1
|
Meena K, Babu R, Pancholi B, Garabadu D. Exploring therapeutic potential of claudin in Flavivirus infection: A review on current advances and future perspectives. Int J Biol Macromol 2025; 309:142936. [PMID: 40203926 DOI: 10.1016/j.ijbiomac.2025.142936] [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: 01/20/2025] [Revised: 03/25/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Flavivirus such as Dengue, Zika, West Nile, Japanese encephalitis, and yellow fever virus, composed of single-stranded positive-sense RNA, predominantly contaminated through arthropods. Flavivirus infection characterises from asymptomatic signs to severe hemorrhagic fever and encephalitis. The host's immune system detects these viruses and provides a defence mechanism to sustain their life and growth. However, flaviviruses through different mechanisms compromise the host's immune defence. The current pharmacotherapeutic strategies against Flavivirus infection target different stages of the Flavivirus life cycle and its proteins. On the contrary, the host's immune defence mechanism is equally important to restrict their growth. It has been suggested that flaviviruses compromise claudins to sustain their life and growth inside the mammalian cells. This review primarily focuses on the effect of Flavivirus on claudins (CLDNs), transmembrane proteins that form tight junctions in mammalian cells. CLDNs are crucial in viral entry and pathogenesis by regulating paracellular permeability, particularly in tissues and the blood-brain barrier. Recent studies indicate that the Dengue and Zika viruses can potentially be treated by targeting specific CLDNs-specifically CLDN 1, CLDN 5, and CLDN 7 to inhibit viral entry and fusion. Additionally, it highlights the current challenges and future prospects in developing claudin-based antiviral agents against Flavivirus infections.
Collapse
Affiliation(s)
- Kiran Meena
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
| | - Raja Babu
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
| | | | - Debapriya Garabadu
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India.
| |
Collapse
|
2
|
Nakayama E, Tang B, Stewart R, Cox AL, Yan K, Bishop CR, Dumenil T, Nguyen W, Slonchak A, Sng J, Khromykh AA, Lutzky VP, Rawle DJ, Suhrbier A. Evolution of Zika virus in Rag1-deficient mice selects for unique envelope glycosylation motif mutants that show enhanced replication fitness. Virus Evol 2025; 11:veaf021. [PMID: 40291117 PMCID: PMC12024116 DOI: 10.1093/ve/veaf021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
N-linked glycosylation of flavivirus envelope proteins is widely viewed as being required for optimal folding, processing and/or transit of envelope proteins, and the assembling virons, through the endoplasmic reticulum (ER) and the Golgi. Zika virus (ZIKV) has a single N-linked envelope glycan located adjacent to the fusion loop. Herein we show that independent serial passage of ZIKVNatal in Rag1 -/- mice for 223 or 386 days generated two unique envelope glycan-deficient mutants, ZIKV-V153D and ZIKV-N154D, respectively. Surprisingly, these mutants grew to titres ∼1 to 2.6 logs higher than the glycosylated parental ZIKVNatal in Vero E6 cells and human brain organoids. RNA-Seq of infected organoids suggested that this increased replication fitness was associated with upregulation of the unfolded protein response (UPR). Cell death, cellular viral RNA, and viral protein levels were not significantly affected, arguing that these glycan mutants enjoyed faster ER/Golgi folding, processing, assembly, transit, and virion egress, assisted by an upregulated UPR. Thus, ZIKV envelope N-linked glycosylation is not essential for promoting envelope folding, assembly, and transit through the ER/Golgi, since aspartic acid (D) substitutions in the glycosylation motif can achieve this with significantly greater efficiency. Instead, the evolution of glycan mutants in Rag1 -/- mice indicates that such envelope glycosylation can have a fitness cost in an environment devoid of virus-specific antibody responses. The V153D and N154D mutations, generated by natural selection in Rag1 -/- mice, have to date not been employed in orthoflavivirus envelope glycosylation studies. Instead, genetic engineering has been used to generate mutant viruses that, for instance, contain a N154A substitution. The latter may impart confounding unfavourable properties, such as envelope protein insolubility, that have a detrimental impact on virus replication. The V153D and N154D substitutions may avoid imparting unfavourable properties by preserving the surface negative charge provided by the glycan moiety in the parental ZIKVNatal envelope protein. In Ifnar1 -/- mice ZIKV-V153D and -N154D showed faster viremia onsets, but reduced viremic periods, than the parental ZIKVNatal, consistent with an established contention that such glycans have evolved to delay neutralizing antibody activity.
Collapse
Affiliation(s)
- Eri Nakayama
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
- Department of Virology I, National Institute of Infectious Diseases, Shinjuku City, Tokyo 162-0052 Japan
| | - Bing Tang
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Romal Stewart
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Abigail L Cox
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Kexin Yan
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Cameron R Bishop
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Troy Dumenil
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Wilson Nguyen
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Andrii Slonchak
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, QLD 4029 and 4072, Australia
| | - Julian Sng
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, QLD 4029 and 4072, Australia
| | - Viviana P Lutzky
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Daniel J Rawle
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Andreas Suhrbier
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
- Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, QLD 4029 and 4072, Australia
| |
Collapse
|
3
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Toll-like receptor response to Zika virus infection: progress toward infection control. NPJ VIRUSES 2025; 3:20. [PMID: 40295746 PMCID: PMC11906774 DOI: 10.1038/s44298-025-00102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025]
Abstract
Infection with the Zika virus (ZIKV) poses a threat to human health. An improved understanding of the host Toll-like receptor response, disease onset, and viral clearance in vivo and in vitro may lead to the development of therapeutic or prophylactic interventions against viral infections. Currently, no clinically approved ZIKV vaccine is available, highlighting the need for its development. In this study, we discuss the progress in the Zika vaccine, including advances in the use of Toll-like receptor agonists as vaccine adjuvants to enhance vaccine efficacy.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, Bangladesh.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
4
|
Torii S, Lord JS, Lavina M, Prot M, Lecuyer A, Diagne CT, Faye O, Faye O, Sall AA, Bonsall MB, Simon-Lorière E, Montagutelli X, Lambrechts L. Polygenic viral factors enable efficient mosquito-borne transmission of African Zika virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634482. [PMID: 39896559 PMCID: PMC11785240 DOI: 10.1101/2025.01.23.634482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus primarily transmitted among humans by Aedes aegypti. Over the past two decades, it has caused significant outbreaks associated with birth defects and neurological disorders. Phylogenetically, ZIKV consists of two main genotypes referred to as the African and Asian lineages, each exhibiting distinct biological properties. African lineage strains are transmitted more efficiently by mosquitoes, but pinpointing the genetic basis of this difference has remained challenging. Here, we address this question by comparing recent African and Asian strains using chimeric viruses, in which segments of the parental genomes are swapped. Our results show that the structural genes from the African strain enhance viral internalization, while the non-structural genes improve genome replication and infectious particle production in mosquito cells. In vivo mosquito transmission is most significantly influenced by the structural genes, although no single viral gene alone determines this effect. Additionally, we develop a stochastic model of in vivo viral dynamics in mosquitoes that mirrors the observed patterns, suggesting that the primary difference between the African and Asian strains lies in their ability to traverse the mosquito salivary glands. Overall, our findings suggest that the polygenic nature of ZIKV transmissibility has prevented Asian lineage strains from achieving the same epidemic potential as African lineage strains, underscoring the importance of lineage-specific adaptive landscapes in shaping ZIKV evolution and emergence.
Collapse
Affiliation(s)
- Shiho Torii
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Jennifer S. Lord
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Morgane Lavina
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Matthieu Prot
- Institut Pasteur, Université Paris Cité, Paris, Evolutionary Genomics of RNA Viruses Unit, Paris France
| | - Alicia Lecuyer
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Cheikh T. Diagne
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Oumar Faye
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Ousmane Faye
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Amadou A. Sall
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | | | - Etienne Simon-Lorière
- Institut Pasteur, Université Paris Cité, Paris, Evolutionary Genomics of RNA Viruses Unit, Paris France
| | - Xavier Montagutelli
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Louis Lambrechts
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| |
Collapse
|
5
|
Wang J, Yu Z, Chen Z, Ye F, Sun Z. The Potential Role of Zika and Dengue Virus Infection in the Urogenital System Disorders: An Overview. Rev Med Virol 2025; 35:e70010. [PMID: 39804234 DOI: 10.1002/rmv.70010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 05/02/2025]
Abstract
Arboviruses currently are regarded as a major worldwide public health concern. The clinical outcomes associated with this group of viruses may vary from asymptomatic infections to severe forms of haemorrhagic fever characterised by bleeding disorders. Similar to other systemic viral infections, arboviruses can either directly or indirectly affect different parts of the body, such as the urogenital system. The human urogenital system anatomically consists of two major subdivisions: (i) the urinary system, including the kidneys, ureters, bladder, and urethra, which plays a significant role in osmoregulation, control of blood volume, pressure, and PH, absorption/excretion of different ions, and toxin metabolism, and (ii) the genital system, composed of the prostate, uterus, testes, ovaries, penis, and vagina, which are responsible for reproductive functions. Arboviruses can impair normal urogenital system functions by direct viral pathogen activity, systemic forms of inflammation, haemorrhagic events and related dysfunctions, and the nephrotoxic side effects of specific medications employed for treatment leading to various urogenital disorders. The present review provides an overview of the potential capacity of two main arboviruses, known as Zika and dengue viruses, to affect the urogenital system. Moreover, it addresses Zika virus as a potential therapeutic oncolytic virus for urogenital cancers.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zongze Yu
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
| | - Zhigui Chen
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Darmuzey M, Touret F, Slowikowski E, Gladwyn-Ng I, Ahuja K, Sanchez-Felipe L, de Lamballerie X, Verfaillie C, Marques PE, Neyts J, Kaptein SJF. Epidemic Zika virus strains from the Asian lineage induce an attenuated fetal brain pathogenicity. Nat Commun 2024; 15:10870. [PMID: 39738084 PMCID: PMC11686291 DOI: 10.1038/s41467-024-55155-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
The 2015-2016 Zika virus (ZIKV) outbreak in the Americas revealed the ability of ZIKV from the Asian lineage to cause birth defects, generically called congenital Zika syndrome (CZS). Notwithstanding the long circulation history of Asian ZIKV, no ZIKV-associated CZS cases were reported prior to the outbreaks in French Polynesia (2013) and Brazil (2015). Whether the sudden emergence of CZS resulted from an evolutionary event of Asian ZIKV has remained unclear. We performed a comparative analysis of the pathogenicity of pre-epidemic and epidemic Asian ZIKV strains in mouse embryonic brains using a female immunocompetent intraplacental infection mouse model. All studied Asian ZIKV strains are neurovirulent, but pre-epidemic strains are consistently more pathogenic in the embryos than their epidemic equivalents. Pathogenicity is not directly linked to viral replication. By contrast, an influx of macrophages/microglial cells is noted in infected fetal brains for both pre-epidemic and epidemic ZIKV strains. Moreover, all tested ZIKV strains trigger an immunological response, whereby the intensity of the response differs between strains, and with epidemic ZIKV strains generally mounting a more attenuated immunostimulatory response. Our study reveals that Asian ZIKV strains evolved towards pathogenic attenuation, potentially resulting in CZS emergence in neonates rather than premature death in utero.
Collapse
Affiliation(s)
- Maïlis Darmuzey
- Virology and Immunology Unit, GIGA-Infection, Immunity and Inflammation, University of Liège, Liège, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium
| | - Franck Touret
- Unité Des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Emily Slowikowski
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, Leuven, Belgium
| | - Ivan Gladwyn-Ng
- Department of Application Scientists, Taconic Biosciences, Leverkusen, Germany
| | - Karan Ahuja
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium
| | - Xavier de Lamballerie
- Unité Des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Pedro E Marques
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium.
| | - Suzanne J F Kaptein
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium.
| |
Collapse
|
7
|
Abdelbasset M, Saron WAA, Ma D, Rathore APS, Kozaki T, Zhong C, Mantri CK, Tan Y, Tung CC, Tey HL, Chu JJH, Chen J, Ng LG, Wang H, Ginhoux F, St John AL. Differential contributions of fetal mononuclear phagocytes to Zika virus neuroinvasion versus neuroprotection during congenital infection. Cell 2024; 187:7511-7532.e20. [PMID: 39532096 DOI: 10.1016/j.cell.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/08/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Fetal immune cell functions during congenital infections are poorly understood. Zika virus (ZIKV) can vertically transmit from mother to fetus, causing nervous system infection and congenital ZIKV syndrome (CZS). We identified differential functional roles for fetal monocyte/macrophage cell types and microglia in ZIKV dissemination versus clearance using mouse models. Trafficking of ZIKV-infected primitive macrophages from the yolk sac allowed initial fetal virus inoculation, while recruited monocytes promoted non-productive neuroinflammation. Conversely, brain-resident differentiated microglia were protective, limiting infection and neuronal death. Single-cell RNA sequencing identified transcriptional profiles linked to the protective versus detrimental contributions of mononuclear phagocyte subsets. In human brain organoids, microglia also promoted neuroprotective transcriptional changes and infection clearance. Thus, microglia are protective before birth, contrasting with the disease-enhancing roles of primitive macrophages and monocytes. Differential modulation of myeloid cell phenotypes by genetically divergent ZIKVs underscores the potential of immune cells to regulate diverse outcomes during fetal infections.
Collapse
Affiliation(s)
- Muhammad Abdelbasset
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wilfried A A Saron
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Dongliang Ma
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Abhay P S Rathore
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA
| | - Tatsuya Kozaki
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chengwei Zhong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chinmay Kumar Mantri
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Yingrou Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Chi-Ching Tung
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Hong Liang Tey
- National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine affiliated Renji Hospital, Shanghai, China
| | - Hongyan Wang
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Ashley L St John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA; SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| |
Collapse
|
8
|
Chowdhury D, Lin XH, Seo SH. RT-qPCR Analysis of Inflammatory & Apoptotic Factors-Related Gene Expression in ZIKV-Infected IFNAR1 -/- Mice. Curr Microbiol 2024; 81:418. [PMID: 39432111 DOI: 10.1007/s00284-024-03943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
ZIKV was a mosquito-borne neglected tropical pathogen until it spread into the Pacific and South America, followed by large human outbreaks related to congenital abnormalities in neonates and neurological disorders in adults. The following study used the C57BL/6 IFNAR1 receptor knockout (IFN AR1-/-) mouse model to understand the role of selected cytokines and apoptotic factors in the pathogenicity of ZIKV strain PRVABC59. Mice infected with 102 particles of Zika viruses died until 9 days post infection. The brain, spleen, and lung were collected from intramuscularly infected mice on day 6 post infection (pi) to quantify the mRNA expression of targeted cytokines and apoptosis-mediated factors by RT-qPCR. Upregulation of IL-6, IL-17α, IFN-α, and IFN-β were found in the brain and lung of infected mice. IFN-γ was also significantly upregulated in the infected brain and spleen. The collective findings from our study indicate that a strong immune response was developed against ZIKV PRVABC59 in the infected mice brain.
Collapse
Affiliation(s)
- Dibakar Chowdhury
- Laboratory of Influenza Research, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Xiao Han Lin
- Laboratory of Influenza Research, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Sang Heui Seo
- Laboratory of Influenza Research, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Republic of Korea.
- Institute of Influenza Virus, Chungnam National University, 99 Dae-Hak Ro, Yuseong Gu, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
9
|
Macha NO, Komarasamy TV, Harun S, Adnan NAA, Hassan SS, Balasubramaniam VRMT. Cross Talk between MicroRNAs and Dengue Virus. Am J Trop Med Hyg 2024; 110:856-867. [PMID: 38579704 PMCID: PMC11066346 DOI: 10.4269/ajtmh.23-0546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/19/2023] [Indexed: 04/07/2024] Open
Abstract
Dengue fever (DF) is an endemic infectious tropical disease and is rapidly becoming a global problem. Dengue fever is caused by one of the four dengue virus (DENV) serotypes and is spread by the female Aedes mosquito. Clinical manifestations of DF may range from asymptomatic to life-threatening severe illness with conditions of hemorrhagic fever and shock. Early and precise diagnosis is vital to avoid mortality from DF. A different approach is required to combat DF because of the challenges with the vaccines currently available, which are nonspecific; each is capable of causing cross-reaction and disease-enhancing antibody responses against the residual serotypes. MicroRNAs (miRNAs) are known to be implicated in DENV infection and are postulated to be involved in most of the host responses. Thus, they might be a suitable target for new strategies against the disease. The involvement of miRNAs in cellular activities and pathways during viral infections has been explored under numerous conditions. Interestingly, miRNAs have also been shown to be involved in viral replication. In this review, we summarize the role of known miRNAs, specifically the role of miRNA Let-7c (miR-Let-7c), miR-133a, miR-30e, and miR-146a, in the regulation of DENV replication and their possible effects on the initial immune reaction.
Collapse
Affiliation(s)
- Nur Omar Macha
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Sarahani Harun
- Institute of Systems Biology Malaysia, National University of Malaysia, Selangor, Malaysia
| | - Nur Amelia Azreen Adnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Sharifah Syed Hassan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| | - Vinod R. M. T. Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
| |
Collapse
|
10
|
Henrio Marcellin DF, Huang J. Exploring Zika Virus Impact on Endothelial Permeability: Insights into Transcytosis Mechanisms and Vascular Leakage. Viruses 2024; 16:629. [PMID: 38675970 PMCID: PMC11054372 DOI: 10.3390/v16040629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Treating brain disease is challenging, and the Zika virus (ZIKV) presents a unique obstacle due to its neuroinvasive nature. In this review, we discuss the immunopathogenesis of ZIKV and explore how the virus interacts with the body's immune responses and the role of the protein Mfsd2a in maintaining the integrity of the blood-brain barrier (BBB) during ZIKV neuroinvasion. ZIKV has emerged as a significant public health concern due to its association with severe neurological problems, including microcephaly and Gillain-Barré Syndrome (GBS). Understanding its journey through the brain-particularly its interaction with the placenta and BBB-is crucial. The placenta, which is designed to protect the fetus, becomes a pathway for ZIKV when infected. The BBB is composed of brain endothelial cells, acts as a second barrier, and protects the fetal brain. However, ZIKV finds ways to disrupt these barriers, leading to potential damage. This study explores the mechanisms by which ZIKV enters the CNS and highlights the role of transcytosis, which allows the virus to move through the cells without significantly disrupting the BBB. Although the exact mechanisms of transcytosis are unclear, research suggests that ZIKV may utilize this pathway.
Collapse
Affiliation(s)
| | - Jufang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, China;
| |
Collapse
|
11
|
Nissly RH, Lim L, Keller MR, Bird IM, Bhushan G, Misra S, Chothe SK, Sill MC, Kumar NV, Sivakumar AVN, Naik BR, Jayarao BM, Kuchipudi SV. The Susceptibility of Chickens to Zika Virus: A Comprehensive Study on Age-Dependent Infection Dynamics and Host Responses. Viruses 2024; 16:569. [PMID: 38675911 PMCID: PMC11054531 DOI: 10.3390/v16040569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Zika virus (ZIKV) remains a public health concern, with epidemics in endemic regions and sporadic outbreaks in new areas posing significant threats. Several mosquito-borne flaviviruses that can cause human illness, including West Nile, Usutu, and St. Louis encephalitis, have associations with birds. However, the susceptibility of chickens to ZIKV and their role in viral epidemiology is not currently known. We investigated the susceptibility of chickens to experimental ZIKV infection using chickens ranging from 1-day-old chicks to 6-week-old birds. ZIKV caused no clinical signs in chickens of all age groups tested. Viral RNA was detected in the blood and tissues during the first 5 days post-inoculation in 1-day and 4-day-old chicks inoculated with a high viral dose, but ZIKV was undetectable in 6-week-old birds at all timepoints. Minimal antibody responses were observed in 6-week-old birds, and while present in younger chicks, they waned by 28 days post-infection. Innate immune responses varied significantly between age groups. Robust type I interferon and inflammasome responses were measured in older chickens, while limited innate immune activation was observed in younger chicks. Signal transducer and activator of transcription 2 (STAT2) is a major driver of host restriction to ZIKV, and chicken STAT2 is distinct from human STAT2, potentially contributing to the observed resistance to ZIKV infection. The rapid clearance of the virus in older chickens coincided with an effective innate immune response, highlighting age-dependent susceptibility. Our study indicates that chickens are not susceptible to productive ZIKV infection and are unlikely to play a role in the ZIKV epidemiology.
Collapse
Affiliation(s)
- Ruth H. Nissly
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802, USA; (R.H.N.); (L.L.); (M.R.K.); (I.M.B.); (G.B.); (B.M.J.)
| | - Levina Lim
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802, USA; (R.H.N.); (L.L.); (M.R.K.); (I.M.B.); (G.B.); (B.M.J.)
- DermBiont, Inc., 451 D Street, Suite 908, Boston, MA 02210, USA
| | - Margo R. Keller
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802, USA; (R.H.N.); (L.L.); (M.R.K.); (I.M.B.); (G.B.); (B.M.J.)
| | - Ian M. Bird
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802, USA; (R.H.N.); (L.L.); (M.R.K.); (I.M.B.); (G.B.); (B.M.J.)
- Applied Biological Sciences Group, The Johns Hopkins University Applied Physics Laboratory, Laurel, MD 20723, USA
| | - Gitanjali Bhushan
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802, USA; (R.H.N.); (L.L.); (M.R.K.); (I.M.B.); (G.B.); (B.M.J.)
- College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Sougat Misra
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.M.); (S.K.C.)
| | - Shubhada K. Chothe
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.M.); (S.K.C.)
| | - Miranda C. Sill
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA;
| | - Nagaram Vinod Kumar
- College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517 602, Andhra Pradesh, India; (N.V.K.); (A.V.N.S.); (B.R.N.)
| | - A. V. N. Sivakumar
- College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517 602, Andhra Pradesh, India; (N.V.K.); (A.V.N.S.); (B.R.N.)
| | - B. Rambabu Naik
- College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517 602, Andhra Pradesh, India; (N.V.K.); (A.V.N.S.); (B.R.N.)
| | - Bhushan M. Jayarao
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, PA 16802, USA; (R.H.N.); (L.L.); (M.R.K.); (I.M.B.); (G.B.); (B.M.J.)
| | - Suresh V. Kuchipudi
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (S.M.); (S.K.C.)
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
12
|
Rippee-Brooks MD, Wu W, Dong J, Pappolla M, Fang X, Bao X. Viral Infections, Are They a Trigger and Risk Factor of Alzheimer's Disease? Pathogens 2024; 13:240. [PMID: 38535583 PMCID: PMC10974111 DOI: 10.3390/pathogens13030240] [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: 02/01/2024] [Revised: 03/02/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024] Open
Abstract
Alzheimer's Disease (AD), a progressive and debilitating condition, is reported to be the most common type of dementia, with at least 55 million people believed to be currently affected. Many causation hypotheses of AD exist, yet the intriguing link between viral infection and its possible contribution to the known etiology of AD has become an attractive focal point of research for the field and a challenging study task. In this review, we will explore the historical perspective and milestones that led the field to investigate the viral connection to AD. Specifically, several viruses such as Herpes Simplex Virus 1 (HSV-1), Zika virus (ZIKV), and severe cute respiratory syndrome coronavirus 2 (SARS-CoV-2), along with several others mentioned, include the various viruses presently considered within the field. We delve into the strong evidence implicating these viruses in the development of AD such as the lytic replication and axonal transport of HSV-1, the various mechanisms of ZIKV neurotropism through the human protein Musashi-1 (MSI1), and the spread of SARS-CoV-2 through the transfer of the virus through the BBB endothelial cells to glial cells and then to neurons via transsynaptic transfer. We will also explore beyond these mere associations by carefully analyzing the potential mechanisms by which these viruses may contribute to AD pathology. This includes but is not limited to direct neuronal infections, the dysregulation of immune responses, and the impact on protein processing (Aβ42 and hyperphosphorylated tau). Controversies and challenges of the virus-AD relationship emerge as we tease out these potential mechanisms. Looking forward, we emphasize future directions, such as distinct questions and proposed experimentations to explore, that the field should take to tackle the remaining unanswered questions and the glaring research gaps that persist. Overall, this review aims to provide a comprehensive survey of the past, present, and future of the potential link between viral infections and their association with AD development while encouraging further discussion.
Collapse
Affiliation(s)
- Meagan D. Rippee-Brooks
- Microbiology and Immunology Graduate Program, Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Jianli Dong
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Miguel Pappolla
- Department of Neurology and Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Xiang Fang
- Department of Neurology and Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Xiaoyong Bao
- Microbiology and Immunology Graduate Program, Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77550, USA
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77550, USA
- The Institute of Translational Sciences, The University of Texas Medical Branch, Galveston, TX 77550, USA
- The Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX 77550, USA
| |
Collapse
|
13
|
Koenig MR, Vazquez J, Leyva Jaimes FB, Mitzey AM, Stanic AK, Golos TG. Decidual leukocytes respond to African lineage Zika virus infection with mild anti-inflammatory changes during acute infection in rhesus macaques. Front Immunol 2024; 15:1363169. [PMID: 38515747 PMCID: PMC10954895 DOI: 10.3389/fimmu.2024.1363169] [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: 12/29/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024] Open
Abstract
Zika virus (ZIKV) can be vertically transmitted during pregnancy resulting in a range of adverse pregnancy outcomes. The decidua is commonly found to be infected by ZIKV, yet the acute immune response to infection remains understudied in vivo. We hypothesized that in vivo African-lineage ZIKV infection induces a pro-inflammatory response in the decidua. To test this hypothesis, we evaluated the decidua in pregnant rhesus macaques within the first two weeks following infection with an African-lineage ZIKV and compared our findings to gestationally aged-matched controls. Decidual leukocytes were phenotypically evaluated using spectral flow cytometry, and cytokines and chemokines were measured in tissue homogenates from the decidua, placenta, and fetal membranes. The results of this study did not support our hypothesis. Although ZIKV RNA was detected in the decidual tissue samples from all ZIKV infected dams, phenotypic changes in decidual leukocytes and differences in cytokine profiles suggest that the decidua undergoes mild anti-inflammatory changes in response to that infection. Our findings emphasize the immunological state of the gravid uterus as a relatively immune privileged site that prioritizes tolerance of the fetus over mounting a pro-inflammatory response to clear infection.
Collapse
Affiliation(s)
- Michelle R. Koenig
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Jessica Vazquez
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Fernanda B. Leyva Jaimes
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Ann M. Mitzey
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Aleksandar K. Stanic
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Thaddeus G. Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
14
|
Elliott KC, Mattapallil JJ. Zika Virus-A Reemerging Neurotropic Arbovirus Associated with Adverse Pregnancy Outcomes and Neuropathogenesis. Pathogens 2024; 13:177. [PMID: 38392915 PMCID: PMC10892292 DOI: 10.3390/pathogens13020177] [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: 12/24/2023] [Revised: 02/07/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Zika virus (ZIKV) is a reemerging flavivirus that is primarily spread through bites from infected mosquitos. It was first discovered in 1947 in sentinel monkeys in Uganda and has since been the cause of several outbreaks, primarily in tropical and subtropical areas. Unlike earlier outbreaks, the 2015-2016 epidemic in Brazil was characterized by the emergence of neurovirulent strains of ZIKV strains that could be sexually and perinatally transmitted, leading to the Congenital Zika Syndrome (CZS) in newborns, and Guillain-Barre Syndrome (GBS) along with encephalitis and meningitis in adults. The immune response elicited by ZIKV infection is highly effective and characterized by the induction of both ZIKV-specific neutralizing antibodies and robust effector CD8+ T cell responses. However, the structural similarities between ZIKV and Dengue virus (DENV) lead to the induction of cross-reactive immune responses that could potentially enhance subsequent DENV infection, which imposes a constraint on the development of a highly efficacious ZIKV vaccine. The isolation and characterization of antibodies capable of cross-neutralizing both ZIKV and DENV along with cross-reactive CD8+ T cell responses suggest that vaccine immunogens can be designed to overcome these constraints. Here we review the structural characteristics of ZIKV along with the evidence of neuropathogenesis associated with ZIKV infection and the complex nature of the immune response that is elicited by ZIKV infection.
Collapse
Affiliation(s)
- Kenneth C. Elliott
- Department of Microbiology & Immunology, The Henry M Jackson Foundation for Military Medicine, Uniformed Services University, Bethesda, MD 20814, USA
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - Joseph J. Mattapallil
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
15
|
Ferreira JCCG, Christoff RR, Rabello T, Ferreira RO, Batista C, Mourão PJP, Rossi ÁD, Higa LM, Bellio M, Tanuri A, Garcez PP. Postnatal Zika virus infection leads to morphological and cellular alterations within the neurogenic niche. Dis Model Mech 2024; 17:dmm050375. [PMID: 38415826 PMCID: PMC10924234 DOI: 10.1242/dmm.050375] [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: 06/29/2023] [Accepted: 01/22/2024] [Indexed: 02/29/2024] Open
Abstract
The Zika virus received significant attention in 2016, following a declaration by the World Health Organization of an epidemic in the Americas, in which infections were associated with microcephaly. Indeed, prenatal Zika virus infection is detrimental to fetal neural stem cells and can cause premature cell loss and neurodevelopmental abnormalities in newborn infants, collectively described as congenital Zika syndrome. Contrastingly, much less is known about how neonatal infection affects the development of the newborn nervous system. Here, we investigated the development of the dentate gyrus of wild-type mice following intracranial injection of the virus at birth (postnatal day 0). Through this approach, we found that Zika virus infection affected the development of neurogenic regions within the dentate gyrus and caused reactive gliosis, cell death and a decrease in cell proliferation. Such infection also altered volumetric features of the postnatal dentate gyrus. Thus, we found that Zika virus exposure to newborn mice is detrimental to the subgranular zone of the dentate gyrus. These observations offer insight into the cellular mechanisms that underlie the neurological features of congenital Zika syndrome in children.
Collapse
Affiliation(s)
- Jéssica C. C. G. Ferreira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Raissa R. Christoff
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Tailene Rabello
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Raiane O. Ferreira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Carolina Batista
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Pedro Junior Pinheiro Mourão
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Átila D. Rossi
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Luiza M. Higa
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Maria Bellio
- Microbiology Institute Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Amilcar Tanuri
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Patricia P. Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| |
Collapse
|
16
|
Zoladek J, Nisole S. Mosquito-borne flaviviruses and type I interferon: catch me if you can! Front Microbiol 2023; 14:1257024. [PMID: 37965539 PMCID: PMC10642725 DOI: 10.3389/fmicb.2023.1257024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Mosquito-borne flaviviruses include many viruses that are important human pathogens, including Yellow fever virus, Dengue virus, Zika virus and West Nile virus. While these viruses have long been confined to tropical regions, they now pose a global public health concern, as the geographical distribution of their mosquito vectors has dramatically expanded. The constant threat of flavivirus emergence and re-emergence underlines the need for a better understanding of the relationships between these viruses and their hosts. In particular, unraveling how these viruses manage to bypass antiviral immune mechanisms could enable the design of countermeasures to limit their impact on human health. The body's first line of defense against viral infections is provided by the interferon (IFN) response. This antiviral defense mechanism takes place in two waves, namely the induction of type I IFNs triggered by viral infection, followed by the IFN signaling pathway, which leads to the synthesis of interferon-stimulated genes (ISGs), whose products inhibit viral replication. In order to spread throughout the body, viruses must race against time to replicate before this IFN-induced antiviral state hinders their dissemination. In this review, we summarize our current knowledge on the multiple strategies developed by mosquito-borne flaviviruses to interfere with innate immune detection and signaling pathways, in order to delay, if not prevent, the establishment of an antiviral response.
Collapse
Affiliation(s)
| | - Sébastien Nisole
- Viral Trafficking, Restriction and Innate Signaling, CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| |
Collapse
|
17
|
Song GY, Huang XY, He MJ, Zhou HY, Li RT, Tian Y, Wang Y, Cheng ML, Chen X, Zhang RR, Zhou C, Zhou J, Fang XY, Li XF, Qin CF. A single amino acid substitution in the capsid protein of Zika virus contributes to a neurovirulent phenotype. Nat Commun 2023; 14:6832. [PMID: 37884553 PMCID: PMC10603150 DOI: 10.1038/s41467-023-42676-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Increasing evidence shows the African lineage Zika virus (ZIKV) displays a more severe neurovirulence compared to the Asian ZIKV. However, viral determinants and the underlying mechanisms of enhanced virulence phenotype remain largely unknown. Herein, we identify a panel of amino acid substitutions that are unique to the African lineage of ZIKVs compared to the Asian lineage by phylogenetic analysis and sequence alignment. We then utilize reverse genetic technology to generate recombinant ZIKVs incorporating these lineage-specific substitutions based on an infectious cDNA clone of Asian ZIKV. Through in vitro characterization, we discover a mutant virus with a lysine to arginine substitution at position 101 of capsid (C) protein (termed K101R) displays a larger plaque phenotype, and replicates more efficiently in various cell lines. Moreover, K101R replicates more efficiently in mouse brains and induces stronger inflammatory responses than the wild type (WT) virus in neonatal mice. Finally, a combined analysis reveals the K101R substitution promotes the production of mature C protein without affecting its binding to viral RNA. Our study identifies the role of K101R substitution in the C protein in contributing to the enhanced virulent phenotype of the African lineage ZIKV, which expands our understanding of the complexity of ZIKV proteins.
Collapse
Affiliation(s)
- Guang-Yuan Song
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Xing-Yao Huang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Meng-Jiao He
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Hang-Yu Zhou
- Suzhou Institute of System Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 215123, Suzhou, Jiangsu, China
| | - Rui-Ting Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Ying Tian
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Yan Wang
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Meng-Li Cheng
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Xiang Chen
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Rong-Rong Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Chao Zhou
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Jia Zhou
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Xian-Yang Fang
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China.
| | - Cheng-Feng Qin
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China.
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China.
| |
Collapse
|
18
|
Narayan R, Sharma M, Yadav R, Biji A, Khatun O, Kaur S, Kanojia A, Joy CM, Rajmani R, Sharma PR, Jeyasankar S, Rani P, Shandil RK, Narayanan S, Rao DC, Satchidanandam V, Das S, Agarwal R, Tripathi S. Picolinic acid is a broad-spectrum inhibitor of enveloped virus entry that restricts SARS-CoV-2 and influenza A virus in vivo. Cell Rep Med 2023; 4:101127. [PMID: 37463584 PMCID: PMC10439173 DOI: 10.1016/j.xcrm.2023.101127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 02/06/2023] [Accepted: 06/23/2023] [Indexed: 07/20/2023]
Abstract
The COVID-19 pandemic highlights an urgent need for effective antivirals. Targeting host processes co-opted by viruses is an attractive antiviral strategy with a high resistance barrier. Picolinic acid (PA) is a tryptophan metabolite endogenously produced in mammals. Here, we report the broad-spectrum antiviral activity of PA against enveloped viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza A virus (IAV), flaviviruses, herpes simplex virus, and parainfluenza virus. Mechanistic studies reveal that PA inhibits enveloped virus entry by compromising viral membrane integrity, inhibiting virus-cellular membrane fusion, and interfering with cellular endocytosis. More importantly, in pre-clinical animal models, PA exhibits promising antiviral efficacy against SARS-CoV-2 and IAV. Overall, our data establish PA as a broad-spectrum antiviral with promising pre-clinical efficacy against pandemic viruses SARS-CoV-2 and IAV.
Collapse
Affiliation(s)
- Rohan Narayan
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Mansi Sharma
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Rajesh Yadav
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Abhijith Biji
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Oyahida Khatun
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Sumandeep Kaur
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Aditi Kanojia
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Christy Margrat Joy
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Raju Rajmani
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru 560012, India
| | - Pallavi Raj Sharma
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Sharumathi Jeyasankar
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Priya Rani
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Radha Krishan Shandil
- Foundation for Neglected Disease Research, KIADB Industrial Area, Doddaballapur, Bengaluru 561203, India
| | - Shridhar Narayanan
- Foundation for Neglected Disease Research, KIADB Industrial Area, Doddaballapur, Bengaluru 561203, India
| | - Durga Chilakalapudi Rao
- Department of Biological Sciences, School of Engineering and Sciences, SRM University, Andhra Pradesh 522240, India
| | - Vijaya Satchidanandam
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Saumitra Das
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Rachit Agarwal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Shashank Tripathi
- Emerging Viral Pathogens Laboratory, Infosys Wing, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru 560012, India; Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bengaluru 560012, India.
| |
Collapse
|
19
|
Jeong GU, Lee S, Kim DY, Lyu J, Yoon GY, Kim KD, Ku KB, Ko J, Kwon YC. Zika Virus Infection Induces Interleukin-1β-Mediated Inflammatory Responses by Macrophages in the Brain of an Adult Mouse Model. J Virol 2023; 97:e0055623. [PMID: 37191498 PMCID: PMC10308908 DOI: 10.1128/jvi.00556-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 04/23/2023] [Indexed: 05/17/2023] Open
Abstract
During the 2015-2016 Zika virus (ZIKV) epidemic, ZIKV-associated neurological diseases were reported in adults, including microcephaly, Guillain-Barre syndrome, myelitis, meningoencephalitis, and fatal encephalitis. However, the mechanisms underlying the neuropathogenesis of ZIKV infection are not yet fully understood. In this study, we used an adult ZIKV infection mouse model (Ifnar1-/-) to investigate the mechanisms underlying neuroinflammation and neuropathogenesis. ZIKV infection induced the expression of proinflammatory cytokines, including interleukin-1β (IL-1β), IL-6, gamma interferon, and tumor necrosis factor alpha, in the brains of Ifnar1-/- mice. RNA-seq analysis of the infected mouse brain also revealed that genes involved in innate immune responses and cytokine-mediated signaling pathways were significantly upregulated at 6 days postinfection. Furthermore, ZIKV infection induced macrophage infiltration and activation and augmented IL-1β expression, whereas microgliosis was not observed in the brain. Using human monocyte THP-1 cells, we confirmed that ZIKV infection promotes inflammatory cell death and increases IL-1β secretion. In addition, expression of the complement component C3, which is associated with neurodegenerative diseases and known to be upregulated by proinflammatory cytokines, was induced by ZIKV infection through the IL-1β-mediated pathway. An increase in C5a produced by complement activation in the brains of ZIKV-infected mice was also verified. Taken together, our results suggest that ZIKV infection in the brain of this animal model augments IL-1β expression in infiltrating macrophages and elicits IL-1β-mediated inflammation, which can lead to the destructive consequences of neuroinflammation. IMPORTANCE Zika virus (ZIKV) associated neurological impairments are an important global health problem. Our results suggest that ZIKV infection in the mouse brain can induce IL-1β-mediated inflammation and complement activation, thereby contributing to the development of neurological disorders. Thus, our findings reveal a mechanism by which ZIKV induces neuroinflammation in the mouse brain. Although we used adult type I interferon receptor IFNAR knockout (Ifnar1-/-) mice owing to the limited mouse models of ZIKV pathogenesis, our conclusions contributed to the understanding ZIKV-associated neurological diseases to develop treatment strategies for patients with ZIKV infection based on these findings.
Collapse
Affiliation(s)
- Gi Uk Jeong
- Department of Convergent Research for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sumin Lee
- Department of Convergent Research for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Do Yeon Kim
- Department of Convergent Research for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medical Chemistry and Pharmacology, University of Science and Technology, Daejeon, Republic of Korea
| | - Jaemyun Lyu
- Arontier Co., Ltd., Seoul, Republic of Korea
| | - Gun Young Yoon
- Department of Convergent Research for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Kyun-Do Kim
- Department of Convergent Research for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Keun Bon Ku
- Department of Convergent Research for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Junsu Ko
- Arontier Co., Ltd., Seoul, Republic of Korea
| | - Young-Chan Kwon
- Department of Convergent Research for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medical Chemistry and Pharmacology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
20
|
Lu AY, Gustin A, Newhouse D, Gale M. Viral Protein Accumulation of Zika Virus Variants Links with Regulation of Innate Immunity for Differential Control of Viral Replication, Spread, and Response to Interferon. J Virol 2023; 97:e0198222. [PMID: 37162358 PMCID: PMC10231147 DOI: 10.1128/jvi.01982-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
Asian lineage Zika virus (ZIKV) strains emerged globally, causing outbreaks linked with critical clinical disease outcomes unless the virus is effectively restricted by host immunity. We have previously shown that retinoic acid-inducible gene-I (RIG-I) senses ZIKV to trigger innate immunity to direct interferon (IFN) production and antiviral responses that can control ZIKV infection. However, ZIKV proteins have been demonstrated to antagonize IFN. Here, we conducted in vitro analyses to assess how divergent prototypic ZIKV variants differ in virologic properties, innate immune regulation, and infection outcome. We comparatively assessed African lineage ZIKV/Dakar/1984/ArD41519 (ZIKV/Dakar) and Asian lineage ZIKV/Malaysia/1966/P6740 (ZIKV/Malaysia) in a human epithelial cell infection model. De novo viral sequence determination identified amino acid changes within the ZIKV/Dakar genome compared to ZIKV/Malaysia. Viral growth analyses revealed that ZIKV/Malaysia accumulated viral proteins and genome copies earlier and to higher levels than ZIKV/Dakar. Both ZIKV strains activated RIG-I/IFN regulatory factor (IRF3) and NF-κB pathways to induce inflammatory cytokine expression and types I and III IFNs. However, ZIKV/Malaysia, but not ZIKV/Dakar, potently blocked downstream IFN signaling. Remarkably, ZIKV/Dakar protein accumulation and genome replication were rescued in RIG-I knockout (KO) cells late in acute infection, resulting in ZIKV/Dakar-mediated blockade of IFN signaling. We found that RIG-I signaling specifically restricts viral protein accumulation late in acute infection where early accumulation of viral proteins in infected cells confers enhanced ability to limit IFN signaling, promoting viral replication and spread. Our results demonstrate that RIG-I-mediated innate immune signaling imparts restriction of ZIKV protein accumulation, which permits IFN signaling and antiviral actions controlling ZIKV infection. IMPORTANCE ZIKV isolates are classified under African or Asian lineages. Infection with emerging Asian lineage-derived ZIKV strains is associated with increased incidence of neurological symptoms that were not previously reported during infection with African or preemergent Asian lineage viruses. In this study, we utilized in vitro models to compare the virologic properties of and innate immune responses to two prototypic ZIKV strains from distinct lineages: African lineage ZIKV/Dakar and Asian lineage ZIKV/Malaysia. Compared to ZIKV/Dakar, ZIKV/Malaysia accumulates viral proteins earlier, replicates to higher levels, and robustly blocks IFN signaling during acute infection. Early accumulation of ZIKV/Malaysia NS5 protein confers enhanced ability to antagonize IFN signaling, dampening innate immune responses to promote viral spread. Our data identify the kinetics of viral protein accumulation as a major regulator of host innate immunity, influencing host-mediated control of ZIKV replication and spread. Importantly, these findings provide a novel framework for evaluating the virulence of emerging variants.
Collapse
Affiliation(s)
- Amy Y. Lu
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Andrew Gustin
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Daniel Newhouse
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
21
|
He MJ, Wang HJ, Yan XL, Lou YN, Song GY, Li RT, Zhu Z, Zhang RR, Qin CF, Li XF. Key Residue in the Precursor Region of M Protein Contributes to the Neurovirulence and Neuroinvasiveness of the African Lineage of Zika Virus. J Virol 2023; 97:e0180122. [PMID: 36840584 PMCID: PMC10062131 DOI: 10.1128/jvi.01801-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/31/2023] [Indexed: 02/24/2023] Open
Abstract
The Zika virus (ZIKV) represents an important global health threat due to its unusual association with congenital Zika syndrome. ZIKV strains are phylogenetically grouped into the African and Asian lineages. However, the viral determinants underlying the phenotypic differences between the lineages remain unknown. Here, multiple sequence alignment revealed a highly conserved residue at position 21 of the premembrane (prM) protein, which is glutamic acid and lysine in the Asian and African lineages, respectively. Using reverse genetics, we generated a recombinant virus carrying an E21K mutation based on the genomic backbone of the Asian lineage strain FSS13025 (termed E21K). The E21K mutation significantly increased viral replication in multiple neural cell lines with a higher ratio of M to prM production. Animal studies showed E21K exhibited increased neurovirulence in suckling mice, leading to more severe defects in mouse brains by causing more neural cell death and destruction of hippocampus integrity. Moreover, the E21K substitution enhanced neuroinvasiveness in interferon alpha/beta (IFN-α/β) receptor knockout mice, as indicated by the increased mortality, and enhanced replication in mouse brains. The global transcriptional analysis showed E21K infection profoundly altered neuron development networks and induced stronger antiviral immune response than wild type (WT) in both neural cells and mouse brains. More importantly, the reverse K21E mutation based on the genomic backbone of the African strain MR766 caused less mouse neurovirulence. Overall, our findings support the 21st residue of prM functions as a determinant for neurovirulence and neuroinvasiveness of the African lineage of ZIKV. IMPORTANCE The suspected link of Zika virus (ZIKV) to birth defects led the World Health Organization to declare ZIKV a Public Health Emergency of International Concern. ZIKV has been identified to have two dominant phylogenetic lineages, African and Asian. Significant differences exist between the two lineages in terms of neurovirulence and neuroinvasiveness in mice. However, the viral determinants underlying the phenotypic differences are still unknown. Here, combining reverse genetics, animal studies, and global transcriptional analysis, we provide evidence that a single E21K mutation of prM confers to the Asian lineage strain FSS130125 significantly enhanced replication in neural cell lines and more neurovirulent and neuroinvasiveness phenotypes in mice. Our findings support that the highly conserved residue at position 21 of prM functions as a determinant of neurovirulence and neuroinvasiveness of the African lineage of ZIKV in mice.
Collapse
Affiliation(s)
- Meng-Jiao He
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Hong-Jiang Wang
- Department of Research, The Chinese People’s Liberation Army Strategic Support Force Medical Center, Beijing, China
| | - Xiu-Li Yan
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ya-Nan Lou
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Guang-Yuan Song
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Rui-Ting Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Zhu Zhu
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Rong-Rong Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Wang K, Zou S, Chen H, Higazy D, Gao X, Zhang Y, Cao S, Cui M. Zika virus replication on endothelial cells and invasion into the central nervous system by inhibiting interferon β translation. Virology 2023; 582:23-34. [PMID: 36996689 DOI: 10.1016/j.virol.2023.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/02/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023]
Abstract
The blood-brain barrier (BBB) is one of the tightest physical barriers to prevent pathogens from invading the central nervous system (CNS). However, the mechanism by which Zika virus (ZIKV) crossing the BBB remains unresolved. We found ZIKV induced high morbidity and mortality in newborn mice, accompanied by inflammatory injury on CNS. ZIKV was found to replicate primarily in the cortex and hippocampus in neonatal mouse brains. An in vitro model revealed that ZIKV had no impact on hBMECs permeability but led to endothelial activation, as shown by the enhancement of adhesion molecules expression and F-actin redistribution. ZIKV replication in hBMECs might be associated with the suppression of IFN-β translation via inhibiting RPS6 phosphorylation. On the other hand, ZIKV infection induced IFN-stimulated genes (ISGs), activated the mitogen-activated protein kinase (MAPK) signaling pathway, and promoted chemokine secretion. This study provides an understanding of virus replication and transmigration across the BBB during ZIKV infection.
Collapse
|
23
|
Li N, Deng CL, Li Q, Chen XL, Zhang B, Ye HQ. A safe replication-defective Zika virus vaccine protects mice from viral infection and vertical transmission. Antiviral Res 2023; 211:105549. [PMID: 36690159 DOI: 10.1016/j.antiviral.2023.105549] [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: 10/23/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
With the explosive emergence of Zika virus (ZIKV) and the consequent devastating fetal malformations in infected expectant women, a safe and effective vaccine is urgently needed. Here, using our established NS1 trans-complementation system, we generated high titer of replication-defective ZIKV with NS1 deletion (ZIKV-ΔNS1) in the BHK-21 cell line stably expressing NS1 (BHKNS1). NS1 deletion of ZIKV-ΔNS1 was stably maintained as no replicative virus was found in naïve BHK-21 cells after continuous passaging of ZIKV-ΔNS1 in BHKNS1 cells. The safety of ZIKV-ΔNS1 was demonstrated when a high dose of ZIKV-ΔNS1 (107 IU) was used to infect the highly susceptible type I and type II interferon (IFN) receptor-deficient mice. ZIKV-ΔNS1 could induce antibody responses in both immunocompetent (BALB/c) and immunodeficient mice and a single dose of ZIKV-ΔNS1 vaccine protected the immunodeficient mice from a highly lethal dosage of challenge with WT ZIKV. ZIKV-ΔNS1 immunization also attenuated vertical transmission during pregnancy of type I IFN receptor-deficient IFNAR-/- mice and protected fetuses from ZIKV infection. Our data reported here not only provide a promising ZIKV vaccine candidate with a satisfied balance between safety and efficacy, but also demonstrate the potential of the NS1 trans-complementation system as a platform for flavivirus vaccine development, especially for highly pathogenic flaviviruses.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qi Li
- College of Pharmacy and Drug Discovery Center for Infectious Diseases, Nankai University, Tianjin, 300350, China
| | - Xiao-Ling Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Han-Qing Ye
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
24
|
Chan YT, Cheok YY, Cheong HC, Tang TF, Sulaiman S, Hassan J, Looi CY, Tan KK, AbuBakar S, Wong WF. Immune Recognition versus Immune Evasion Systems in Zika Virus Infection. Biomedicines 2023; 11:biomedicines11020642. [PMID: 36831177 PMCID: PMC9952926 DOI: 10.3390/biomedicines11020642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/22/2023] Open
Abstract
The reemergence of the Zika virus (ZIKV) infection in recent years has posed a serious threat to global health. Despite being asymptomatic or mildly symptomatic in a majority of infected individuals, ZIKV infection can result in severe manifestations including neurological complications in adults and congenital abnormalities in newborns. In a human host, ZIKV is primarily recognized by RIG-like receptors and Toll-like receptors that elicit anti-viral immunity through the secretion of type I interferon (IFN) to limit viral survival, replication, and pathogenesis. Intriguingly, ZIKV evades its host immune system through various immune evasion strategies, including suppressing the innate immune receptors and signaling pathways, mutation of viral structural and non-structural proteins, RNA modulation, or alteration of cellular pathways. Here, we present an overview of ZIKV recognition by the host immune system and the evasion strategies employed by ZIKV. Characterization of the host-viral interaction and viral disease mechanism provide a platform for the rational design of novel prophylactic and therapeutic strategies against ZIKV infection.
Collapse
Affiliation(s)
- Yee Teng Chan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Ting Fang Tang
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Sofiah Sulaiman
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Jamiyah Hassan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor’s University, 1, Jalan Taylors, Subang Jaya 47500, Malaysia
| | - Kim-Kee Tan
- Tropical Infectious Diseases Research and Education Centre (TIDREC), Higher Education Center of Excellence (HICoE), University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Centre (TIDREC), Higher Education Center of Excellence (HICoE), University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: ; Tel.: +60-(3)-7967-6672
| |
Collapse
|
25
|
Schmitt K, Curlin JZ, Remling-Mulder L, Aboellail T, Akkina R. Zika virus induced microcephaly and aberrant hematopoietic cell differentiation modeled in novel neonatal humanized mice. Front Immunol 2023; 14:1060959. [PMID: 36825016 PMCID: PMC9941325 DOI: 10.3389/fimmu.2023.1060959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Immunocompetent and immunocompromised murine models have been instrumental in answering important questions regarding ZIKV pathogenesis and vertical transmission. However, mimicking human congenital zika syndrome (CZS) characteristics in these murine models has been less than optimal and does not address the potential viral effects on the human immune system. Methods Here, we utilized neonatal humanized Rag2-/-γc-/- mice to model CZS and evaluate the potential viral effects on the differentiation of human hematopoietic stem cells in vivo. Newborn Rag2-/-γc-/- mice were engrafted with ZIKV-infected hematopoietic stem cells (HSC) and monitored for symptoms and lesions. Results Within 13 days, mice displayed outward clinical symptoms that encompassed stunted growth, hunched posture, ruffled fur, and ocular defects. Striking gross pathologies in the brain and visceral organs were noted. Our results also confirmed that ZIKV actively infected human CD34+ hematopoietic stem cells and restricted the development of terminally differentiated B cells. Histologically, there was multifocal mineralization in several different regions of the brain together with ZIKV antigen co-localization. Diffuse necrosis of pyramidal neurons was seen with collapse of the hippocampal formation. Discussion Overall, this model recapitulated ZIKV microcephaly and CZS together with viral adverse effects on the human immune cell ontogeny thus providing a unique in vivo model to assess the efficacy of novel therapeutics and immune interventions.
Collapse
|
26
|
Gilbert RK, Petersen LR, Honein MA, Moore CA, Rasmussen SA. Zika virus as a cause of birth defects: Were the teratogenic effects of Zika virus missed for decades? Birth Defects Res 2023; 115:265-274. [PMID: 36513609 PMCID: PMC10552063 DOI: 10.1002/bdr2.2134] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022]
Abstract
Zika virus (ZIKV) was identified as a teratogen in 2016 when an increase in severe microcephaly and other brain defects was observed in fetuses and newborns following outbreaks in French Polynesia (2013-2014) and Brazil (2015-2016) and among travelers to other countries experiencing outbreaks. Some have questioned why ZIKV was not recognized as a teratogen before these outbreaks: whether novel genetic changes in ZIKV had increased its teratogenicity or whether its association with birth defects had previously been undetected. Here we examine the evidence for these two possibilities. We describe evidence for specific mutations that arose before the French Polynesia outbreak that might have increased ZIKV teratogenicity. We also present information on children born with findings consistent with congenital Zika syndrome (CZS) as early as 2009 and epidemiological evidence that suggests increases in CZS-type birth defects before 2013. We also explore reasons why a link between ZIKV and birth defects might have been missed, including issues with surveillance of ZIKV infections and of birth defects, challenges to ZIKV diagnostic testing, and the susceptibility of different populations to ZIKV infection at the time of pregnancy. Although it is not possible to prove definitively that ZIKV had teratogenic properties before 2013, several pieces of evidence support the hypothesis that its teratogenicity had been missed in the past. These findings emphasize the need for further investments in global surveillance for emerging infections and for birth defects so that infectious teratogens can be identified more expeditiously in the future.
Collapse
Affiliation(s)
- Rachel K. Gilbert
- University of Florida College of Medicine, Gainesville, Florida, USA
| | - Lyle R. Petersen
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Margaret A. Honein
- Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Cynthia A. Moore
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Goldbelt Professional Services, LLC, Chesapeake, Virginia, USA
| | - Sonja A. Rasmussen
- Departments of Pediatrics and Obstetrics and Gynecology, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Epidemiology, College of Medicine and College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
27
|
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
|
28
|
Pathogenicity and Structural Basis of Zika Variants with Glycan Loop Deletions in the Envelope Protein. J Virol 2022; 96:e0087922. [PMID: 36377874 PMCID: PMC9749469 DOI: 10.1128/jvi.00879-22] [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: 11/16/2022] Open
Abstract
The glycan loop of Zika virus (ZIKV) envelope protein (E) contains the glycosylation site and has been well documented to be important for viral pathogenesis and transmission. In the present study, we report that deletions in the E glycan loop, which were recorded in African ZIKV strains previously, have re-emerged in their contemporary Asian lineages. Here, we generated recombinant ZIKV containing specific deletions in the E glycan loop by reverse genetics. Extensive in vitro and in vivo characterization of these deletion mutants demonstrated an attenuated phenotype in an adult A129 mouse model and reduced oral infections in mosquitoes. Surprisingly, these glycan loop deletion mutants exhibited an enhanced neurovirulence phenotype, and resulted in a more severe microcephalic brain in neonatal mouse models. Crystal structures of the ZIKV E protein and a deletion mutant at 2.5 and 2.6 Å, respectively, revealed that deletion of the glycan loop induces encephalitic flavivirus-like conformational alterations, including the appearance of perforations on the surface and a clear change in the topology of the loops. Overall, our results demonstrate that the E glycan loop deletions represent neonatal mouse neurovirulence markers of ZIKV. IMPORTANCE Zika virus (ZIKV) has been identified as a cause of microcephaly and acquired evolutionary mutations since its discovery. Previously deletions in the E glycan loop were recorded in African ZIKV strains, which have re-emerged in the contemporary Asian lineages recently. The glycan loop deletion mutants are not glycosylated, which are attenuated in adult A129 mouse model and reduced oral infections in mosquitoes. More importantly, the glycan loop deletion mutants induce an encephalitic flavivirus-like conformational alteration in the E homodimer, resulting in a significant enhancement of neonatal mouse neurovirulence. This study underscores the critical role of glycan loop deletion mutants in ZIKV pathogenesis, highlighting a need for global virological surveillance for such ZIKV variants.
Collapse
|
29
|
The Innate Defense in the Zika-Infected Placenta. Pathogens 2022; 11:pathogens11121410. [PMID: 36558744 PMCID: PMC9787577 DOI: 10.3390/pathogens11121410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Zika virus (ZIKV) is an arthropod-borne virus that belongs to the Flaviviridae family, genus Flavivirus and was first isolated 1947 in Uganda, Africa, from the serum of a sentinel Rhesus monkey. Since its discovery, the virus was responsible for major outbreaks in several different countries, being linked to severe complications in pregnant women, neonatal birth defects and the congenital zika syndrome. Maternal-fetal transmission of ZIKV can occur in all trimesters of pregnancy, and the role of the placenta and its cells in these cases is yet to be fully understood. The decidua basalis and chorionic villi, maternal-fetal components of the placenta, contain a rich immunological infiltrate composed by Hofbauer cells, mastocytes, dendritic cells and macrophages, primary cells of the innate immune response that have a role that still needs to be better investigated in ZIKV infection. Recent studies have already described several histopathological features and the susceptibility and permissiveness of placenta cells to infection by the Zika virus. In this review, we address some of the current knowledge on the innate immune responses against ZIKV, especially in the placenta.
Collapse
|
30
|
Chen ZL, Yin ZJ, Qiu TY, Chen J, Liu J, Zhang XY, Xu JQ. Revealing the characteristics of ZIKV infection through tissue-specific transcriptome sequencing analysis. BMC Genomics 2022; 23:697. [PMID: 36209057 PMCID: PMC9546753 DOI: 10.1186/s12864-022-08919-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recently, Zika virus (ZIKV) re-emerged in India and was potentially associated with microcephaly. However, the molecular mechanisms underlying ZIKV pathogenesis remain to be explored. RESULTS Herein, we performed a comprehensive RNA-sequencing analysis on ZIKV-infected JEG-3, U-251 MG, and HK-2 cells versus corresponding uninfected controls. Combined with a series of functional analyses, including gene annotation, pathway enrichment, and protein-protein interaction (PPI) network analysis, we defined the molecular characteristics induced by ZIKV infection in different tissues and invasion time points. Data showed that ZIKV infection and replication in each susceptible organ commonly stimulated interferon production and down-regulated metabolic-related processes. Also, tissue-specific immune responses or biological processes (BPs) were induced after ZIKV infection, including GnRH signaling pathway in JEG-3 cells, MAPK signaling pathway in U-251 MG cells, and PPAR signaling pathway in HK-2 cells. Of note, ZIKV infection induced delayed antiviral interferon responses in the placenta-derived cell lines, which potentially explains the molecular mechanism by which ZIKV replicates rapidly in the placenta and subsequential vertical transmission occurs. CONCLUSIONS Together, these data may provide a systemic insight into the pathogenesis of ZIKV infection in distinct human tissue-derived cell lines, which is likely to help develop prophylactic and therapeutic strategies against ZIKV infection.
Collapse
Affiliation(s)
- Zhi-Lu Chen
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Zuo-Jing Yin
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tian-Yi Qiu
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Immunotherapy and Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jian Chen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jian Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Xiao-Yan Zhang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China. .,Department of Immunotherapy and Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jian-Qing Xu
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China. .,Department of Immunotherapy and Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
31
|
Abstract
Zika virus (ZIKV) is unusual among flaviviruses in its ability to spread between humans through sexual contact, as well as by mosquitoes. Sexual transmission has the potential to change the epidemiology and geographic range of ZIKV compared to mosquito-borne transmission and potentially could produce distinct clinical manifestations, so it is important to understand the host mechanisms that control susceptibility to sexually transmitted ZIKV. ZIKV replicates poorly in wild-type mice following subcutaneous inoculation, so most ZIKV pathogenesis studies use mice lacking type I interferon (IFN-αβ) signaling (e.g., Ifnar1-/-). We found that wild-type mice support ZIKV replication following intravaginal infection, consistent with prior studies, although the infection remained localized to the lower female reproductive tract. Vaginal ZIKV infection required a high-progesterone state (pregnancy or pretreatment with depot medroxyprogesterone acetate [DMPA]) even in Ifnar1-/- mice that otherwise are highly susceptible to ZIKV infection. Progesterone-mediated susceptibility did not appear to result from a compromised epithelial barrier, blunted antiviral gene induction, or changes in vaginal leukocyte populations, leaving open the mechanism by which progesterone confers susceptibility to vaginal ZIKV infection. DMPA treatment is a key component of mouse vaginal infection models for herpes simplex virus and Chlamydia, but the mechanisms by which DMPA increases susceptibility to those pathogens also remain poorly defined. Understanding how progesterone mediates susceptibility to ZIKV vaginal infection may provide insights into host mechanisms influencing susceptibility to diverse sexually transmitted pathogens. IMPORTANCE Zika virus (ZIKV) is transmitted by mosquitoes, similar to other flaviviruses. However, ZIKV is unusual among flaviviruses in its ability also to spread through sexual transmission. We found that ZIKV was able to replicate in the vaginas of wild-type mice, even though these mice do not support ZIKV replication by other routes, suggesting that the vagina is particularly susceptible to ZIKV infection. Vaginal susceptibility was dependent on a high-progesterone state, which is a common feature of mouse vaginal infection models for other pathogens, through mechanisms that have remained poorly defined. Understanding how progesterone mediates susceptibility to ZIKV vaginal infection may provide insights into host mechanisms that influence susceptibility to diverse sexually transmitted pathogens.
Collapse
|
32
|
Shang J, Li C, Jin Z, Zu S, Chen S, Chen J, Chen Z, Tang H, Qin CF, Ye Q, Wu A. Immune profiles in mouse brain and testes infected by Zika virus with variable pathogenicity. Front Cell Infect Microbiol 2022; 12:948980. [PMID: 35992167 PMCID: PMC9385972 DOI: 10.3389/fcimb.2022.948980] [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: 05/20/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The Zika virus is responsible for neurological diseases such as microcephaly, Guillain-Barré syndrome, neuropathy, and myelitis in human adults and children. Previous studies have shown that the Zika virus can infect nerve progenitor cells and interfere with neural development. However, it is unclear how the immune system responds to infection with Zika viruses with variable pathogenicity. Here, we used two Zika strains with relatively different pathogenicity, the Asian ancestral strain CAM/2010 and the America pandemic strain GZ01/2016, to infect the brains of mice. We found that both strains elicited a strong immune response. Notably, the strain with relatively high pathogenicity, GZ01/2016, caused more intense immune regulation, with stronger CD8+ T cell and macrophage activation at 14 days post infection (dpi), as well as a greater immune gene disturbance. Notably, several TNF family genes were upregulated at 14 dpi, including Tnfrsf9, Tnfsf13, Tnfrsf8, Cd40, and Tnfsf10. It was notable that GZ01/2016 could maintain the survival of nerve cells at 7dpi but caused neurological disorders at 14dpi. These results indicate that Zika viruses with high pathogenicity may induce sustained activation of the immune system leading to nerve tissue damage.
Collapse
Affiliation(s)
- Jingzhe Shang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Chunfeng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States
| | - Zhujia Jin
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Shulong Zu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Songjie Chen
- Departments of Genetics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Junlan Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology, Ministry of Education, Hospital of Stomatology, Faculty of Medical Sciences, Wuhan University, Wuhan, China
| | - Ziyi Chen
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Hua Tang
- Institute of Immunology, Shandong First Medical University, Tai’an, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Qing Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- *Correspondence: Qing Ye, ; Aiping Wu,
| | - Aiping Wu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
- *Correspondence: Qing Ye, ; Aiping Wu,
| |
Collapse
|
33
|
Kirschen GW, Panda S, Burd I. Congenital Infection Influence on Early Brain Development Through the Gut-Brain Axis. Front Neurosci 2022; 16:894955. [PMID: 35844234 PMCID: PMC9280077 DOI: 10.3389/fnins.2022.894955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/23/2022] [Indexed: 11/21/2022] Open
Abstract
The mechanisms by which various pathogens cause congenital infections have been studied extensively, aiding in the understanding of the detrimental effects these infections can have on fetal/neonatal neurological development. Recent studies have focused on the gut-brain axis as pivotal in neurodevelopment, with congenital infections causing substantial disruptions. There remains controversy surrounding the purported sterility of the placenta as well as concerns regarding the effects of exposure to antibiotics used during pregnancy on neonatal microbiome development and how early exposure to microbes or antibiotics can shape the gut-brain axis. Long-term neurodevelopmental consequences, such as autism spectrum disorder, attention deficit hyperactivity disorder, and cerebral palsy, may be attributable, in part, to early life infection and changes in the immature gut microbiome. The goal of this review is thus to critically evaluate the current evidence related to early life infection affecting neurodevelopment through the gut-brain axis.
Collapse
Affiliation(s)
- Gregory W. Kirschen
- Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD, United States
- Integrated Center for Fetal Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Snigdha Panda
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Irina Burd
- Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD, United States
- Integrated Center for Fetal Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
34
|
Casazza RL, Philip DT, Lazear HM. Interferon Lambda Signals in Maternal Tissues to Exert Protective and Pathogenic Effects in a Gestational Stage-Dependent Manner. mBio 2022; 13:e0385721. [PMID: 35471083 PMCID: PMC9239100 DOI: 10.1128/mbio.03857-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/28/2022] [Indexed: 01/10/2023] Open
Abstract
Interferon lambda (IFN-λ) (type III IFN) is constitutively secreted from human placental cells in culture and reduces Zika virus (ZIKV) transplacental transmission in mice. However, the roles of IFN-λ during healthy pregnancy and in restricting congenital infection remain unclear. Here, we used mice lacking the IFN-λ receptor (Ifnlr1-/-) to generate pregnancies lacking either maternal or fetal IFN-λ responsiveness and found that the antiviral effect of IFN-λ resulted from signaling exclusively in maternal tissues. This protective effect depended on gestational stage, as infection earlier in pregnancy (E7 rather than E9) resulted in enhanced transplacental transmission of ZIKV. In Ifnar1-/- dams, which sustain robust ZIKV infection, maternal IFN-λ signaling caused fetal resorption and intrauterine growth restriction. Pregnancy pathology elicited by poly(I·C) treatment also was mediated by maternal IFN-λ signaling, specifically in maternal leukocytes, and also occurred in a gestational stage-dependent manner. These findings identify an unexpected effect of IFN-λ signaling, specifically in maternal (rather than placental or fetal) tissues, which is distinct from the pathogenic effects of IFN-αβ (type I IFN) during pregnancy. These results highlight the complexity of immune signaling at the maternal-fetal interface, where disparate outcomes can result from signaling at different gestational stages. IMPORTANCE Pregnancy is an immunologically complex situation, which must balance protecting the fetus from maternal pathogens with preventing maternal immune rejection of non-self fetal and placental tissue. Cytokines, such as interferon lambda (IFN-λ), contribute to antiviral immunity at the maternal-fetal interface. We found in a mouse model of congenital Zika virus infection that IFN-λ can have either a protective antiviral effect or cause immune-mediated pathology, depending on the stage of gestation when IFN-λ signaling occurs. Remarkably, both the protective and pathogenic effects of IFN-λ occurred through signaling exclusively in maternal immune cells rather than in fetal or placental tissues or in other maternal cell types, identifying a new role for IFN-λ at the maternal-fetal interface.
Collapse
Affiliation(s)
- Rebecca L. Casazza
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Drake T. Philip
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Helen M. Lazear
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
35
|
Aggio JB, Porto BN, Duarte dos Santos CN, Mosimann ALP, Wowk PF. Human Neutrophils Present Mild Activation by Zika Virus But Reduce the Infection of Susceptible Cells. Front Immunol 2022; 13:784443. [PMID: 35747137 PMCID: PMC9210994 DOI: 10.3389/fimmu.2022.784443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
The emergence of the Zika virus (ZIKV) has highlighted the need for a deeper understanding of virus-host interactions in order to pave the way for the development of antiviral therapies. The present work aimed to address the response of neutrophils during ZIKV infection. Neutrophils are important effector cells in innate immunity implicated in the host’s response to neurotropic arboviruses. Our results indicate that human neutrophils were not permissive to Asian or African ZIKV strain replication. In fact, after stimulation with ZIKV, neutrophils were mild primed against the virus as evaluated through CD11b and CD62L modulation, secretion of inflammatory cytokines and granule content, production of reactive oxygen species, and neutrophil extracellular traps formation. Overall, neutrophils did not affect ZIKV infectivity. Moreover, in vitro ZIKV infection of primary innate immune cells did not trigger neutrophil migration. However, neutrophils co-cultured with ZIKV susceptible cell lineages resulted in lower cell infection frequencies, possibly due to cell-to-cell contact. In vivo, neutrophil depletion in immunocompetent mice did not affect ZIKV spreading to the draining lymph nodes. The data suggest that human neutrophils do not play an antiviral role against ZIKV per se, but these cells might participate in an infected environment shaping the ZIKV infection in other target cells.
Collapse
Affiliation(s)
- Juliana Bernardi Aggio
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
| | - Bárbara Nery Porto
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | | | - Ana Luiza Pamplona Mosimann
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
- *Correspondence: Pryscilla Fanini Wowk, ; Ana Luiza Pamplona Mosimann,
| | - Pryscilla Fanini Wowk
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
- *Correspondence: Pryscilla Fanini Wowk, ; Ana Luiza Pamplona Mosimann,
| |
Collapse
|
36
|
Tamura T, Torii S, Kajiwara K, Anzai I, Fujioka Y, Noda K, Taguwa S, Morioka Y, Suzuki R, Fauzyah Y, Ono C, Ohba Y, Okada M, Fukuhara T, Matsuura Y. Secretory glycoprotein NS1 plays a crucial role in the particle formation of flaviviruses. PLoS Pathog 2022; 18:e1010593. [PMID: 35658055 PMCID: PMC9200304 DOI: 10.1371/journal.ppat.1010593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 06/15/2022] [Accepted: 05/13/2022] [Indexed: 11/19/2022] Open
Abstract
Flaviviruses, which are globally distributed and cause a spectrum of potentially severe illnesses, pose a major threat to public health. Although Flaviviridae viruses, including flaviviruses, possess similar genome structures, only the flaviviruses encode the non-structural protein NS1, which resides in the endoplasmic reticulum (ER) and is secreted from cells after oligomerization. The ER-resident NS1 is known to be involved in viral genome replication, but the essential roles of secretory NS1 in the virus life cycle are not fully understood. Here we characterized the roles of secretory NS1 in the particle formation of flaviviruses. We first identified an amino acid residue essential for the NS1 secretion but not for viral genome replication by using protein-protein interaction network analyses and mutagenesis scanning. By using the recombinant flaviviruses carrying the identified NS1 mutation, we clarified that the mutant flaviviruses employed viral genome replication. We then constructed a recombinant NS1 with the identified mutation and demonstrated by physicochemical assays that the mutant NS1 was unable to form a proper oligomer or associate with liposomes. Finally, we showed that the functions of NS1 that were lost by the identified mutation could be compensated for by the in trans-expression of Erns of pestiviruses and host exchangeable apolipoproteins, which participate in the infectious particle formation of pestiviruses and hepaciviruses in the family Flaviviridae, respectively. Collectively, our study suggests that secretory NS1 plays a role in the particle formation of flaviviruses through its interaction with the lipid membrane. It is difficult to characterize the function of NS1 in the post-genome replication stages in the virus life cycle of flaviviruses. Here, by means of protein-protein interaction network analyses and mutagenesis scanning, we identified a unique mutation in NS1 by which the protein loses its secretory capacity while retaining its genome replication activity. Physicochemical assays using the mutant NS1 revealed that oligomerization of NS1 is responsible for the lipid association and secretion of NS1. In addition, we established a complementation assay that can evaluate the particle formation of Flaviviridae viruses. By using recombinant flaviviruses possessing the identified mutation in NS1, we clarified that NS1 is involved in particle formation. Our findings reveal that the flavivirus NS1 has at least two roles in the virus life cycles—namely, a role in infectious particle formation and a role in viral genome replication.
Collapse
Affiliation(s)
- Tomokazu Tamura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shiho Torii
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Kentaro Kajiwara
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Itsuki Anzai
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Hokkaido, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Sapporo, Hokkaido, Japan
| | - Kisho Noda
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shuhei Taguwa
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Yuhei Morioka
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuzy Fauzyah
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Chikako Ono
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Hokkaido, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Sapporo, Hokkaido, Japan
| | - Masato Okada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Takasuke Fukuhara
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- * E-mail: (TF); (YoM)
| | - Yoshiharu Matsuura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- * E-mail: (TF); (YoM)
| |
Collapse
|
37
|
Peng NYG, Amarilla AA, Hugo LE, Modhiran N, Sng JDJ, Slonchak A, Watterson D, Setoh YX, Khromykh AA. The distinguishing NS5-M114V mutation in American Zika virus isolates has negligible impacts on virus replication and transmission potential. PLoS Negl Trop Dis 2022; 16:e0010426. [PMID: 35536870 PMCID: PMC9122223 DOI: 10.1371/journal.pntd.0010426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 05/20/2022] [Accepted: 04/18/2022] [Indexed: 11/18/2022] Open
Abstract
During 2015–2016, outbreaks of Zika virus (ZIKV) occurred in Southeast Asia and the Americas. Most ZIKV infections in humans are asymptomatic, while clinical manifestation is usually a self-limiting febrile disease with maculopapular rash. However, ZIKV is capable of inducing a range of severe neurological complications collectively described as congenital Zika syndrome (CZS). Notably, the scale and magnitude of outbreaks in Southeast Asia were significantly smaller compared to those in the Americas. Sequence comparison between epidemic-associated ZIKV strains from Southeast Asia with those from the Americas revealed a methionine to valine substitution at residue position 114 of the NS5 protein (NS5-M114V) in all the American isolates. Using an American isolate of ZIKV (Natal), we investigated the impact of NS5-M114V mutation on virus replication in cells, virulence in interferon (IFN) α/β receptor knockout (Ifnar-/-) mice, as well as replication and transmission potential in Aedes aegypti mosquitoes. We demonstrated that NS5-M114V mutation had insignificant effect on ZIKV replication efficiency in cells, its ability to degrade STAT2, and virulence in vivo, albeit viremia was slightly prolonged in mice. Furthermore, NS5-M114V mutation decreased mosquito infection and dissemination rates but had no effect on virus secretion into the saliva. Taken together, our findings support the notion that NS5-M114V mutation is unlikely to be a major determinant for virus replication and transmission potential. Zika virus (ZIKV) emerged to cause outbreaks in Southeast Asia and the Americas during 2015–2016. However, the scale of outbreaks in Southeast Asia were significantly smaller compared to epidemic in the Americas. A methionine to valine amino acid mutation at residue position 114 of the NS5 protein (NS5-M114V) is hypothesized to influence the epidemic outcomes of ZIKV, which led to the large-scale epidemic that occurred in the Americas. By analyzing infection of mammalian and mosquito cells, IFNα/β receptor knockout (Ifnar-/-) mice and Aedes aegypti mosquitoes with engineered ZIKV isolates containing either methionine or valine at residue position 114 of the NS5 protein, we demonstrated that the NS5-M114V mutation did not affect virus replication efficiency and STAT2 degradation in cells, virulence in mice, or virus secretion into the mosquito saliva. The NS5-M114V mutation slightly prolonged viremia in Ifnar-/- mice and reduced mosquito infection rate. Collectively, our findings suggest that the NS5-M114V mutation is unlikely to have significantly influenced the ZIKV epidemic in the Americas.
Collapse
Affiliation(s)
- Nias Y. G. Peng
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Alberto A. Amarilla
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Leon E. Hugo
- Mosquito Control Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Queensland, Brisbane, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Julian D. J. Sng
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Andrii Slonchak
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Queensland, Brisbane, Australia
- * E-mail: (DW); (YXS); (AAK)
| | - Yin Xiang Setoh
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
- * E-mail: (DW); (YXS); (AAK)
| | - Alexander A. Khromykh
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Queensland, Brisbane, Australia
- * E-mail: (DW); (YXS); (AAK)
| |
Collapse
|
38
|
Intranasal Immunization with Zika Virus Envelope Domain III-Flagellin Fusion Protein Elicits Systemic and Mucosal Immune Responses and Protection against Subcutaneous and Intravaginal Virus Challenges. Pharmaceutics 2022; 14:pharmaceutics14051014. [PMID: 35631599 PMCID: PMC9144594 DOI: 10.3390/pharmaceutics14051014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022] Open
Abstract
Zika virus (ZIKV) infections in humans are mainly transmitted by the mosquito vectors, but human-to-human sexual transmission is also another important route. Developing a ZIKV mucosal vaccine that can elicit both systemic and mucosal immune responses is of particular interest. In this study, we constructed a recombinant ZIKV envelope DIII (ZDIII) protein genetically fused with Salmonella typhimurium flagellin (FliC-ZDIII) as a novel mucosal antigen for intranasal immunization. The results indicated that the FliC-ZDIII fusion proteins formulated with E. coli heat-labile enterotoxin B subunit (LTIIb-B5) adjuvant greatly increased the ZDIII-specific IgG, IgA, and neutralizing titers in sera, and the ZDIII-specific IgA titers in bronchoalveolar lavage and vaginal fluids. Protective immunity was further assessed by subcutaneous and intravaginal ZIKV challenges. The second-generation FliCΔD3-2ZDIII was shown to result in a reduced titer of anti-FliC IgG antibodies in sera and still retained the same levels of serum IgG, IgA, and neutralizing antibodies and mucosal IgA antibodies without compromising the vaccine antigenicity. Therefore, intranasal immunization with FliCΔD3-2ZDIII fusion proteins formulated with LTIIb-B5 adjuvant elicited the greatest protective immunity against subcutaneous and intravaginal ZIKV challenges. Our findings indicated that the combination of FliCΔD3-2ZDIII fusion proteins and LTIIb-B5 adjuvant for intranasal immunization can be used for developing ZIKV mucosal vaccines.
Collapse
|
39
|
Measles-based Zika vaccine induces long-term immunity and requires NS1 antibodies to protect the female reproductive tract. NPJ Vaccines 2022; 7:43. [PMID: 35440656 PMCID: PMC9018676 DOI: 10.1038/s41541-022-00464-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Zika virus (ZIKV) can cause devastating effects in the unborn fetus of pregnant women. To develop a candidate vaccine that can protect human fetuses, we generated a panel of live measles vaccine (MV) vectors expressing ZIKV-E and -NS1. Our MV-based ZIKV-E vaccine, MV-E2, protected mice from the non-lethal Zika Asian strain (PRVABC59) and the lethal African strain (MR766) challenge. Despite 100% survival of the MV-E2 mice, however, complete viral clearance was not achieved in the brain and reproductive tract of the lethally challenged mice. We then tested MV-based vaccines that expressed E and NS1 together or separately in two different vaccines. We observed complete clearance of ZIKV from the female reproductive tract and complete fetal protection in the lethal African challenge model in animals that received the dual antigen vaccines. Additionally, MV-E2 and MV-NS1, when administered together, induced durable plasma cell responses. Our findings suggest that NS1 antibodies are required to enhance the protection of ZIKV-E antibodies in the female reproductive tract.
Collapse
|
40
|
Lesage S, Chazal M, Beauclair G, Batalie D, Cerboni S, Couderc E, Lescure A, Del Nery E, Tangy F, Martin A, Manel N, Jouvenet N. Discovery of Genes that Modulate Flavivirus Replication in an Interferon-Dependent Manner. J Mol Biol 2022; 434:167277. [PMID: 34599939 PMCID: PMC8480147 DOI: 10.1016/j.jmb.2021.167277] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/02/2022]
Abstract
Establishment of the interferon (IFN)-mediated antiviral state provides a crucial initial line of defense against viral infection. Numerous genes that contribute to this antiviral state remain to be identified. Using a loss-of-function strategy, we screened an original library of 1156 siRNAs targeting 386 individual curated human genes in stimulated microglial cells infected with Zika virus (ZIKV), an emerging RNA virus that belongs to the flavivirus genus. The screen recovered twenty-one potential host proteins that modulate ZIKV replication in an IFN-dependent manner, including the previously known IFITM3 and LY6E. Further characterization contributed to delineate the spectrum of action of these genes towards other pathogenic RNA viruses, including Hepatitis C virus and SARS-CoV-2. Our data revealed that APOL3 acts as a proviral factor for ZIKV and several other related and unrelated RNA viruses. In addition, we showed that MTA2, a chromatin remodeling factor, possesses potent flavivirus-specific antiviral functions induced by IFN. Our work identified previously unrecognized genes that modulate the replication of RNA viruses in an IFN-dependent manner, opening new perspectives to target weakness points in the life cycle of these viruses.
Collapse
Affiliation(s)
- Sarah Lesage
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Virus Sensing and Signaling Unit, F-75015 Paris, France
| | - Maxime Chazal
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Virus Sensing and Signaling Unit, F-75015 Paris, France
| | - Guillaume Beauclair
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Virus Sensing and Signaling Unit, F-75015 Paris, France; Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Damien Batalie
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Molecular Genetics of RNA Viruses Unit, F-75015 Paris, France
| | - Silvia Cerboni
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Elodie Couderc
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Virus Sensing and Signaling Unit, F-75015 Paris, France; Institut Pasteur, Université de Paris, CNRS UMR 2000, Insect-Virus Interactions Unit, F-75015 Paris, France
| | - Aurianne Lescure
- Institut Curie, PSL Research University, Department of Translational Research-Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Elaine Del Nery
- Institut Curie, PSL Research University, Department of Translational Research-Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Frédéric Tangy
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Viral Genomics and Vaccination Unit, F-75015 Paris, France
| | - Annette Martin
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Molecular Genetics of RNA Viruses Unit, F-75015 Paris, France
| | - Nicolas Manel
- Institut Curie, PSL Research University, INSERM U932, Paris, France. https://twitter.com/NicolasManellab
| | - Nolwenn Jouvenet
- Institut Pasteur, Université de Paris, CNRS UMR 3569, Virus Sensing and Signaling Unit, F-75015 Paris, France.
| |
Collapse
|
41
|
Creisher PS, Lei J, Sherer ML, Dziedzic A, Jedlicka AE, Narasimhan H, Chudnovets A, Campbell AD, Liu A, Pekosz A, Burd I, Klein SL. Downregulation of transcriptional activity, increased inflammation, and damage in the placenta following in utero Zika virus infection is associated with adverse pregnancy outcomes. FRONTIERS IN VIROLOGY 2022; 2:782906. [PMID: 35573818 PMCID: PMC9104602 DOI: 10.3389/fviro.2022.782906] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Zika virus (ZIKV) infection during pregnancy causes serious adverse outcomes to the developing fetus, including fetal loss and birth defects known as congenital Zika syndrome (CZS). The mechanism by which ZIKV infection causes these adverse outcomes and specifically, the interplay between the maternal immune response and ZIKV replication has yet to be fully elucidated. Using an immunocompetent mouse model of transplacental ZIKV transmission and adverse pregnancy outcomes, we have previously shown that Asian lineage ZIKV disrupts placental morphology and induces elevated secretion of IL-1β. In the current manuscript, we characterized placental damage and inflammation during in utero African lineage ZIKV infection. Within 48 hours after ZIKV infection at embryonic day 10, viral RNA was detected in placentas and fetuses from ZIKA infected dams, which corresponded with placental damage and reduced fetal viability as compared with mock infected dams. Dams infected with ZIKV had reduced proportions of trophoblasts and endothelial cells and disrupted placental morphology compared to mock infected dams. While placental IL-1β was increased in the placenta, but not the spleen, within 3 hours post infection, this was not caused by activation of the NLRP3 inflammasome. Using bulk mRNAseq from placentas of ZIKV and mock infected dams, ZIKV infection caused profound downregulation of the transcriptional activity of genes that may underly tissue morphology, neurological development, metabolism, cell signaling and inflammation, illustrating that in utero ZIKV infections causes disruption of pathways associated with CZS in our model.
Collapse
Affiliation(s)
- Patrick S. Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Morgan L. Sherer
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Amanda Dziedzic
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anne E. Jedlicka
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Harish Narasimhan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anna Chudnovets
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ariana D. Campbell
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anguo Liu
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Shin M, Kim K, Lee HJ, Lee R, Jung YJ, Park J, Hahn TW. Zika virus baculovirus-expressed envelope protein elicited humoral and cellular immunity in immunocompetent mice. Sci Rep 2022; 12:660. [PMID: 35027643 PMCID: PMC8758750 DOI: 10.1038/s41598-021-04713-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/24/2021] [Indexed: 12/05/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that has a high risk of inducing Guillain-Barré syndrome and microcephaly in newborns. Because vaccination is considered the most effective strategy against ZIKV infection, we designed a recombinant vaccine utilizing the baculovirus expression system with two strains of ZIKV envelope protein (MR766, Env_M; ZBRX6, Env_Z). Animals inoculated with Env_M and Env_Z produced ZIKV-specific antibodies and secreted effector cytokines such as interferon-γ, tumor necrosis factor-α, and interleukin-12. Moreover, the progeny of immunized females had detectable maternal antibodies that protected them against two ZIKV strains (MR766 and PRVABC59) and a Dengue virus strain. We propose that the baculovirus expression system ZIKV envelope protein recombinant provides a safe and effective vaccine strategy.
Collapse
Affiliation(s)
- Minna Shin
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Kiju Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyo-Ji Lee
- College of Biological Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rangyeon Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Yu-Jin Jung
- College of Biological Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jeongho Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Tae-Wook Hahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
43
|
Vue D, Tang Q. Zika Virus Overview: Transmission, Origin, Pathogenesis, Animal Model and Diagnosis. ZOONOSES (BURLINGTON, MASS.) 2021; 1:10.15212/zoonoses-2021-0017. [PMID: 34957474 PMCID: PMC8698461 DOI: 10.15212/zoonoses-2021-0017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Zika virus (ZIKV) was first discovered in 1947 in Uganda. ZIKV did not entice much attention until Brazil hosted the 2016 Summer Olympics Game, where ZIKV attracted a global audience. ZIKV is a flavivirus that can be transmitted chiefly through the biting of the mosquito or sexually or by breastfeeding at a lower scale. As time passed, the recent discovery of how the ZIKV causes congenital neurodevelopmental defects, including microcephaly, makes us reevaluate the importance of ZIKV interaction with centrosome organization because centrosome plays an important role in cell division. When the ZIKV disrupts centrosome organization and mitotic abnormalities, this will alter neural progenitor differentiation. Altering the neural progenitor differentiation will lead to cell cycle arrest, increase apoptosis, and inhibit the neural progenitor cell differentiation, as this can lead to abnormalities in neural cell development resulting in microcephaly. Understanding the importance of ZIKV infection throughout the years, this review article gives an overview of the history, transmission routes, pathogenesis, animal models, and diagnosis.
Collapse
Affiliation(s)
- Dallas Vue
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW Washington, DC 20059, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW Washington, DC 20059, USA
| |
Collapse
|
44
|
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
|
45
|
van Leur SW, Heunis T, Munnur D, Sanyal S. Pathogenesis and virulence of flavivirus infections. Virulence 2021; 12:2814-2838. [PMID: 34696709 PMCID: PMC8632085 DOI: 10.1080/21505594.2021.1996059] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 11/01/2022] Open
Abstract
The Flavivirus genus consists of >70 members including several that are considered significant human pathogens. Flaviviruses display a broad spectrum of diseases that can be roughly categorised into two phenotypes - systemic disease involving haemorrhage exemplified by dengue and yellow Fever virus, and neurological complications associated with the likes of West Nile and Zika viruses. Attempts to develop vaccines have been variably successful against some. Besides, mosquito-borne flaviviruses can be vertically transmitted in the arthropods, enabling long term persistence and the possibility of re-emergence. Therefore, developing strategies to combat disease is imperative even if vaccines become available. The cellular interactions of flaviviruses with their human hosts are key to establishing the viral lifecycle on the one hand, and activation of host immunity on the other. The latter should ideally eradicate infection, but often leads to immunopathological and neurological consequences. In this review, we use Dengue and Zika viruses to discuss what we have learned about the cellular and molecular determinants of the viral lifecycle and the accompanying immunopathology, while highlighting current knowledge gaps which need to be addressed in future studies.
Collapse
Affiliation(s)
| | - Tiaan Heunis
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OxfordOX1 3RE, UK
| | - Deeksha Munnur
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OxfordOX1 3RE, UK
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OxfordOX1 3RE, UK
| |
Collapse
|
46
|
Are Zika virus cross-reactive antibodies against aquaporin-4 associated to Neuromyelitis Optica Spectrum Disorder? J Neuroimmunol 2021; 360:577697. [PMID: 34461359 DOI: 10.1016/j.jneuroim.2021.577697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/03/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022]
Abstract
Zika virus (ZIKV) infection has been associated with the development of Neuromyelitis Optica Spectrum Disorder (NMOSD). ZIKV-induced antibodies that putatively cross-react to aquaporin-4 (AQP4) protein are suggested to cause inflammation of the optic nerve. A region of similarity between AQP4 and the ZIKV NS2B protein was identified. Our data showed that ZIKV-associated NMOSD patients develop anti-AQP4 antibodies, but not anti-ZIKV NS2B antibodies, revealing that cross-reacting antibodies are not the underlying cause of this phenotype. ZIKV infection in mice showed persistent viral replication in the eye tissue, suggesting that NMOSD symptoms are consequence of viral infection of the optic nerve cells.
Collapse
|
47
|
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
|
48
|
Zaccaria G, Malatesta D, Jurisic L, Marcacci M, Di Teodoro G, Conte A, Teodori L, Monaco F, Marini V, Casaccia C, Savini G, Di Gennaro A, Rossi E, D'Innocenzo V, D'Alterio N, Lorusso A. The envelope protein of Usutu virus attenuates West Nile virus virulence in immunocompetent mice. Vet Microbiol 2021; 263:109262. [PMID: 34715462 DOI: 10.1016/j.vetmic.2021.109262] [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: 08/14/2020] [Accepted: 10/17/2021] [Indexed: 11/28/2022]
Abstract
West Nile virus (WNV) and Usutu virus (USUV) are the two most widespread mosquito-borne flaviviruses in Europe causing severe neuroinvasive disease in humans. Here, following standardization of the murine model with wild type (wt) viruses, we engineered WNV and USUV genome by reverse genetics. A recombinant virus carrying the 5' UTR of WNV within the USUV genome backbone (r-USUV5'-UTR WNV) was rescued; when administered to mice this virus did not cause signs or disease as wt USUV suggesting that 5' UTR of a marked neurotropic parental WNV was not per se a virulence factor. Interestingly, a chimeric virus carrying the envelope (E) protein of USUV in the WNV genome backbone (r-WNVE-USUV) showed an attenuated profile in mice compared to wt WNV but significantly more virulent than wt USUV. Moreover, except when tested against serum samples originating from a live WNV infection, r-WNVE-USUV showed an identical antigenic profile to wt USUV confirming that E is also the major immunodominant protein of USUV.
Collapse
Affiliation(s)
- Guendalina Zaccaria
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Daniela Malatesta
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Lucija Jurisic
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Maurilia Marcacci
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy; Dipartimento di Medicina Veterinaria, University of Bari, Valenzano, Bari, Italy
| | - Giovanni Di Teodoro
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Annamaria Conte
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Liana Teodori
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Federica Monaco
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Valeria Marini
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Claudia Casaccia
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Giovanni Savini
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Annapia Di Gennaro
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Emanuela Rossi
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Vincenzo D'Innocenzo
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Nicola D'Alterio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy
| | - Alessio Lorusso
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise (IZSAM), Campo Boario, 64100 Teramo, Italy.
| |
Collapse
|
49
|
The Compound SBI-0090799 Inhibits Zika Virus Infection by Blocking De Novo Formation of the Membranous Replication Compartment. J Virol 2021; 95:e0099621. [PMID: 34468177 DOI: 10.1128/jvi.00996-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen classified by the World Health Organization (WHO) as a public health emergency of international concern in 2016, and it is still identified as a priority disease. Although most infected individuals are asymptomatic or show mild symptoms, a risk of neurologic complications is associated with infection in adults. Additionally, infection during pregnancy is directly linked to microcephaly and other congenital malformations. Since there are no currently available vaccines or approved therapeutics for this virus, there is a critical unmet need in developing treatments to prevent future ZIKV outbreaks. Toward this end, we performed a large-scale cell-based high-content screen of 51,520 chemical compounds to identify potential antiviral drug candidates. The compound (2E)-N-benzyl-3-(4-butoxyphenyl)prop-2-enamide (SBI-0090799) was found to inhibit replication of multiple ZIKV strains and in different cell systems. SBI-0090799 did not affect viral entry or RNA translation but suppressed RNA replication by preventing the formation of the membranous replication compartment. Selection of drug-resistant viruses identified single-amino-acid substitutions in the N-terminal region of nonstructural protein NS4A, arguing this is the likely drug target. These resistance mutations rescued viral RNA replication and restored the formation of the membranous replication compartment. This mechanism of action is similar to clinically approved NS5A inhibitors for hepatitis C virus (HCV). Taken together, SBI-0090799 represents a promising lead candidate for the development of an antiviral treatment against ZIKV infection for the mitigation of severe complications and potential resurgent outbreaks of the virus. IMPORTANCE This study describes the elucidation of (2E)-N-benzyl-3-(4-butoxyphenyl)prop-2-enamide (SBI-0090799) as a selective and potent inhibitor of Zika virus (ZIKV) replication using a high-throughput screening approach. Mapping and resistance studies, supported by electron microscopy observations, indicate that the small molecule is functioning through inhibition of NS4A-mediated formation of ZIKV replication compartments in the endoplasmic reticulum (ER). Intriguingly, this defines a novel nonenzymatic target and chemical matter for the development of a new class of ZIKV antivirals. Moreover, chemical modulation affecting this nonstructural protein mirrors the identification and development of hepatitis C virus (HCV) NS5A inhibitor daclatasvir and its derivatives, similarly interfering with the formation of the viral replication compartment and also targeting a protein with no enzymatic activity, which have been part of a curative strategy for HCV.
Collapse
|
50
|
Are the Organoid Models an Invaluable Contribution to ZIKA Virus Research? Pathogens 2021; 10:pathogens10101233. [PMID: 34684182 PMCID: PMC8537471 DOI: 10.3390/pathogens10101233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
In order to prevent new pathogen outbreaks and avoid possible new global health threats, it is important to study the mechanisms of microbial pathogenesis, screen new antiviral agents and test new vaccines using the best methods. In the last decade, organoids have provided a groundbreaking opportunity for modeling pathogen infections in human brains, including Zika virus (ZIKV) infection. ZIKV is a member of the Flavivirus genus, and it is recognized as an emerging infectious agent and a serious threat to global health. Organoids are 3D complex cellular models that offer an in-scale organ that is physiologically alike to the original one, useful for exploring the mechanisms behind pathogens infection; additionally, organoids integrate data generated in vitro with traditional tools and often support those obtained in vivo with animal model. In this mini-review the value of organoids for ZIKV research is examined and sustained by the most recent literature. Within a 3D viewpoint, tissue engineered models are proposed as future biological systems to help in deciphering pathogenic processes and evaluate preventive and therapeutic strategies against ZIKV. The next steps in this field constitute a challenge that may protect people and future generations from severe brain defects.
Collapse
|