [PMID: 33270635 PMCID: PMC7738164 DOI: 10.1371/journal.pntd.0008896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/15/2020] [Accepted: 10/15/2020] [Indexed: 11/19/2022] Open
Abstract
Zika virus (ZIKV) is a significant global health threat due to its potential for rapid emergence and association with severe congenital malformations during infection in pregnancy. Despite the urgent need, accurate diagnosis of ZIKV infection is still a major hurdle that must be overcome. Contributing to the inaccuracy of most serologically-based diagnostic assays for ZIKV, is the substantial geographic and antigenic overlap with other flaviviruses, including the four serotypes of dengue virus (DENV). Within this study, we have utilized a novel T cell receptor (TCR) sequencing platform to distinguish between ZIKV and DENV infections. Using high-throughput TCR sequencing of lymphocytes isolated from DENV and ZIKV infected mice, we were able to develop an algorithm which could identify virus-associated TCR sequences uniquely associated with either a prior ZIKV or DENV infection in mice. Using this algorithm, we were then able to separate mice that had been exposed to ZIKV or DENV infection with 97% accuracy. Overall this study serves as a proof-of-principle that T cell receptor sequencing can be used as a diagnostic tool capable of distinguishing between closely related viruses. Our results demonstrate the potential for this innovative platform to be used to accurately diagnose Zika virus infection and potentially the next emerging pathogen(s). Diagnostic differentiation between dengue virus and Zika virus infections is a challenge due to serological cross-reactivity. In this study, we used a novel T cell receptor sequencing platform to identify T cell receptor sequences significantly associated with either dengue or Zika virus infection in HLA-A2 transgenic mice. These libraries were used to computationally train diagnostic classifiers which were capable of distinguishing between dengue and Zika virus in independent cohorts of infected mice.
Collapse
|
14
|
Castanospermine reduces Zika virus infection-associated seizure by inhibiting both the viral load and inflammation in mouse models. Antiviral Res 2020; 183:104935. [PMID: 32949636 PMCID: PMC7492813 DOI: 10.1016/j.antiviral.2020.104935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 11/30/2022]
Abstract
Zika virus (ZIKV) outbreaks have been reported worldwide, including a recent occurrence in Brazil where it spread rapidly, and an association with increased cases of microcephaly was observed in addition to neurological issues such as GBS that were reported during previous outbreaks. Following infection of neuronal tissues, ZIKV can cause inflammation, which may lead to neuronal abnormalities, including seizures and paralysis. Therefore, a drug containing both anti-viral and immunosuppressive properties would be of great importance in combating ZIKV related neurological abnormalities. Castanospermine (CST) is potentially a right candidate drug as it reduced viral load and brain inflammation with the resulting appearance of delayed neuronal disorders, including seizures and paralysis in an Ifnar1−/− mouse. Anti-ZIKV activity of castanospermine (CST) In vivo and in vitro. CST reduces ZIKV induced inflammation of brain. CST delays the ZIKV induced seizure and improves neuronal disorders such as motor function. CST gives marginal improvement in survivability in Ifnar1−/− mice.
Collapse
|
15
|
The involvement of annexin A1 in human placental response to maternal Zika virus infection. Antiviral Res 2020; 179:104809. [PMID: 32360947 DOI: 10.1016/j.antiviral.2020.104809] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/02/2020] [Accepted: 04/25/2020] [Indexed: 12/22/2022]
Abstract
The association of Zika virus infection (ZIKV) with congenital malformation and neurological sequelae brought a significant global concern. Recent studies have shown that maternal viral infection leads to inflammation in the placental tissue. In this context, the antiinflammatory protein annexin 1 (ANXA1) has a major determination of the resolution of inflammation and it has been positively associated with antiparasitic activity in infected placental explants. Although these effects have been explored to some degree, ANXA1 expression and potential properties have not yet been fully elucidated in placentas infected with ZIKV. This study was conducted to evaluate the histopathology, inflammatory process and elucidate if ANXA1 were differently expressed in placentas of ZIKV-infected mothers. Three classification groups were used in this study: Neg/Neg (mother and placenta negative for the virus), Pos/Neg (infected mother, but no virus detected in placenta) and Pos/Pos (mother and placenta infected with ZIKV). ANXA1 was expressed in syncytiotrophoblast cells of all studied groups, and its expression was decreased in Pos/Neg group, which displayed also an increase of the inflammatory response, as evinced from the recruitment of inflammatory cells, increased levels of placenta cytokines, and evidence of impaired tissue repair. The presence of ZIKV in placentas of Pos/Pos group shows structural alterations, including detachment and disorganization of the trophoblastic epithelium. In summary, our results suggest that maternal infection with ZIKV, even without direct tissue infection, leads to a placental inflammatory response probably related to the modulation of ANXA1. After placental infection, structural changes - including inflammatory cells influx - are observed leading to placental dysfunction and reduced fetal weight. Our study sheds additional light on the outcomes of ZIKV infection in trophoblast and reveals a potential involvement of ANXA1 in the placental biology.
Collapse
|
16
|
Morrey JD, Oliveira ALR, Wang H, Zukor K, de Castro MV, Siddharthan V. Zika virus infection causes temporary paralysis in adult mice with motor neuron synaptic retraction and evidence for proximal peripheral neuropathy. Sci Rep 2019; 9:19531. [PMID: 31862897 PMCID: PMC6925114 DOI: 10.1038/s41598-019-55717-3] [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: 07/31/2019] [Accepted: 12/01/2019] [Indexed: 12/27/2022] Open
Abstract
Clinical evidence is mounting that Zika virus can contribute to Guillain-Barré syndrome which causes temporary paralysis, yet the mechanism is unknown. We investigated the mechanism of temporary acute flaccid paralysis caused by Zika virus infection in aged interferon αβ-receptor knockout mice used for their susceptibility to infection. Twenty-five to thirty-five percent of mice infected subcutaneously with Zika virus developed motor deficits including acute flaccid paralysis that peaked 8-10 days after viral challenge. These mice recovered within a week. Despite Zika virus infection in the spinal cord, motor neurons were not destroyed. We examined ultrastructures of motor neurons and synapses by transmission electron microscopy. The percent coverage of motor neurons by boutons was reduced by 20%; more specifically, flattened-vesicle boutons were reduced by 46%, and were normalized in recovering mice. Using electromyographic procedures employed in people to help diagnose Guillain-Barré syndrome, we determined that nerve conduction velocities between the sciatic notch and the gastrocnemius muscle were unchanged in paralyzed mice. However, F-wave latencies were increased in paralyzed mice, which suggests that neuropathy may exist between the sciatic notch to the nerve rootlets. Reversible synaptic retraction may be a previously unrecognized cofactor along with peripheral neuropathy for the development of Guillain-Barré syndrome during Zika virus outbreaks.
Collapse
Affiliation(s)
- John D Morrey
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, United States of America.
| | | | - Hong Wang
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, United States of America
| | - Katherine Zukor
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, United States of America
| | | | - Venkatraman Siddharthan
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, United States of America
| |
Collapse
|
17
|
Bellmann J, Monette A, Tripathy V, Sójka A, Abo-Rady M, Janosh A, Bhatnagar R, Bickle M, Mouland AJ, Sterneckert J. Viral Infections Exacerbate FUS-ALS Phenotypes in iPSC-Derived Spinal Neurons in a Virus Species-Specific Manner. Front Cell Neurosci 2019; 13:480. [PMID: 31695598 PMCID: PMC6817715 DOI: 10.3389/fncel.2019.00480] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) arises from an interplay of genetic mutations and environmental factors. ssRNA viruses are possible ALS risk factors, but testing their interaction with mutations such as in FUS, which encodes an RNA-binding protein, has been difficult due to the lack of a human disease model. Here, we use isogenic induced pluripotent stem cell (iPSC)-derived spinal neurons (SNs) to investigate the interaction between ssRNA viruses and mutant FUS. We find that rabies virus (RABV) spreads ALS phenotypes, including the formation of stress granules (SGs) with aberrant composition due to increased levels of FUS protein, as well as neurodegeneration and reduced restriction activity by FUS mutations. Consistent with this, iPSC-derived SNs harboring mutant FUS are more sensitive to human immunodeficiency virus (HIV-1) and Zika viruses (ZIKV). We demonstrate that RABV and HIV-1 exacerbate cytoplasmic mislocalization of FUS. Our results demonstrate that viral infections worsen ALS pathology in SNs with genetic risk factors, suggesting a novel role for viruses in modulating patient phenotypes.
Collapse
Affiliation(s)
- Jessica Bellmann
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anne Monette
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Vadreenath Tripathy
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anna Sójka
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Masin Abo-Rady
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Antje Janosh
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrew J Mouland
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Jared Sterneckert
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
18
|
Sariyer IK, Gordon J, Burdo TH, Wollebo HS, Gianti E, Donadoni M, Bellizzi A, Cicalese S, Loomis R, Robinson JA, Carnevale V, Steiner J, Ozdener MH, Miller AD, Amini S, Klein ML, Khalili K. Suppression of Zika Virus Infection in the Brain by the Antiretroviral Drug Rilpivirine. Mol Ther 2019; 27:2067-2079. [PMID: 31653397 DOI: 10.1016/j.ymthe.2019.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 01/02/2023] Open
Abstract
Zika virus (ZIKV) infection is associated with microcephaly in neonates and Guillain-Barré syndrome in adults. ZIKV produces a class of nonstructural (NS) regulatory proteins that play a critical role in viral transcription and replication, including NS5, which possesses RNA-dependent RNA polymerase (RdRp) activity. Here we demonstrate that rilpivirine (RPV), a non-nucleoside reverse transcriptase inhibitor (NNRTI) used in the treatment of HIV-1 infection, inhibits the enzymatic activity of NS5 and suppresses ZIKV infection and replication in primary human astrocytes. Similarly, other members of the NNRTI family, including etravirine and efavirenz, showed inhibitory effects on viral infection of brain cells. Site-directed mutagenesis identified 14 amino acid residues within the NS5 RdRp domain (AA265-903), which are important for the RPV interaction and the inhibition of NS5 polymerase activity. Administration of RPV to ZIKV-infected interferon-alpha/beta receptor (IFN-A/R) knockout mice improved the clinical outcome and prevented ZIKV-induced mortality. Histopathological examination of the brains from infected animals revealed that RPV reduced ZIKV RNA levels in the hippocampus, frontal cortex, thalamus, and cerebellum. Repurposing of NNRTIs, such as RPV, for the inhibition of ZIKV replication offers a possible therapeutic strategy for the prevention and treatment of ZIKV-associated disease.
Collapse
Affiliation(s)
- Ilker Kudret Sariyer
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Jennifer Gordon
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Tricia H Burdo
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Hassen S Wollebo
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Eleonora Gianti
- Department of Chemistry, Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1901 N. 12(th) Street, Philadelphia, PA 19122, USA
| | - Martina Donadoni
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Anna Bellizzi
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Stephanie Cicalese
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Regina Loomis
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Jake A Robinson
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Vincenzo Carnevale
- Department of Chemistry, Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1901 N. 12(th) Street, Philadelphia, PA 19122, USA
| | - Joseph Steiner
- Translational Neuroscience Center, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Mehmet H Ozdener
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Andrew D Miller
- College of Veterinary Medicine, Cornell University, T5-006A Veterinary Research Tower, Ithaca, NY 14853, USA
| | - Shohreh Amini
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Michael L Klein
- Department of Chemistry, Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1901 N. 12(th) Street, Philadelphia, PA 19122, USA
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
19
|
Alfano C, Gladwyn-Ng I, Couderc T, Lecuit M, Nguyen L. The Unfolded Protein Response: A Key Player in Zika Virus-Associated Congenital Microcephaly. Front Cell Neurosci 2019; 13:94. [PMID: 30971894 PMCID: PMC6445045 DOI: 10.3389/fncel.2019.00094] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/26/2019] [Indexed: 01/08/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that belongs to the Flaviviridae family, together with dengue, yellow fever, and West Nile viruses. In the wake of its emergence in the French Polynesia and in the Americas, ZIKV has been shown to cause congenital microcephaly. It is the first arbovirus which has been proven to be teratogenic and sexually transmissible. Confronted with this major public health challenge, the scientific and medical communities teamed up to precisely characterize the clinical features of congenital ZIKV syndrome and its underlying pathophysiological mechanisms. This review focuses on the critical impact of the unfolded protein response (UPR) on ZIKV-associated congenital microcephaly. ZIKV infection of cortical neuron progenitors leads to high endoplasmic reticulum (ER) stress. This results in both the stalling of indirect neurogenesis, and UPR-dependent neuronal apoptotic death, and leads to cortical microcephaly. In line with these results, the administration of molecules inhibiting UPR prevents ZIKV-induced cortical microcephaly. The discovery of the link between ZIKV infection and UPR activation has a broader relevance, since this pathway plays a crucial role in many distinct cellular processes and its induction by ZIKV may account for several reported ZIKV-associated defects.
Collapse
Affiliation(s)
- Christian Alfano
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège, Belgium
| | - Ivan Gladwyn-Ng
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège, Belgium
| | - Thérèse Couderc
- Institut Pasteur, Biology of Infection Unit, Paris, France.,INSERM U1117, Biologie des Infections, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France.,INSERM U1117, Biologie des Infections, Paris, France.,Paris Descartes University, Division of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades Hospital, Institut Imagine, Sorbonne Paris Cité, Paris, France
| | - Laurent Nguyen
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège, Belgium
| |
Collapse
|
20
|
Bartolini L, Libbey JE, Ravizza T, Fujinami RS, Jacobson S, Gaillard WD. Viral Triggers and Inflammatory Mechanisms in Pediatric Epilepsy. Mol Neurobiol 2018; 56:1897-1907. [PMID: 29978423 DOI: 10.1007/s12035-018-1215-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/27/2018] [Indexed: 12/21/2022]
Abstract
Experimental and clinical findings suggest a crucial role for inflammation in the onset of pediatric seizures; this mechanism is not targeted by conventional antiepileptic drugs and may contribute to refractory epilepsy. Several triggers, including infection with neurotropic viruses such as human herpesvirus 6 (HHV-6), other herpesviruses, and picornaviruses, appear to induce activation of the innate and adaptive immune systems, which results in several neuroinflammatory responses, leading to enhanced neuronal excitability, and ultimately contributing to epileptogenesis. This review discusses the proposed mechanisms by which infection with herpesviruses, and particularly with HHV-6, and ensuing inflammation may lead to seizure generation, and later development of epilepsy. We also examine the evidence that links herpesvirus and picornavirus infections with acute seizures and chronic forms of epilepsy. Understanding the mechanisms by which specific viruses may trigger a cascade of alterations in the CNS ultimately leading to epilepsy appears critical for the development of therapeutic agents that may target the virus or inflammatory mechanisms early and prevent progression of epileptogenesis.
Collapse
Affiliation(s)
- Luca Bartolini
- Clinical Epilepsy Section, National Institute of Neurological Disorders and Stroke, NIH, Building 10, room 7-5680, 10 Center Drive, Bethesda, MD, 20814, USA. .,Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, NIH, 10 Center Drive, Bethesda, MD, 20892, USA. .,Center for Neuroscience, Children's National Medical Center, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA.
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Teresa Ravizza
- Neuroscience Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Steven Jacobson
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - William D Gaillard
- Center for Neuroscience, Children's National Medical Center, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA
| |
Collapse
|