1
|
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 DOI: 10.1038/s41467-024-55155-4] [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: 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
|
2
|
Hu Y, Hu Y, Yin A, Lv Y, Li J, Fan J, Qian B, Song J, Zhang Y. IP-10 acts early in CV-A16 infection to induce BBB destruction and promote virus entry into the CNS by increasing TNF-α expression. Front Immunol 2024; 15:1374447. [PMID: 39559356 PMCID: PMC11570546 DOI: 10.3389/fimmu.2024.1374447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024] Open
Abstract
The mechanisms underlying pathological changes in the central nervous system (CNS) following Coxsackievirus A16 (CV-A16) infection have not yet been elucidated. IFN-γ-inducible protein-10 (IP-10) is often used as a predictive factor to monitor early virus infection. It has also been reported that IP-10 plays a pivotal role in neuroinflammation. In this study, we aimed to explore the role of IP-10 in the neuropathogenesis of CV-A16 infection. We observed that the level of IP-10, as well as the TLR3-TRIF-TRAF3-TBK1-NF-κB and RIG-I/MDA5-MAVS-TRAFS-TBK1-NF-κB pathways, which are the upstream of IP-10, were significantly elevated during the course of CV-A16 infection. This increase was accompanied by an increase in a series of inflammatory cytokines at different time-points during CV-A16 infection. To determine whether IP-10 influences BBB integrity, we examined junctional complexes. Our results revealed that the expression levels of Claudin5, Occludin, ZO-1 and VE-Cadherin were notably decreased in CV-A16-infected HUVECs, but these indicators were restored in CV-A16-infected HUVECs with Eldelumab treatment. Nevertheless, IP-10 is only a chemokine that primarily traffics CXCR3-positive immune cells to inflammatory sites or promotes the production of inflammatory cytokines. Therefore, the interactions between IP-10 and inflammatory cytokines were evaluated. Our data revealed that IP-10 mediated the production of TNF-α, which was also observed to change the junctional complexes. Moreover, in a suckling mouse model, IP-10 and TNF-α treatments exacerbated clinical symptoms, mortality and pathological changes in the brain of CV-A16-infected mice, but Anti-IP-10 and Anti-TNF-α treatments alleviated these changes. Our data also revealed that IP-10 may be detected early in CV-A16 infection, whereas TNF-α was detected late in CV-A16 infection, and the production of TNF-α was also found to be positively correlated with IP-10. In addition, IP-10 and TNF-α were observed to reduce junctional complexes and enhance virus entry into the CNS. Taken together, this study provides the first evidence that CV-A16 activates the IP-10/TNF-α regulatory axis to cause BBB damage and accelerate the formation of neuroinflammation in infected hosts, which not only provides a new understanding of the neuropathogenesis caused by CV-A16, but also offers a promising target for the development of CV-A16 antiviral drugs.
Collapse
Affiliation(s)
- Yajie Hu
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yunguang Hu
- National and Local Engineering Center for Infectious Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Anguo Yin
- National and Local Engineering Center for Infectious Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Yaming Lv
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiang Li
- National and Local Engineering Center for Infectious Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Jingyuan Fan
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Baojiang Qian
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jie Song
- National and Local Engineering Center for Infectious Biological Products, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Yunhui Zhang
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
3
|
Metzler AD, Tang H. Zika Virus Neuropathogenesis-Research and Understanding. Pathogens 2024; 13:555. [PMID: 39057782 PMCID: PMC11279898 DOI: 10.3390/pathogens13070555] [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: 05/02/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, is prominently associated with microcephaly in babies born to infected mothers as well as Guillain-Barré Syndrome in adults. Each cell type infected by ZIKV-neuronal cells (radial glial cells, neuronal progenitor cells, astrocytes, microglia cells, and glioblastoma stem cells) and non-neuronal cells (primary fibroblasts, epidermal keratinocytes, dendritic cells, monocytes, macrophages, and Sertoli cells)-displays its own characteristic changes to their cell physiology and has various impacts on disease. Here, we provide an in-depth review of the ZIKV life cycle and its cellular targets, and discuss the current knowledge of how infections cause neuropathologies, as well as what approaches researchers are currently taking to further advance such knowledge. A key aspect of ZIKV neuropathogenesis is virus-induced neuronal apoptosis via numerous mechanisms including cell cycle dysregulation, mitochondrial fragmentation, ER stress, and the unfolded protein response. These, in turn, result in the activation of p53-mediated intrinsic cell death pathways. A full spectrum of infection models including stem cells and co-cultures, transwells to simulate blood-tissue barriers, brain-region-specific organoids, and animal models have been developed for ZIKV research.
Collapse
Affiliation(s)
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
4
|
Ohki CMY, Benazzato C, van der Linden V, França JV, Toledo CM, Machado RRG, Araujo DB, Oliveira DBL, Neris RS, Assunção-Miranda I, de Oliveira Souza IN, Nogueira CO, Leite PEC, van der Linden H, Figueiredo CP, Durigon EL, Clarke JR, Russo FB, Beltrão-Braga PCB. Zika virus infection impairs synaptogenesis, induces neuroinflammation, and could be an environmental risk factor for autism spectrum disorder outcome. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167097. [PMID: 38408544 DOI: 10.1016/j.bbadis.2024.167097] [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: 08/14/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Zika virus (ZIKV) infection was first associated with Central Nervous System (CNS) infections in Brazil in 2015, correlated with an increased number of newborns with microcephaly, which ended up characterizing the Congenital Zika Syndrome (CZS). Here, we investigated the impact of ZIKV infection on the functionality of iPSC-derived astrocytes. Besides, we extrapolated our findings to a Brazilian cohort of 136 CZS children and validated our results using a mouse model. Interestingly, ZIKV infection in neuroprogenitor cells compromises cell migration and causes apoptosis but does not interfere in astrocyte generation. Moreover, infected astrocytes lost their ability to uptake glutamate while expressing more glutamate transporters and secreted higher levels of IL-6. Besides, infected astrocytes secreted factors that impaired neuronal synaptogenesis. Since these biological endophenotypes were already related to Autism Spectrum Disorder (ASD), we extrapolated these results to a cohort of children, now 6-7 years old, and found seven children with ASD diagnosis (5.14 %). Additionally, mice infected by ZIKV revealed autistic-like behaviors, with a significant increase of IL-6 mRNA levels in the brain. Considering these evidence, we inferred that ZIKV infection during pregnancy might lead to synaptogenesis impairment and neuroinflammation, which could increase the risk for ASD.
Collapse
Affiliation(s)
| | - Cecília Benazzato
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Julia V França
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carmen M Toledo
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Romulo S Neris
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iranaia Assunção-Miranda
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Clara O Nogueira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Emilio Corrêa Leite
- Clinical Research Unit of the Antonio Pedro Hospital, Federal Fluminense University, Rio de Janeiro, Brazil
| | | | - Claudia P Figueiredo
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edison Luiz Durigon
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Institut Pasteur de São Paulo, São Paulo, Brazil
| | - Julia R Clarke
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
5
|
de Lima Cavalcanti TYV, Lima MC, Bargi-Souza P, Franca RFO, Peliciari-Garcia RA. Zika Virus Infection Alters the Circadian Clock Expression in Human Neuronal Monolayer and Neurosphere Cultures. Cell Mol Neurobiol 2023; 44:10. [PMID: 38141078 PMCID: PMC11407173 DOI: 10.1007/s10571-023-01445-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023]
Abstract
Rhythmic regulations are virtually described in all physiological processes, including central nervous system development and immunologic responses. Zika virus (ZIKV), a neurotropic arbovirus, has been recently linked to a series of birth defects and neurodevelopmental disorders. Given the well-characterized role of the intrinsic cellular circadian clock within neurogenesis, cellular metabolism, migration, and differentiation among other processes, this study aimed to characterize the influence of ZIKV infection in the circadian clock expression in human neuronal cells. For this, in vitro models of human-induced neuroprogenitor cells (hiNPCs) and neuroblastoma cell line SH-SY5Y, cultured as monolayer and neurospheres, were infected by ZIKV, followed by RNA-Seq and RT-qPCR investigation, respectively. Targeted circadian clock components presented mRNA oscillations only after exogenous synchronizing stimuli (Forskolin) in SH-SY5Y monolayer culture. Interestingly, when these cells were grown as 3D-arranged neurospheres, an intrinsic oscillatory expression pattern was observed for some core clock components without any exogenous stimulation. The ZIKV infection significantly disturbed the mRNA expression pattern of core clock components in both neuroblastoma cell culture models, which was also observed in hiNPCs infected with different strains of ZIKV. The ZIKV-mediated desynchronization of the circadian clock expression in human cells might further contribute to the virus impairment of neuronal metabolism and function observed in adults and ZIKV-induced congenital syndrome. In vitro models of Zika virus (ZIKV) neuronal infection. Human neuroprogenitor cells were cultured as monolayer and neurospheres and infected by ZIKV. Monolayer-cultured cells received forskolin (FSK) as a coupling factor for the circadian clock rhythmicity, while 3D-arranged neurospheres showed an intrinsic oscillatory pattern in the circadian clock expression. The ZIKV infection affected the mRNA expression pattern of core clock components in both cell culture models. The ZIKV-mediated desynchronization of the circadian clock machinery might contribute to the impairment of neuronal metabolism and function observed in both adults (e.g., Guillain-Barré syndrome) and ZIKV-induced congenital syndrome (microcephaly). The graphical abstract has been created with Canva at the canva.com website.
Collapse
Affiliation(s)
- Thaíse Yasmine Vasconcelos de Lima Cavalcanti
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation - FIOCRUZ, Av. Professor Moraes Rego, S/N, Cidade Universitária, Recife, PE, CEP 50740-465, Brazil
| | - Morganna Costa Lima
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation - FIOCRUZ, Av. Professor Moraes Rego, S/N, Cidade Universitária, Recife, PE, CEP 50740-465, Brazil
| | - Paula Bargi-Souza
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Rafael Freitas Oliveira Franca
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation - FIOCRUZ, Av. Professor Moraes Rego, S/N, Cidade Universitária, Recife, PE, CEP 50740-465, Brazil.
| | - Rodrigo Antonio Peliciari-Garcia
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation - FIOCRUZ, Av. Professor Moraes Rego, S/N, Cidade Universitária, Recife, PE, CEP 50740-465, Brazil.
- Morphophysiology & Pathology Sector, Department of Biological Sciences, Federal University of São Paulo, Rua São Nicolau, 210, Diadema, SP, CEP 09913-030, Brazil.
| |
Collapse
|
6
|
Verkhratsky A, Butt A, Li B, Illes P, Zorec R, Semyanov A, Tang Y, Sofroniew MV. Astrocytes in human central nervous system diseases: a frontier for new therapies. Signal Transduct Target Ther 2023; 8:396. [PMID: 37828019 PMCID: PMC10570367 DOI: 10.1038/s41392-023-01628-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 10/14/2023] Open
Abstract
Astroglia are a broad class of neural parenchymal cells primarily dedicated to homoeostasis and defence of the central nervous system (CNS). Astroglia contribute to the pathophysiology of all neurological and neuropsychiatric disorders in ways that can be either beneficial or detrimental to disorder outcome. Pathophysiological changes in astroglia can be primary or secondary and can result in gain or loss of functions. Astroglia respond to external, non-cell autonomous signals associated with any form of CNS pathology by undergoing complex and variable changes in their structure, molecular expression, and function. In addition, internally driven, cell autonomous changes of astroglial innate properties can lead to CNS pathologies. Astroglial pathophysiology is complex, with different pathophysiological cell states and cell phenotypes that are context-specific and vary with disorder, disorder-stage, comorbidities, age, and sex. Here, we classify astroglial pathophysiology into (i) reactive astrogliosis, (ii) astroglial atrophy with loss of function, (iii) astroglial degeneration and death, and (iv) astrocytopathies characterised by aberrant forms that drive disease. We review astroglial pathophysiology across the spectrum of human CNS diseases and disorders, including neurotrauma, stroke, neuroinfection, autoimmune attack and epilepsy, as well as neurodevelopmental, neurodegenerative, metabolic and neuropsychiatric disorders. Characterising cellular and molecular mechanisms of astroglial pathophysiology represents a new frontier to identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania.
| | - Arthur Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04109, Leipzig, Germany
| | - Robert Zorec
- Celica Biomedical, Lab Cell Engineering, Technology Park, 1000, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Alexey Semyanov
- Department of Physiology, Jiaxing University College of Medicine, 314033, Jiaxing, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Nogueira CO, Rocha T, Messor DF, Souza INO, Clarke JR. Fundamental neurochemistry review: Glutamatergic dysfunction as a central mechanism underlying flavivirus-induced neurological damage. J Neurochem 2023; 166:915-927. [PMID: 37603368 DOI: 10.1111/jnc.15935] [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: 05/15/2023] [Revised: 07/06/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
The Flaviviridae family comprises positive-sense single-strand RNA viruses mainly transmitted by arthropods. Many of these pathogens are especially deleterious to the nervous system, and a myriad of neurological symptoms have been associated with infections by Zika virus (ZIKV), West Nile virus (WNV), and Japanese encephalitis virus (JEV) in humans. Studies suggest that viral replication in neural cells and the massive release of pro-inflammatory mediators lead to morphological alterations of synaptic spine structure and changes in the balance of excitatory/inhibitory neurotransmitters and receptors. Glutamate is the predominant excitatory neurotransmitter in the brain, and studies propose that either enhanced release or impaired uptake of this amino acid contributes to brain damage in several conditions. Here, we review existing evidence suggesting that glutamatergic dysfunction-induced by flaviviruses is a central mechanism for neurological damage and clinical outcomes of infection. We also discuss current data suggesting that pharmacological approaches that counteract glutamatergic dysfunction show benefits in animal models of such viral diseases.
Collapse
Affiliation(s)
- Clara O Nogueira
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tamires Rocha
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel F Messor
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isis N O Souza
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia R Clarke
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Rengifo AC, Rivera J, Álvarez-Díaz DA, Naizaque J, Santamaria G, Corchuelo S, Gómez CY, Torres-Fernández O. Morphological and Molecular Changes in the Cortex and Cerebellum of Immunocompetent Mice Infected with Zika Virus. Viruses 2023; 15:1632. [PMID: 37631975 PMCID: PMC10458311 DOI: 10.3390/v15081632] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Zika virus (ZIKV) disease continues to be a threat to public health, and it is estimated that millions of people have been infected and that there have been more cases of serious complications than those already reported. Despite many studies on the pathogenesis of ZIKV, several of the genes involved in the malformations associated with viral infection are still unknown. In this work, the morphological and molecular changes in the cortex and cerebellum of mice infected with ZIKV were evaluated. Neonatal BALB/c mice were inoculated with ZIKV intraperitoneally, and the respective controls were inoculated with a solution devoid of the virus. At day 10 postinoculation, the mice were euthanized to measure the expression of the markers involved in cortical and cerebellar neurodevelopment. The infected mice presented morphological changes accompanied by calcifications, as well as a decrease in most of the markers evaluated in the cortex and cerebellum. The modifications found could be predictive of astrocytosis, dendritic pathology, alterations in the regulation systems of neuronal excitation and inhibition, and premature maturation, conditions previously described in other models of ZIKV infection and microcephaly.
Collapse
Affiliation(s)
- Aura Caterine Rengifo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Jorge Rivera
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Diego Alejandro Álvarez-Díaz
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
- Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia
| | - Julián Naizaque
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Gerardo Santamaria
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Sheryll Corchuelo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Claudia Yadira Gómez
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| |
Collapse
|
9
|
Yu X, Wang S, Wu W, Chang H, Shan P, Yang L, Zhang W, Wang X. Exploring New Mechanism of Depression from the Effects of Virus on Nerve Cells. Cells 2023; 12:1767. [PMID: 37443801 PMCID: PMC10340315 DOI: 10.3390/cells12131767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Depression is a common neuropsychiatric disorder with long-term recurrent depressed mood, pain and despair, pessimism and anxiety, and even suicidal tendencies as the main symptoms. Depression usually induces or aggravates the development of other related diseases, such as sleep disorders and endocrine disorders. In today's society, the incidence of depression is increasing worldwide, and its pathogenesis is complex and generally believed to be related to genetic, psychological, environmental, and biological factors. Current studies have shown the key role of glial cells in the development of depression, and it is noteworthy that some recent evidence suggests that the development of depression may be closely related to viral infections, such as SARS-CoV-2, BoDV-1, ZIKV, HIV, and HHV6, which infect the organism and cause some degree of glial cells, such as astrocytes, oligodendrocytes, and microglia. This can affect the transmission of related proteins, neurotransmitters, and cytokines, which in turn leads to neuroinflammation and depression. Based on the close relationship between viruses and depression, this paper provides an in-depth analysis of the new mechanism of virus-induced depression, which is expected to provide a new perspective on the mechanism of depression and a new idea for the diagnosis of depression in the future.
Collapse
Affiliation(s)
- Xinxin Yu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (X.Y.); (W.W.)
| | - Shihao Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (S.W.); (H.C.); (W.Z.)
| | - Wenzheng Wu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (X.Y.); (W.W.)
| | - Hongyuan Chang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (S.W.); (H.C.); (W.Z.)
| | - Pufan Shan
- College of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Lin Yang
- College of Nursing, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Wenjie Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (S.W.); (H.C.); (W.Z.)
| | - Xiaoyu Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (X.Y.); (W.W.)
| |
Collapse
|
10
|
Viettri M, Caraballo G, Sanchez ME, Espejel-Nuñez A, Betanzos A, Ortiz-Navarrete V, Estrada-Gutierrez G, Nava P, Ludert JE. Comparative Infections of Zika, Dengue, and Yellow Fever Viruses in Human Cytotrophoblast-Derived Cells Suggest a Gating Role for the Cytotrophoblast in Zika Virus Placental Invasion. Microbiol Spectr 2023; 11:e0063023. [PMID: 37227282 PMCID: PMC10269719 DOI: 10.1128/spectrum.00630-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/01/2023] [Indexed: 05/26/2023] Open
Abstract
The Zika virus (ZIKV) is teratogenic and considered a TORCH pathogen (toxoplasmosis [Toxoplasma gondii], rubella, cytomegalovirus, herpes simplex virus [HSV], and other microorganisms capable of crossing the blood-placenta barrier). In contrast, the related flavivirus dengue virus (DENV) and the attenuated yellow fever virus vaccine strain (YFV-17D) are not. Understanding the mechanisms used by ZIKV to cross the placenta is necessary. In this work, parallel infections with ZIKV of African and Asian lineages, DENV, and YFV-17D were compared for kinetics and growth efficiency, activation of mTOR pathways, and cytokine secretion profile using cytotrophoblast-derived HTR8 cells and monocytic U937 cells differentiated to M2 macrophages. In HTR8 cells, ZIKV replication, especially the African strain, was significantly more efficient and faster than DENV or YFV-17D. In macrophages, ZIKV replication was also more efficient, although differences between strains were reduced. Greater activation of the mTORC1 and mTORC2 pathways in HTR8 cells infected with ZIKV than with DENV or YFV-17D was observed. HTR8 cells treated with mTOR inhibitors showed a 20-fold reduction in ZIKV yield, versus 5- and 3.5-fold reductions for DENV and YFV-17D, respectively. Finally, infection with ZIKV, but not DENV or YFV-17D, efficiently inhibited the interferon (IFN) and chemoattractant responses in both cell lines. These results suggest a gating role for the cytotrophoblast cells in favoring entry of ZIKV, but not DENV and YFV-17D, into the placental stroma. IMPORTANCE Zika virus acquisition during pregnancy is associated with severe fetal damage. The Zika virus is related to dengue virus and yellow fever virus, yet fetal damage has not been related to dengue or inadvertent vaccination for yellow fever during pregnancy. Mechanisms used by the Zika virus to cross the placenta need to be deciphered. By comparing parallel infections of Zika virus strains belonging to the African and Asian lineages, dengue virus, and the yellow fever vaccine virus strain YFV-17D in placenta-derived cytotrophoblast cells and differentiated macrophages, evidence was found that Zika virus infections, especially by the African strains, were more efficient in cytotrophoblast cells than dengue virus or yellow fever vaccine virus strain infections. Meanwhile, no significant differences were observed in macrophages. Robust activation of the mTOR signaling pathways and inhibition of the IFN and chemoattractant response appear to be related to the better growth capacity of the Zika viruses in the cytotrophoblast-derived cells.
Collapse
Affiliation(s)
- Mercedes Viettri
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Gerson Caraballo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Maria Elena Sanchez
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Abigail Betanzos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | | | - Porfirio Nava
- Department of Biophysical Physiology and Neuroscience, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan E. Ludert
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
11
|
Du C, Li G, Han G. Biosafety and mental health: Virus induced cognitive decline. BIOSAFETY AND HEALTH 2023; 5:159-167. [PMID: 40078510 PMCID: PMC11895046 DOI: 10.1016/j.bsheal.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 03/14/2025] Open
Abstract
Biological agents threats people's life through different ways, one of which lies in the impairment of cognition. It is believed cognitive decline may result from biological agents mediated neuron damage directly, or from the activation of the host immune response to eradicate the pathogen. However, direct linkage between infections and cognitive decline is very limited. Here we focus on the mechanisms of how different biological virus or they induced systemic and local inflammation link to the cognitive impairment, focusing on the roles of activated microglia and several molecular pathways mediated neurotoxicity.
Collapse
Affiliation(s)
- Chunxiao Du
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ge Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Gencheng Han
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| |
Collapse
|
12
|
da Silva EV, Fontes-Dantas FL, Dantas TV, Dutra A, Nascimento OJM, Alves-Leon SV. Shared Molecular Signatures Across Zika Virus Infection and Multiple Sclerosis Highlight AP-1 Transcription Factor as a Potential Player in Post-ZIKV MS-Like Phenotypes. Mol Neurobiol 2023:10.1007/s12035-023-03305-y. [PMID: 37046138 DOI: 10.1007/s12035-023-03305-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/08/2023] [Indexed: 04/14/2023]
Abstract
Zika virus (ZIKV) is an arbovirus of the Flaviviridae genus that has rapidly disseminated from across the Pacific to the Americas. Robust evidence has indicated a crucial role of ZIKV in congenital virus syndrome, including neonatal microcephaly. Moreover, emerging evidence suggests an association between ZIKV infection and the development of an extensive spectrum of central nervous system inflammatory demyelinating diseases (CNS IDD), such as multiple sclerosis-like clinical phenotypes. However, the underlying mechanisms of host-pathogen neuro-immune interactions remain to be elucidated. This study aimed to identify common transcriptional signatures between multiple sclerosis (MS) and ZIKV infection to generate molecular interaction networks, thereby leading to the identification of deregulated processes and pathways, which could give an insight of these underlying molecular mechanisms. Our investigation included publicly available transcriptomic data from MS patients in either relapse or remission (RR-MS) and datasets of subjects acutely infected by ZIKV for both immune peripheral cells and central nervous system cells. The protein-protein interaction (PPI) analysis showed upregulated AP-1 transcription factors (JUN and FOS) among the top hub and bottleneck genes in RR-MS and ZIKV data. Gene enrichment analysis retrieved a remarkable presence of ontologies and pathways linked to oxidative stress responses, immune cell function, inflammation, interleukin signaling, cell division, and transcriptional regulation commonly enriched in both scenarios. Considering the recent findings concerning AP-1 function in immunological tolerance breakdown, regulation of inflammation, and its function as an oxidative stress sensor, we postulate that the ZIKV trigger may contribute as a boost for the activation of such AP-1-regulated mechanisms that could favor the development of MS-like phenotypes following ZIKV infection in a genetically susceptible individual.
Collapse
Affiliation(s)
- Elielson Veloso da Silva
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de pós-graduação em Medicina (Neurologia/Neurociências), Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Fabrícia Lima Fontes-Dantas
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thiago Viana Dantas
- Programa de Engenharia de Sistemas e Computação-COPPE, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda Dutra
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Osvaldo J M Nascimento
- Programa de pós-graduação em Medicina (Neurologia/Neurociências), Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Soniza Vieira Alves-Leon
- Laboratório de Neurociências Translacional, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.
- Hospital Universitário Clementino Fraga Filho, Centro de Referência em Doenças Inflamatórias Desmielinizantes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Quincozes-Santos A, Bobermin LD, Costa NLF, Thomaz NK, Almeida RRDS, Beys-da-Silva WO, Santi L, Rosa RL, Capra D, Coelho-Aguiar JM, DosSantos MF, Heringer M, Cirne-Lima EO, Guimarães JA, Schuler-Faccini L, Gonçalves CA, Moura-Neto V, Souza DO. The role of glial cells in Zika virus-induced neurodegeneration. Glia 2023. [PMID: 36866453 DOI: 10.1002/glia.24353] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Zika virus (ZIKV) is a strongly neurotropic flavivirus whose infection has been associated with microcephaly in neonates. However, clinical and experimental evidence indicate that ZIKV also affects the adult nervous system. In this regard, in vitro and in vivo studies have shown the ability of ZIKV to infect glial cells. In the central nervous system (CNS), glial cells are represented by astrocytes, microglia, and oligodendrocytes. In contrast, the peripheral nervous system (PNS) constitutes a highly heterogeneous group of cells (Schwann cells, satellite glial cells, and enteric glial cells) spread through the body. These cells are critical in both physiological and pathological conditions; as such, ZIKV-induced glial dysfunctions can be associated with the development and progression of neurological complications, including those related to the adult and aging brain. This review will address the effects of ZIKV infection on CNS and PNS glial cells, focusing on cellular and molecular mechanisms, including changes in the inflammatory response, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate homeostasis, neural metabolism, and neuron-glia communication. Of note, preventive and therapeutic strategies that focus on glial cells may emerge to delay and/or prevent the development of ZIKV-induced neurodegeneration and its consequences.
Collapse
Affiliation(s)
- André Quincozes-Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Naithan Ludian Fernandes Costa
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Natalie K Thomaz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rômulo Rodrigo de Souza Almeida
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Lucélia Santi
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Rafael L Rosa
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Daniela Capra
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana M Coelho-Aguiar
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcos Fabio DosSantos
- Laboratório de Propriedades Mecânicas e Biologia Celular, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Manoela Heringer
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
14
|
Immune Functions of Astrocytes in Viral Neuroinfections. Int J Mol Sci 2023; 24:ijms24043514. [PMID: 36834929 PMCID: PMC9960577 DOI: 10.3390/ijms24043514] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Neuroinfections of the central nervous system (CNS) can be triggered by various pathogens. Viruses are the most widespread and have the potential to induce long-term neurologic symptoms with potentially lethal outcomes. In addition to directly affecting their host cells and inducing immediate changes in a plethora of cellular processes, viral infections of the CNS also trigger an intense immune response. Regulation of the innate immune response in the CNS depends not only on microglia, which are fundamental immune cells of the CNS, but also on astrocytes. These cells align blood vessels and ventricle cavities, and consequently, they are one of the first cell types to become infected after the virus breaches the CNS. Moreover, astrocytes are increasingly recognized as a potential viral reservoir in the CNS; therefore, the immune response initiated by the presence of intracellular virus particles may have a profound effect on cellular and tissue physiology and morphology. These changes should be addressed in terms of persisting infections because they may contribute to recurring neurologic sequelae. To date, infections of astrocytes with different viruses originating from genetically distinct families, including Flaviviridae, Coronaviridae, Retroviridae, Togaviridae, Paramyxoviridae, Picomaviridae, Rhabdoviridae, and Herpesviridae, have been confirmed. Astrocytes express a plethora of receptors that detect viral particles and trigger signaling cascades, leading to an innate immune response. In this review, we summarize the current knowledge on virus receptors that initiate the release of inflammatory cytokines from astrocytes and depict the involvement of astrocytes in immune functions of the CNS.
Collapse
|
15
|
Sher AA, Lao YT, Coombs KM. HLA-A, HSPA5, IGFBP5 and PSMA2 Are Restriction Factors for Zika Virus Growth in Astrocytic Cells. Viruses 2022; 15:97. [PMID: 36680137 PMCID: PMC9863221 DOI: 10.3390/v15010097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
(1) Background: Zika virus (ZIKV), an arbo-flavivirus, is transmitted via Aeges aegyptii mosquitoes Following its major outbreaks in 2013, 2014 and 2016, WHO declared it a Public Health Emergency of International Concern. Symptoms of ZIKV infection include acute fever, conjunctivitis, headache, muscle & joint pain and malaise. Cases of its transmission also have been reported via perinatal, sexual and transfusion transmission. ZIKV pathologies include meningo-encephalitis and myelitis in the central nervous system (CNS) and Guillain-Barré syndrome and acute transient polyneuritis in the peripheral nervous system (PNS). Drugs like azithromycin have been tested as inhibitors of ZIKV infection but no vaccines or treatments are currently available. Astrocytes are the most abundant cells in the CNS and among the first cells in CNS infected by ZIKV; (2) Methods: We previously used SOMAScan proteomics to study ZIKV-infected astrocytic cells. Here, we use mass spectrometric analyses to further explain dysregulations in the cellular expression profile of glioblastoma astrocytoma U251 cells. We also knocked down (KD) some of the U251 cellular proteins using siRNAs and observed the impact on ZIKV replication and infectivity; (3) Results & Conclusions: The top ZIKV dysregulated cellular networks were antimicrobial response, cell death, and energy production while top dysregulated functions were antigen presentation, viral replication and cytopathic impact. Th1 and interferon signaling pathways were among the top dysregulated canonical pathways. siRNA-mediated KD of HLA-A, IGFBP5, PSMA2 and HSPA5 increased ZIKV titers and protein synthesis, indicating they are ZIKV restriction factors. ZIKV infection also restored HLA-A expression in HLA-A KD cells by 48 h post-infection, suggesting interactions between this gene product and ZIKV.
Collapse
Affiliation(s)
- Affan A. Sher
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Ying Tenny Lao
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Kevin M. Coombs
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
16
|
Losada PX, Bosch I, Frydman GH, Gehrke L, Narváez CF. Dengue and Zika virus differential infection of human megakaryoblast MEG-01 reveals unique cellular markers. Virology 2022; 577:16-23. [PMID: 36257128 DOI: 10.1016/j.virol.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/26/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022]
Abstract
Platelet count is widely used for the diagnosis and follow-up of patients with dengue. Despite its close viral structural and symptomatic homology, ZIKV infection does not typically induce significant thrombocytopenia. To determine the effect of DENV-2 and ZIKV infection on human platelet precursors we utilized MEG-01 cell line to evaluate the viral infection, viability, innate gene expression and release of platelet-like particles (PLPs). DENV-2 induced a higher proportion of cell death at 48-72 h post-infection than ZIKV. The median range of intracellular NS1+/E+ cells was 11.2% (3.3%-25%) and 5% (3%-8.1%) for DENV-2 and ZIKV, respectively (p = 0.03). MEG-01 cells infected with DENV-2 quickly expressed higher levels of IFN-β, indolamine 2,3-dioxygenase and CXCL10 mRNA compared to ZIKV infected cells and DENV-2 but not ZIKV infection reduced the number PLPs from stimulated MEG-01 cells. The results shed light into mechanisms including thrombocytopenia present in patients with DENV but absent in ZIKV infections.
Collapse
Affiliation(s)
- Paula X Losada
- División de Inmunología, Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, 410001, Huila, Colombia
| | - Irene Bosch
- Institute for Medical Engineering and Science and the Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Galit H Frydman
- Institute for Medical Engineering and Science and the Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Lee Gehrke
- Institute for Medical Engineering and Science and the Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Carlos F Narváez
- División de Inmunología, Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, 410001, Huila, Colombia.
| |
Collapse
|
17
|
Puig-Pijuan T, Souza LRQ, Pedrosa CDSG, Higa LM, Monteiro FL, Tanuri A, Valverde RHF, Einicker-Lamas M, Rehen SK. Copper regulation disturbance linked to oxidative stress and cell death during Zika virus infection in human astrocytes. J Cell Biochem 2022; 123:1997-2008. [PMID: 36063501 DOI: 10.1002/jcb.30323] [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: 02/08/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 12/24/2022]
Abstract
The Zika virus (ZIKV) caused neurological abnormalities in more than 3500 Brazilian newborns between 2015 and 2020. Data have pointed to oxidative stress in astrocytes as well as to dysregulations in neural cell proliferation and cell cycle as important events accounting for the cell death and neurological complications observed in Congenital Zika Syndrome. Copper imbalance has been shown to induce similar alterations in other pathologies, and disturbances in copper homeostasis have already been described in viral infections. Here, we investigated copper homeostasis imbalance as a factor that could contribute to the cytotoxic effects of ZIKV infection in astrocytes. Human induced pluripotent stem cell-derived astrocytes were infected with ZIKV; changes in the gene expression of copper homeostasis proteins were analyzed. The effect of the administration of CuCl2 or a copper chelator on oxidative stress, cell viability and percentage of infection were also studied. ZIKV infection leads to a downregulation of one of the transporters mediating copper release, ATP7B protein. We also observed the activation of mechanisms that counteract high copper levels, including the synthesis of copper chaperones and the reduction of the copper importer protein CTR1. Finally, we show that chelator-mediated copper sequestration in ZIKV-infected astrocytes reduces the levels of reactive oxygen species and improves cell viability, but does not change the overall percentage of infected cells. In summary, our results show that copper homeostasis imbalance plays a role in the pathology of ZIKV in astrocytes, indicating that it may also be a factor accounting for the developmental abnormalities in the central nervous system following viral infection. Evaluating micronutrient levels and the use of copper chelators in pregnant women susceptible to ZIKV infection may be promising strategies to manage novel cases of congenital ZIKV syndrome.
Collapse
Affiliation(s)
- Teresa Puig-Pijuan
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.,Laboratory of Biomembranes, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia R Q Souza
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | | | - Luiza M Higa
- Department of Genetics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio Luis Monteiro
- Department of Genetics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amilcar Tanuri
- Department of Genetics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael H F Valverde
- Laboratory of Biomembranes, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Laboratory of Biomembranes, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stevens Kastrup Rehen
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.,Department of Genetics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Mungin JW, Chen X, Liu B. Interferon Epsilon Signaling Confers Attenuated Zika Replication in Human Vaginal Epithelial Cells. Pathogens 2022; 11:853. [PMID: 36014974 PMCID: PMC9415962 DOI: 10.3390/pathogens11080853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 02/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus that causes congenital birth defects and neurological compilations in the human host. Although ZIKV is primarily transmitted through infected mosquitos, recent studies reveal sexual contact as a potential transmission route. In vagina-bearing individuals, the vaginal epithelium constitutes the first line of defense against viruses. However, it is unclear how ZIKV interacts with the vaginal epithelium to initiate ZIKV transmission. In this study, we demonstrate that exposing ZIKV to human vaginal epithelial cells (hVECs) resulted in de novo viral RNA replication, increased envelope viral protein production, and a steady, extracellular release of infectious viral particles. Interestingly, our data show that, despite an increase in viral load, the hVECs did not exhibit significant cytopathology in culture as other cell types typically do. Furthermore, our data reveal that the innate antiviral state of hVECs plays a crucial role in preventing viral cytopathology. For the first time, our data show that interferon epsilon inhibits ZIKV replication. Collectively, our results in this study provide a novel perspective on the viral susceptibility and replication dynamics during ZIKV infection in the human vaginal epithelium. These findings will be instrumental towards developing therapeutic agents aimed at eliminating the pathology caused by the virus.
Collapse
Affiliation(s)
| | | | - Bindong Liu
- Centers for AIDS Health Disparity Research, Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (J.W.M.J.); (X.C.)
| |
Collapse
|
19
|
Brokatzky D, Häcker G. Mitochondria: intracellular sentinels of infections. Med Microbiol Immunol 2022; 211:161-172. [PMID: 35790577 PMCID: PMC9255486 DOI: 10.1007/s00430-022-00742-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/14/2022] [Indexed: 01/19/2023]
Abstract
Structure and integrity of the mitochondrial network play important roles in many cellular processes. Loss of integrity can lead to the activation of a variety of signalling pathways and affect the cell’s response to infections. The activation of such mitochondria-mediated cellular responses has implications for infection recognition, signal transduction and pathogen control. Although we have a basic understanding of mitochondrial factors such as mitochondrial DNA or RNA that may be involved in processes like pro-inflammatory signalling, the diverse roles of mitochondria in host defence remain unclear. Here we will first summarise the functions of mitochondria in the host cell and provide an overview of the major known mitochondrial stress responses. We will then present recent studies that have contributed to the understanding of the role of mitochondria in infectious diseases and highlight a number of recently investigated models of bacterial and viral infections.
Collapse
Affiliation(s)
- Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Medical Centre University of Freiburg, Faculty of Medicine, 79104, Freiburg, Germany
| |
Collapse
|
20
|
Komarasamy TV, Adnan NAA, James W, Balasubramaniam VRMT. Zika Virus Neuropathogenesis: The Different Brain Cells, Host Factors and Mechanisms Involved. Front Immunol 2022; 13:773191. [PMID: 35371036 PMCID: PMC8966389 DOI: 10.3389/fimmu.2022.773191] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/21/2022] [Indexed: 12/16/2022] Open
Abstract
Zika virus (ZIKV), despite being discovered six decades earlier, became a major health concern only after an epidemic in French Polynesia and an increase in the number of microcephaly cases in Brazil. Substantial evidence has been found to support the link between ZIKV and neurological complications in infants. The virus targets various cells in the brain, including radial glial cells, neural progenitor cells (NPCs), astrocytes, microglial and glioblastoma stem cells. It affects the brain cells by exploiting different mechanisms, mainly through apoptosis and cell cycle dysregulation. The modulation of host immune response and the inflammatory process has also been demonstrated to play a critical role in ZIKV induced neurological complications. In addition to that, different ZIKV strains have exhibited specific neurotropism and unique molecular mechanisms. This review provides a comprehensive and up-to-date overview of ZIKV-induced neuroimmunopathogenesis by dissecting its main target cells in the brain, and the underlying cellular and molecular mechanisms. We highlighted the roles of the different ZIKV host factors and how they exploit specific host factors through various mechanisms. Overall, it covers key components for understanding the crosstalk between ZIKV and the brain.
Collapse
Affiliation(s)
- Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Nur Amelia Azreen Adnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - William James
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Vinod R M T Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
21
|
Astrocyte Control of Zika Infection Is Independent of Interferon Type I and Type III Expression. BIOLOGY 2022; 11:biology11010143. [PMID: 35053142 PMCID: PMC8772967 DOI: 10.3390/biology11010143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022]
Abstract
Simple Summary Zika virus (ZIKV) is a mosquito-borne virus first isolated from the Zika forest, Uganda, in 1947, which has been spreading across continents since then. We now know ZIKV causes both microencephaly in newborns and neurological complications in adults; however, no effective treatment options have yet been found. A more complete understanding of Zika-infection-mediated pathogenesis and host responses is required to enable the development of novel treatment strategies. In this study, efforts were made to elucidate the host responses following Zika virus infection using several astrocyte cell models, as astrocytes are a major cell type within the central nervous system (CNS) with significant antiviral ability. Our data suggest that astrocytes can resist ZIKV both in an interferon type I- and III-independent manner and suggest that an early and more diverse antiviral response may be more effective in controlling Zika infection. This study also identifies astrocyte cellular models that appear to display differential abilities in the control of viral infection, which may assist in the study of alternate neurotropic virus infections. Overall, this work adds to the growing body of knowledge surrounding ZIKV-mediated cellular host interactions and will contribute to a better understanding of ZIKV-mediated pathogenesis. Abstract Zika virus (ZIKV) is a pathogenic neurotropic virus that infects the central nervous system (CNS) and results in various neurological complications. Astrocytes are the dominant CNS cell producer of the antiviral cytokine IFN-β, however little is known about the factors involved in their ability to mediate viral infection control. Recent studies have displayed differential responses in astrocytes to ZIKV infection, and this study sought to elucidate astrocyte cell-specific responses to ZIKV using a variety of cell models infected with either the African (MR766) or Asian (PRVABC59) ZIKV strains. Expression levels of pro-inflammatory (TNF-α and IL-1β) and inflammatory (IL-8) cytokines following viral infection were low and mostly comparable within the ZIKV-resistant and ZIKV-susceptible astrocyte models, with better control of proinflammatory cytokines displayed in resistant astrocyte cells, synchronising with the viral infection level at specific timepoints. Astrocyte cell lines displaying ZIKV-resistance also demonstrated early upregulation of multiple antiviral genes compared with susceptible astrocytes. Interestingly, pre-stimulation of ZIKV-susceptible astrocytes with either poly(I:C) or poly(dA:dT) showed efficient protection against ZIKV compared with pre-stimulation with either recombinant IFN-β or IFN-λ, perhaps indicating that a more diverse antiviral gene expression is necessary for astrocyte control of ZIKV, and this is driven in part through interferon-independent mechanisms.
Collapse
|
22
|
Tavčar Verdev P, Potokar M, Korva M, Resman Rus K, Kolenc M, Avšič Županc T, Zorec R, Jorgačevski J. In human astrocytes neurotropic flaviviruses increase autophagy, yet their replication is autophagy-independent. Cell Mol Life Sci 2022; 79:566. [PMID: 36283999 PMCID: PMC9596533 DOI: 10.1007/s00018-022-04578-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 01/18/2023]
Abstract
Astrocytes, an abundant type of glial cells, are the key cells providing homeostasis in the central nervous system. Due to their susceptibility to infection, combined with high resilience to virus-induced cell death, astrocytes are now considered one of the principal types of cells, responsible for virus retention and dissemination within the brain. Autophagy plays an important role in elimination of intracellular components and in maintaining cellular homeostasis and is also intertwined with the life cycle of viruses. The physiological significance of autophagy in astrocytes, in connection with the life cycle and transmission of viruses, remains poorly investigated. In the present study, we investigated flavivirus-induced modulation of autophagy in human astrocytes by monitoring a tandem fluorescent-tagged LC3 probe (mRFP-EGFP-LC3) with confocal and super-resolution fluorescence microscopy. Astrocytes were infected with tick-borne encephalitis virus (TBEV) or West Nile virus (WNV), both pathogenic flaviviruses, and with mosquito-only flavivirus (MOF), which is considered non-pathogenic. The results revealed that human astrocytes are susceptible to infection with TBEV, WNV and to a much lower extent also to MOF. Infection and replication rates of TBEV and WNV are paralleled by increased rate of autophagy, whereas autophagosome maturation and the size of autophagic compartments are not affected. Modulation of autophagy by rapamycin and wortmannin does not influence TBEV and WNV replication rate, whereas bafilomycin A1 attenuates their replication and infectivity. In human astrocytes infected with MOF, the low infectivity and the lack of efficient replication of this flavivirus are mirrored by the absence of an autophagic response.
Collapse
Affiliation(s)
- Petra Tavčar Verdev
- grid.8954.00000 0001 0721 6013Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Potokar
- grid.8954.00000 0001 0721 6013Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia ,grid.433223.7Celica Biomedical, Ljubljana, Slovenia
| | - Miša Korva
- grid.8954.00000 0001 0721 6013Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katarina Resman Rus
- grid.8954.00000 0001 0721 6013Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Marko Kolenc
- grid.8954.00000 0001 0721 6013Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tatjana Avšič Županc
- grid.8954.00000 0001 0721 6013Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- grid.8954.00000 0001 0721 6013Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia ,grid.433223.7Celica Biomedical, Ljubljana, Slovenia
| | - Jernej Jorgačevski
- grid.8954.00000 0001 0721 6013Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia ,grid.433223.7Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
23
|
Qiao X, Zhang W, Zhao W. Role of CXCL10 in Spinal Cord Injury. Int J Med Sci 2022; 19:2058-2070. [PMID: 36483597 PMCID: PMC9724238 DOI: 10.7150/ijms.76694] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury (SCI) results in acute inflammatory responses and secondary damages, including neuronal and glial cell death, axonal damage and demyelination, and blood-brain barrier (BBB) damage, eventually leading to neuronal dysfunction and other complications. C-X-C motif Chemokine Ligand 10 (CXCL10) is expressed after the injury, playing multiple roles in the development and progression of SCI. Moreover, the CXCL10 antagonist can restrict inflammatory immune responses and promote neuronal regeneration and functional recovery. In this review, we summarize the structure and biological functions of CXCL10, and the roles of the CXCL10 / CXCR3 axis in acute inflammatory responses, secondary damages, and complications during SCI, thus providing a potential theoretical basis by highlighting CXCL10 as a new potential drug target for the treatment of SCI.
Collapse
Affiliation(s)
- Xinyu Qiao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, China
| | - Weijiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
24
|
de Mendonça-Vieira LR, Aníbal-Silva CE, Menezes-Neto A, Azevedo EDAN, Zanluqui NG, Peron JPS, Franca RFDO. Reactive Oxygen Species (ROS) Are Not a Key Determinant for Zika Virus-Induced Apoptosis in SH-SY5Y Neuroblastoma Cells. Viruses 2021; 13:v13112111. [PMID: 34834918 PMCID: PMC8622630 DOI: 10.3390/v13112111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/27/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction: ZIKV is a highly neurotropic virus that can cause the death of infected neuroprogenitor cells through mitochondrial damage and intrinsic apoptotic signaling. In this context, the role of reactive oxygen species (ROS) in neuronal cell death caused by ZIKV still remains elusive. Objective: We aimed at evaluating the role of these cellular components in the death of human undifferentiated neuroblastoma cell line infected with ZIKV. Results: ZIKV infection resulted in the extensive death of SH-SY5Y cells with the upregulation of several genes involved in survival and apoptotic responses as well as the colocalization of mitochondrial staining with ZIKV Envelope (E) protein. Notably, levels of intracellular reactive oxygen species (ROS) were not altered during ZIKV infection in undifferentiated SH-SY5Y cells, and consistent with these results, the treatment of infected cells with the widely studied ROS scavenger N-acetylcysteine (NAC) did not prevent cell death in these cells. Conclusion: Altogether, our results suggest that excessive ROS production is not the main trigger of SH-SY5Y cells death in ZIKV infection.
Collapse
Affiliation(s)
- Leila Rodrigues de Mendonça-Vieira
- Oswaldo Cruz Foundation/Fiocruz, Aggeu Magalhães Institute, Recife 50740-465, Brazil; (A.M.-N.); (E.d.A.N.A.)
- Correspondence: (L.R.d.M.-V.); (R.F.d.O.F.)
| | | | - Armando Menezes-Neto
- Oswaldo Cruz Foundation/Fiocruz, Aggeu Magalhães Institute, Recife 50740-465, Brazil; (A.M.-N.); (E.d.A.N.A.)
| | | | - Nágela Ghabdan Zanluqui
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil; (N.G.Z.); (J.P.S.P.)
| | - Jean Pierre Schatzmann Peron
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil; (N.G.Z.); (J.P.S.P.)
| | - Rafael Freitas de Oliveira Franca
- Oswaldo Cruz Foundation/Fiocruz, Aggeu Magalhães Institute, Recife 50740-465, Brazil; (A.M.-N.); (E.d.A.N.A.)
- Correspondence: (L.R.d.M.-V.); (R.F.d.O.F.)
| |
Collapse
|
25
|
Polonio CM, Peron JPS. ZIKV Infection and miRNA Network in Pathogenesis and Immune Response. Viruses 2021; 13:v13101992. [PMID: 34696422 PMCID: PMC8541119 DOI: 10.3390/v13101992] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 01/01/2023] Open
Abstract
Over the years, viral infections have caused severe illness in humans. Zika Virus (ZIKV) is a flavivirus transmitted by mosquito vectors that leads to notable neurological impairment, whose most dramatic impact is the Congenital ZIKV Syndrome (CZS). ZIKV targets neuronal precursor cells leading to apoptosis and further impairment of neuronal development, causing microcephaly, lissencephaly, ventriculomegaly, and calcifications. Several regulators of biological processes are involved in CZS development, and in this context, microRNAs (miRNAs) seem to have a fundamental role. miRNAs are important regulators of protein translation, as they form the RISC silencing complex and interact with complementary mRNA target sequences to further post-transcriptional repression. In this context, little is known about their participation in the pathogenesis of viral infections. In this review, we discuss how miRNAs could relate to ZIKV and other flavivirus infections.
Collapse
Affiliation(s)
- Carolina Manganeli Polonio
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil;
- Laboratory of Neuroimmunology of Arboviruses, Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-020, Brazil
| | - Jean Pierre Schatzmann Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil;
- Laboratory of Neuroimmunology of Arboviruses, Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-020, Brazil
- Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo, São Paulo 01246-000, Brazil
- Correspondence:
| |
Collapse
|
26
|
Dahal B, Lehman CW, Akhrymuk I, Bracci NR, Panny L, Barrera MD, Bhalla N, Jacobs JL, Dinman JD, Kehn-Hall K. PERK Is Critical for Alphavirus Nonstructural Protein Translation. Viruses 2021; 13:892. [PMID: 34065980 PMCID: PMC8151226 DOI: 10.3390/v13050892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is an alphavirus that causes encephalitis. Previous work indicated that VEEV infection induced early growth response 1 (EGR1) expression, leading to cell death via the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) arm of the unfolded protein response (UPR) pathway. Loss of PERK prevented EGR1 induction and decreased VEEV-induced death. The results presented within show that loss of PERK in human primary astrocytes dramatically reduced VEEV and eastern equine encephalitis virus (EEEV) infectious titers by 4-5 log10. Loss of PERK also suppressed VEEV replication in primary human pericytes and human umbilical vein endothelial cells, but it had no impact on VEEV replication in transformed U87MG and 293T cells. A significant reduction in VEEV RNA levels was observed as early as 3 h post-infection, but viral entry assays indicated that the loss of PERK minimally impacted VEEV entry. In contrast, the loss of PERK resulted in a dramatic reduction in viral nonstructural protein translation and negative-strand viral RNA production. The loss of PERK also reduced the production of Rift Valley fever virus and Zika virus infectious titers. These data indicate that PERK is an essential factor for the translation of alphavirus nonstructural proteins and impacts multiple RNA viruses, making it an exciting target for antiviral development.
Collapse
Affiliation(s)
- Bibha Dahal
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
| | - Caitlin W. Lehman
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Ivan Akhrymuk
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Nicole R. Bracci
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Lauren Panny
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Michael D. Barrera
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
| | - Nishank Bhalla
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
| | | | - Jonathan D. Dinman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
27
|
Tavčar P, Potokar M, Kolenc M, Korva M, Avšič-Županc T, Zorec R, Jorgačevski J. Neurotropic Viruses, Astrocytes, and COVID-19. Front Cell Neurosci 2021; 15:662578. [PMID: 33897376 PMCID: PMC8062881 DOI: 10.3389/fncel.2021.662578] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
At the end of 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was discovered in China, causing a new coronavirus disease, termed COVID-19 by the WHO on February 11, 2020. At the time of this paper (January 31, 2021), more than 100 million cases have been recorded, which have claimed over 2 million lives worldwide. The most important clinical presentation of COVID-19 is severe pneumonia; however, many patients present various neurological symptoms, ranging from loss of olfaction, nausea, dizziness, and headache to encephalopathy and stroke, with a high prevalence of inflammatory central nervous system (CNS) syndromes. SARS-CoV-2 may also target the respiratory center in the brainstem and cause silent hypoxemia. However, the neurotropic mechanism(s) by which SARS-CoV-2 affects the CNS remain(s) unclear. In this paper, we first address the involvement of astrocytes in COVID-19 and then elucidate the present knowledge on SARS-CoV-2 as a neurotropic virus as well as several other neurotropic flaviviruses (with a particular emphasis on the West Nile virus, tick-borne encephalitis virus, and Zika virus) to highlight the neurotropic mechanisms that target astroglial cells in the CNS. These key homeostasis-providing cells in the CNS exhibit many functions that act as a favorable milieu for virus replication and possibly a favorable environment for SARS-CoV-2 as well. The role of astrocytes in COVID-19 pathology, related to aging and neurodegenerative disorders, and environmental factors, is discussed. Understanding these mechanisms is key to better understanding the pathophysiology of COVID-19 and for developing new strategies to mitigate the neurotropic manifestations of COVID-19.
Collapse
Affiliation(s)
- Petra Tavčar
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Potokar
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Celica Biomedical, Ljubljana, Slovenia
| | - Marko Kolenc
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Miša Korva
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tatjana Avšič-Županc
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Celica Biomedical, Ljubljana, Slovenia
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
28
|
Geddes VEV, Brustolini OJB, Cavalcante LTDF, Moreira FRR, de Castro FL, Guimarães APDC, Gerber AL, Figueiredo CM, Diniz LP, Neto EDA, Tanuri A, Souza RP, Assunção-Miranda I, Alves-Leon SV, Romão LF, de Souza JPBM, de Vasconcelos ATR, de Aguiar RS. Common Dysregulation of Innate Immunity Pathways in Human Primary Astrocytes Infected With Chikungunya, Mayaro, Oropouche, and Zika Viruses. Front Cell Infect Microbiol 2021; 11:641261. [PMID: 33791243 PMCID: PMC8006316 DOI: 10.3389/fcimb.2021.641261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/28/2021] [Indexed: 12/22/2022] Open
Abstract
Arboviruses pose a major threat throughout the world and represent a great burden in tropical countries of South America. Although generally associated with moderate febrile illness, in more severe cases they can lead to neurological outcomes, such as encephalitis, Guillain-Barré syndrome, and Congenital Syndromes. In this context astrocytes play a central role in production of inflammatory cytokines, regulation of extracellular matrix, and control of glutamate driven neurotoxicity in the central nervous system. Here, we presented a comprehensive genome-wide transcriptome analysis of human primary astrocytes infected with Chikungunya, Mayaro, Oropouche, or Zika viruses. Analyses of differentially expressed genes (DEGs), pathway enrichment, and interactomes have shown that Alphaviruses up-regulated genes related to elastic fiber formation and N-glycosylation of glycoproteins, with down-regulation of cell cycle and DNA stability and chromosome maintenance genes. In contrast, Oropouche virus up-regulated cell cycle and DNA maintenance and condensation pathways while down-regulated extracellular matrix, collagen metabolism, glutamate and ion transporters pathways. Zika virus infection only up-regulated eukaryotic translation machinery while down-regulated interferon pathways. Reactome and integration analysis revealed a common signature in down-regulation of innate immune response, antiviral response, and inflammatory cytokines associated to interferon pathway for all arboviruses tested. Validation of interferon stimulated genes by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) corroborated our transcriptome findings. Altogether, our results showed a co-evolution in the mechanisms involved in the escape of arboviruses to antiviral immune response mediated by the interferon (IFN) pathway.
Collapse
Affiliation(s)
- Victor Emmanuel Viana Geddes
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Biologia Integrativa, Departamento de Genética Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Otávio José Bernardes Brustolini
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Ministério de Ciência Tecnologia e Comunicações, Petrópolis, Brazil
| | - Liliane Tavares de Faria Cavalcante
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Filipe Romero Rebello Moreira
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando Luz de Castro
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula de Campos Guimarães
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Ministério de Ciência Tecnologia e Comunicações, Petrópolis, Brazil
| | - Alexandra Lehmkuhl Gerber
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Ministério de Ciência Tecnologia e Comunicações, Petrópolis, Brazil
| | - Camila Menezes Figueiredo
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eurico de Arruda Neto
- Departamento de Biologia Celular e Molecular, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Amilcar Tanuri
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renan Pedra Souza
- Laboratório de Biologia Integrativa, Departamento de Genética Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Iranaia Assunção-Miranda
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Soniza Vieira Alves-Leon
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Ferreira Romão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ana Tereza Ribeiro de Vasconcelos
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Ministério de Ciência Tecnologia e Comunicações, Petrópolis, Brazil
| | - Renato Santana de Aguiar
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Biologia Integrativa, Departamento de Genética Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
29
|
Bruno SR, Anathy V. Lung epithelial endoplasmic reticulum and mitochondrial 3D ultrastructure: a new frontier in lung diseases. Histochem Cell Biol 2021; 155:291-300. [PMID: 33598824 PMCID: PMC7889473 DOI: 10.1007/s00418-020-01950-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
It has long been appreciated that the endoplasmic reticulum (ER) and mitochondria, organelles important for regular cell function and survival, also play key roles in pathogenesis of various lung diseases, including asthma, fibrosis, and infections. Alterations in processes regulated within these organelles, including but not limited to protein folding in the ER and oxidative phosphorylation in the mitochondria, are important in disease pathogenesis. In recent years it has also become increasingly apparent that organelle structure dictates function. It is now clear that organelles must maintain precise organization and localization for proper function. Newer microscopy capabilities have allowed the scientific community to reveal, via 3D imaging, that the structure of these organelles and their interactions with each other are a main component of regulating function and, therefore, effects on the disease state. In this review, we will examine how 3D imaging through techniques could allow advancements in knowledge of how the ER and mitochondria function and the roles they may play in lung epithelia in progression of lung disease.
Collapse
Affiliation(s)
- Sierra R Bruno
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, 149 Beaumont Ave, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, 149 Beaumont Ave, Burlington, VT, 05405, USA.
| |
Collapse
|
30
|
Schouest B, Peterson TA, Szeltner DM, Scheef EA, Baddoo M, Ungerleider N, Flemington EK, MacLean AG, Maness NJ. Transcriptional signatures of Zika virus infection in astrocytes. J Neurovirol 2021; 27:116-125. [PMID: 33405202 PMCID: PMC7921019 DOI: 10.1007/s13365-020-00931-3] [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: 07/13/2020] [Revised: 11/09/2020] [Accepted: 12/02/2020] [Indexed: 01/18/2023]
Abstract
Astrocytes are an early and important target of Zika virus (ZIKV) infection in the developing brain, but the impacts of infection on astrocyte function remain controversial. Given that nonhuman primate (NHP) models of ZIKV infection replicate aspects of neurologic disease seen in human infections, we cultured primary astrocytes from the brain tissue of infant rhesus macaques and then infected the cells with Asian or African lineage ZIKV to identify transcriptional patterns associated with infection in these cells. The African lineage virus appeared to have greater infectivity and promote stronger antiviral signaling, but infection by either strain ultimately produced typical virus response patterns. Both viruses induced hypoxic stress, but the Asian lineage strain additionally had an effect on metabolic and lipid biosynthesis pathways. Together, these findings describe an NHP astrocyte model that may be used to assess transcriptional signatures following ZIKV infection.
Collapse
Affiliation(s)
- Blake Schouest
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
- Biomedical Sciences Training Program, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tiffany A Peterson
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
- Biomedical Sciences Training Program, Tulane University School of Medicine, New Orleans, LA, USA
| | - Dawn M Szeltner
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Elizabeth A Scheef
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Melody Baddoo
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nathan Ungerleider
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Erik K Flemington
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Andrew G MacLean
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
31
|
Spencer Clinton JL, Tran LL, Vogt MB, Rowley DR, Kimata JT, Rico-Hesse R. IP-10 and CXCR3 signaling inhibit Zika virus replication in human prostate cells. PLoS One 2020; 15:e0244587. [PMID: 33378361 PMCID: PMC7773246 DOI: 10.1371/journal.pone.0244587] [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: 07/11/2020] [Accepted: 12/12/2020] [Indexed: 11/18/2022] Open
Abstract
Our previous studies have shown that Zika virus (ZIKV) replicates in human prostate cells, suggesting that the prostate may serve as a long-term reservoir for virus transmission. Here, we demonstrated that the innate immune responses generated to three distinct ZIKV strains (all isolated from human serum) were significantly different and dependent on their passage history (in mosquito, monkey, or human cells). In addition, some of these phenotypic differences were reduced by a single additional cell culture passage, suggesting that viruses that have been passaged more than 3 times from the patient sample will no longer reflect natural phenotypes. Two of the ZIKV strains analyzed induced high levels of the IP-10 chemokine and IFNγ in human prostate epithelial and stromal mesenchymal stem cells. To further understand the importance of these innate responses on ZIKV replication, we measured the effects of IP-10 and its downstream receptor, CXCR3, on RNA and virus production in prostate cells. Treatment with IP-10, CXCR3 agonist, or CXCR3 antagonist significantly altered ZIKV viral gene expression, depending on their passage in cells of relevant hosts (mosquito or human). We detected differences in gene expression of two primary CXCR3 isoforms (CXCR3-A and CXCR3-B) on the two cell types, possibly explaining differences in viral output. Lastly, we examined the effects of IP-10, agonist, or antagonist on cell death and proliferation under physiologically relevant infection rates, and detected no significant differences. Although we did not measure protein expression directly, our results indicate that CXCR3 signaling may be a target for therapeutics, to ultimately stop sexual transmission of this virus.
Collapse
Affiliation(s)
- Jennifer L. Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Linda L. Tran
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Megan B. Vogt
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - David R. Rowley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rebecca Rico-Hesse
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
32
|
Guzeloglu-Kayisli O, Guo X, Tang Z, Semerci N, Ozmen A, Larsen K, Mutluay D, Guller S, Schatz F, Kayisli UA, Lockwood CJ. Zika Virus-Infected Decidual Cells Elicit a Gestational Age-Dependent Innate Immune Response and Exaggerate Trophoblast Zika Permissiveness: Implication for Vertical Transmission. THE JOURNAL OF IMMUNOLOGY 2020; 205:3083-3094. [PMID: 33139490 DOI: 10.4049/jimmunol.2000713] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Vertical transmission of the Zika virus (ZIKV) causes severe fetal defects, but the exact pathogenic mechanism is unclear. We identified up to a 10,480-fold higher expression of viral attachment factors AXL, GAS6, and PROS1 and a 3880-fold increase in ZIKV infectiousness/propagation in human term decidual stromal cells versus trophoblasts. Moreover, levels of viral attachment factors and ZIKV are significantly increased, whereas expression of innate immune response genes are significantly decreased, in human first trimester versus term decidual cells. ZIKV-infected decidual cell supernatants increased cytotrophoblasts infection up to 252-fold compared with directly infected cytotrophoblasts. Tizoxanide treatment efficiently inhibited Zika infection in both maternal and fetal cells. We conclude that ZIKV permissiveness, as well as innate immune responsiveness of human decidual cells, are gestational age dependent, and decidual cells augment ZIKV infection of primary human cytotrophoblast cultures, which are otherwise ZIKV resistant. Human decidual cells may act as reservoirs for trimester-dependent placental transmission of ZIKV, accounting for the higher Zika infection susceptibility and more severe fetal sequelae observed in early versus late pregnancy. Moreover, tizoxanide is a promising agent in preventing perinatal Zika transmission as well as other RNA viruses such as coronavirus.
Collapse
Affiliation(s)
- Ozlem Guzeloglu-Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Xiaofang Guo
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Zhonghua Tang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Asli Ozmen
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Kellie Larsen
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Duygu Mutluay
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Frederick Schatz
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Umit Ali Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Charles Joseph Lockwood
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| |
Collapse
|
33
|
Castanospermine reduces Zika virus infection-associated seizure by inhibiting both the viral load and inflammation in mouse models. Antiviral Res 2020; 183:104935. [PMID: 32949636 PMCID: PMC7492813 DOI: 10.1016/j.antiviral.2020.104935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 11/30/2022]
Abstract
Zika virus (ZIKV) outbreaks have been reported worldwide, including a recent occurrence in Brazil where it spread rapidly, and an association with increased cases of microcephaly was observed in addition to neurological issues such as GBS that were reported during previous outbreaks. Following infection of neuronal tissues, ZIKV can cause inflammation, which may lead to neuronal abnormalities, including seizures and paralysis. Therefore, a drug containing both anti-viral and immunosuppressive properties would be of great importance in combating ZIKV related neurological abnormalities. Castanospermine (CST) is potentially a right candidate drug as it reduced viral load and brain inflammation with the resulting appearance of delayed neuronal disorders, including seizures and paralysis in an Ifnar1−/− mouse. Anti-ZIKV activity of castanospermine (CST) In vivo and in vitro. CST reduces ZIKV induced inflammation of brain. CST delays the ZIKV induced seizure and improves neuronal disorders such as motor function. CST gives marginal improvement in survivability in Ifnar1−/− mice.
Collapse
|
34
|
Liang Y, Yi P, Ru W, Jie Z, Wang H, Ghanayem T, Wang X, Alamer E, Liu J, Hu H, Soong L, Cai J, Sun J. IL-22 hinders antiviral T cell responses and exacerbates ZIKV encephalitis in immunocompetent neonatal mice. J Neuroinflammation 2020; 17:249. [PMID: 32843067 PMCID: PMC7448338 DOI: 10.1186/s12974-020-01928-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/13/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The Zika virus (ZIKV) outbreak that occurred in multiple countries was linked to increased risk of nervous system injuries and congenital defects. However, host immunity- and immune-mediated pathogenesis in ZIKV infection are not well understood. Interleukin-22 (IL-22) is a crucial cytokine for regulating host immunity in infectious diseases. Whether IL-22 plays, a role in ZIKV infection is unknown. METHODS The cellular source of IL-22 was identified in IFNAR-/- mice and wild-type (WT) neonatal mice during ZIKV infection. To determine the role of IL-22, we challenged 1-day-old WT and IL-22-/- mice with ZIKV and monitored clinical manifestations. Glial cell activation in the brain was assessed by confocal imaging. ZIKV-specific CD8+ T cell responses in both the spleen and brain were analyzed by flow cytometry. In addition, glial cells were cultured in vitro and infected with ZIKV in the presence of IL-22, followed by the evaluation of cell proliferation, cytokine expression, and viral loads. RESULTS We found that γδ T cells were the main source of IL-22 during ZIKV infection in both the spleen and brain. WT mice began to exhibit weight loss, staggered steps, bilateral hind limb paralysis, and weakness at 10 days post-infection (dpi) and ultimately succumbed to infection at 16-19 dpi. IL-22 deficiency lessened weight loss, moderated the systemic inflammatory response, and greatly improved clinical signs of neurological disease and mortality. ZIKV infection also induced the activation of microglia and astrocytes in vitro. Additional analysis demonstrated that the absence of IL-22 resulted in reduced activation of microglia and astrocytes in the cortex. Although IL-22 displayed a negligible effect on glial cells in vitro, IL-22-/- mice mounted more vigorous ZIKV-specific CD8+ T cell responses, which led to a more effective control of ZIKV in the brain. CONCLUSIONS Our data revealed a pathogenic role of IL-22 in ZIKV encephalitis.
Collapse
Affiliation(s)
- Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA.
| | - Panpan Yi
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenjuan Ru
- Department of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Zuliang Jie
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Tamer Ghanayem
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
| | - Xiaofang Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Edrous Alamer
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Jinjun Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Jiyang Cai
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
35
|
Clé M, Eldin P, Briant L, Lannuzel A, Simonin Y, Van de Perre P, Cabié A, Salinas S. Neurocognitive impacts of arbovirus infections. J Neuroinflammation 2020; 17:233. [PMID: 32778106 PMCID: PMC7418199 DOI: 10.1186/s12974-020-01904-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/17/2020] [Indexed: 12/15/2022] Open
Abstract
Arthropod-borne viruses or arbovirus, are most commonly associated with acute infections, resulting on various symptoms ranging from mild fever to more severe disorders such as hemorrhagic fever. Moreover, some arboviral infections can be associated with important neuroinflammation that can trigger neurological disorders including encephalitis, paralysis, ophthalmological impairments, or developmental defects, which in some cases, can lead to long-term defects of the central nervous system (CNS). This is well illustrated in Zika virus-associated congenital brain malformations but also in West Nile virus-induced synaptic dysfunctions that can last well beyond infection and lead to cognitive deficits. Here, we summarize clinical and mechanistic data reporting on cognitive disturbances triggered by arboviral infections, which may highlight growing public health issues spanning the five continents.
Collapse
Affiliation(s)
- Marion Clé
- Pathogenesis and Control of Chronic Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Patrick Eldin
- Institute of Research in Infectiology of Montpellier, CNRS, University of Montpellier, Montpellier, France
| | - Laurence Briant
- Institute of Research in Infectiology of Montpellier, CNRS, University of Montpellier, Montpellier, France
| | - Annie Lannuzel
- Neurology Unit, INSERM CIC 1424, Guadeloupe University Hospital, Université des Antilles, Pointe-à-Pitre, Guadeloupe, France
- INSERM U1127, CNRS, UMR7225, Brain and Spine Institute, Sorbonne University Medical School, Paris, France
| | - Yannick Simonin
- Pathogenesis and Control of Chronic Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic Infections, INSERM, University of Montpellier, Etablissement Français du Sang, CHU Montpellier, Montpellier, France
| | - André Cabié
- INSERM CIC 1424, Infectious Disease and Tropical Medicine Unit, Martinique University Hospital, Université des Antilles EA4537, Martinique, France.
| | - Sara Salinas
- Pathogenesis and Control of Chronic Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Montpellier, France.
| |
Collapse
|
36
|
Vargas G, Medeiros Geraldo LH, Gedeão Salomão N, Viana Paes M, Regina Souza Lima F, Carvalho Alcantara Gomes F. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and glial cells: Insights and perspectives. Brain Behav Immun Health 2020; 7:100127. [PMID: 32838339 PMCID: PMC7423575 DOI: 10.1016/j.bbih.2020.100127] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
In December 2019, a pneumonia outbreak was reported in Wuhan, Hubei province, China. Since then, the World Health Organization declared a public health emergency of international concern due to a growing number of deaths around the globe, as well as unparalleled economic and sociodemographic consequences. The disease called coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel form of human coronavirus. Although coronavirus infections have been associated with neurological manifestations such as febrile seizures, convulsions, change in mental status, and encephalitis, less is known about the impact of SARS-CoV-2 in the brain. Recently, emerging evidence suggests that SARS-CoV-2 is associated with neurological alterations in COVID-19 patients with severe clinical manifestations. The molecular and cellular mechanisms involved in this process, as well as the neurotropic and neuroinvasive properties of SARS-CoV-2, are still poorly understood. Glial cells, such as astrocytes and microglia, play pivotal roles in the brain response to neuroinflammatory insults and neurodegenerative diseases. Further, accumulating evidence has shown that those cells are targets of several neurotropic viruses that severely impact their function. Glial cell dysfunctions have been associated with several neuroinflammatory diseases, suggesting that SARS-CoV-2 likely has a primary effect on these cells in addition to a secondary effect from neuronal damage. Here, we provide an overview of these data and discuss the possible implications of glial cells as targets of SARS-CoV-2. Considering the roles of microglia and astrocytes in brain inflammatory responses, we shed light on glial cells as possible drivers and potential targets of therapeutic strategies against neurological manifestations in patients with COVID-19. The main goal of this review is to highlight the need to consider glial involvement in the progression of COVID-19 and potentially include astrocytes and microglia as mediators of SARS-CoV-2-induced neurological damage.
Collapse
Affiliation(s)
- Gabriele Vargas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Natália Gedeão Salomão
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | - Marciano Viana Paes
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | - Flavia Regina Souza Lima
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
37
|
Estofolete CF, Milhim BHGA, Zini N, Scamardi SN, Selvante JD, Vasilakis N, Nogueira ML. Flavivirus Infection Associated with Cerebrovascular Events. Viruses 2020; 12:v12060671. [PMID: 32580374 PMCID: PMC7354470 DOI: 10.3390/v12060671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) of the genus Flavivirus are distributed globally and cause significant human disease and mortality annually. Flavivirus infections present a spectrum of clinical manifestations, ranging from asymptomatic to severe manifestations, including hemorrhage, encephalitis and death. Herein, we describe 3 case reports of cerebrovascular involvement in patients infected by dengue and Zika viruses in Sao Jose do Rio Preto, São Paulo State, Brazil, a hyperendemic area for arbovirus circulation, including dengue, yellow fever, chikungunya and Saint Louis encephalitis viruses. Our findings highlight the potential threat that unusual clinical manifestations may pose to arbovirus disease management and recovery.
Collapse
Affiliation(s)
- Cássia F Estofolete
- Department of Infectious, Dermatological and Parasitic Infections, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto 15090-000, Brazil
| | - Bruno H G A Milhim
- Department of Infectious, Dermatological and Parasitic Infections, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto 15090-000, Brazil
| | - Nathalia Zini
- Department of Infectious, Dermatological and Parasitic Infections, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto 15090-000, Brazil
| | - Samuel N Scamardi
- Department of Infectious, Dermatological and Parasitic Infections, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto 15090-000, Brazil
| | - Joana D'Arc Selvante
- Department of Infectious, Dermatological and Parasitic Infections, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto 15090-000, Brazil
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0609, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0609, USA
- Center for Tropical Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0609, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0610, USA
| | - Maurício L Nogueira
- Department of Infectious, Dermatological and Parasitic Infections, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto 15090-000, Brazil
| |
Collapse
|
38
|
Mutso M, St John JA, Ling ZL, Burt FJ, Poo YS, Liu X, Žusinaite E, Grau GE, Hueston L, Merits A, King NJ, Ekberg JA, Mahalingam S. Basic insights into Zika virus infection of neuroglial and brain endothelial cells. J Gen Virol 2020; 101:622-634. [PMID: 32375993 PMCID: PMC7414445 DOI: 10.1099/jgv.0.001416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/24/2020] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) has recently emerged as an important human pathogen due to the strong evidence that it causes disease of the central nervous system, particularly microcephaly and Guillain-Barré syndrome. The pathogenesis of disease, including mechanisms of neuroinvasion, may include both invasion via the blood-brain barrier and via peripheral (including cranial) nerves. Cellular responses to infection are also poorly understood. This study characterizes the in vitro infection of laboratory-adapted ZIKV African MR766 and two Asian strains of (1) brain endothelial cells (hCMEC/D3 cell line) and (2) olfactory ensheathing cells (OECs) (the neuroglia populating cranial nerve I and the olfactory bulb; both human and mouse OEC lines) in comparison to kidney epithelial cells (Vero cells, in which ZIKV infection is well characterized). Readouts included infection kinetics, intracellular virus localization, viral persistence and cytokine responses. Although not as high as in Vero cells, viral titres exceeded 104 plaque-forming units (p.f.u.) ml-1 in the endothelial/neuroglial cell types, except hOECs. Despite these substantial titres, a relatively small proportion of neuroglial cells were primarily infected. Immunolabelling of infected cells revealed localization of the ZIKV envelope and NS3 proteins in the cytoplasm; NS3 staining overlapped with that of dsRNA replication intermediate and the endoplasmic reticulum (ER). Infected OECs and endothelial cells produced high levels of pro-inflammatory chemokines. Nevertheless, ZIKV was also able to establish persistent infection in hOEC and hCMEC/D3 cells. Taken together, these results provide basic insights into ZIKV infection of endothelial and neuroglial cells and will form the basis for further study of ZIKV disease mechanisms.
Collapse
Affiliation(s)
- Margit Mutso
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - James A. St John
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Griffith Institute for Drug Discovery, Griffith University, Nathan 4111, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Zheng Lung Ling
- Discipline of Pathology, Bosch Institute, Marie Bashir Institute for Infectious diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Felicity J. Burt
- National Health Laboratory Services, University of the Free State, Bloemfontein, South Africa
| | - Yee Suan Poo
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Xiang Liu
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Eva Žusinaite
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Georges E. Grau
- Vascular Immunology Unit, Discipline of Pathology, Sydney Medical School, University of Sydney, New South Wales 2050, Australia
| | - Linda Hueston
- Arbovirus Emerging Disease Unit, CIDMLS-ICPMR, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Nicholas J.C. King
- Discipline of Pathology, Bosch Institute, Marie Bashir Institute for Infectious diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Jenny A.K. Ekberg
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| | - Suresh Mahalingam
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Southport 4222, Queensland, Australia
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW The present review will outline neuroprotective and neurotoxic effects of central nervous system (CNS) infiltrating T cells during viral infections. Evidence demonstrating differential roles for antiviral effector and resident memory T-cell subsets in virologic control and immunopathology in the CNS will be discussed. Potential therapeutic targets emanating from a growing understanding of T-cell-initiated neuropathology that impacts learning and memory will also be delineated. RECENT FINDINGS The critical role for T cells in preventing and clearing CNS infections became incontrovertible during the era of acquired immunodeficiency syndrome. Recent studies have further defined differential roles of T-cell subsets, including resident memory T cells (Trm), in antiviral immunity and, unexpectedly, in postinfectious cognitive dysfunction. Mechanisms of T-cell-mediated effects include differential innate immune signaling within neural cells that are virus-specific. SUMMARY T-cell cytokines that are essential for cell-mediated virologic control during neurotropic viral infections have recently been identified as potential targets to prevent post-infection memory disorders. Further identification of T-cell subsets, their antigen specificity, and postinfection localization of Trm will enhance the efficacy of immunotherapies through minimization of immunopathology.
Collapse
Affiliation(s)
| | - Robyn S. Klein
- Departments of Medicine
- Pathology and Immunology
- Neurosciences Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
40
|
Ledur PF, Karmirian K, Pedrosa CDSG, Souza LRQ, Assis-de-Lemos G, Martins TM, Ferreira JDCCG, de Azevedo Reis GF, Silva ES, Silva D, Salerno JA, Ornelas IM, Devalle S, Madeiro da Costa RF, Goto-Silva L, Higa LM, Melo A, Tanuri A, Chimelli L, Murata MM, Garcez PP, Filippi-Chiela EC, Galina A, Borges HL, Rehen SK. Zika virus infection leads to mitochondrial failure, oxidative stress and DNA damage in human iPSC-derived astrocytes. Sci Rep 2020; 10:1218. [PMID: 31988337 PMCID: PMC6985105 DOI: 10.1038/s41598-020-57914-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/02/2020] [Indexed: 12/14/2022] Open
Abstract
Zika virus (ZIKV) has been extensively studied since it was linked to congenital malformations, and recent research has revealed that astrocytes are targets of ZIKV. However, the consequences of ZIKV infection, especially to this cell type, remain largely unknown, particularly considering integrative studies aiming to understand the crosstalk among key cellular mechanisms and fates involved in the neurotoxicity of the virus. Here, the consequences of ZIKV infection in iPSC-derived astrocytes are presented. Our results show ROS imbalance, mitochondrial defects and DNA breakage, which have been previously linked to neurological disorders. We have also detected glial reactivity, also present in mice and in post-mortem brains from infected neonates from the Northeast of Brazil. Given the role of glia in the developing brain, these findings may help to explain the observed effects in congenital Zika syndrome related to neuronal loss and motor deficit.
Collapse
Affiliation(s)
| | - Karina Karmirian
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | | | - Gabriela Assis-de-Lemos
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thiago Martino Martins
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Gabriel Ferreira de Azevedo Reis
- Insitute of Biology, Department of Biophysics and Biometrics, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Eduardo Santos Silva
- Insitute of Biology, Department of Biophysics and Biometrics, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Débora Silva
- Laboratory of Neuropathology, State Institute of Brain Paulo Niemeyer, Rio de Janeiro, RJ, Brazil
| | - José Alexandre Salerno
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Sylvie Devalle
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | | | - Livia Goto-Silva
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Luiza Mendonça Higa
- Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Adriana Melo
- Research Institute Prof. Joaquim Amorim Neto (IPESQ), Campina Grande, PB, Brazil
| | - Amilcar Tanuri
- Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Leila Chimelli
- Laboratory of Neuropathology, State Institute of Brain Paulo Niemeyer, Rio de Janeiro, RJ, Brazil
| | - Marcos Massao Murata
- Insitute of Biology, Department of Biophysics and Biometrics, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Patrícia Pestana Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Antonio Galina
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Helena Lobo Borges
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Stevens Kastrup Rehen
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
41
|
Braz-De-Melo HA, Pasquarelli-do-Nascimento G, Corrêa R, das Neves Almeida R, de Oliveira Santos I, Prado PS, Picolo V, de Bem AF, Pizato N, Magalhães KG. Potential neuroprotective and anti-inflammatory effects provided by omega-3 (DHA) against Zika virus infection in human SH-SY5Y cells. Sci Rep 2019; 9:20119. [PMID: 31882804 PMCID: PMC6984748 DOI: 10.1038/s41598-019-56556-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
Zika virus (ZIKV) has a strong tropism for the nervous system and has been related to post-infection neurological syndromes. Once neuronal cells are infected, the virus is capable of modulating cell metabolism, leading to neurotoxicity and cellular death. The negative effect of ZIKV in neuron cells has been characterized. However, the description of molecules capable of reversing these cytotoxic effects is still under investigation. In this context, it has been largely demonstrated that docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid, is highly neuroprotective. Here, we hypothesized that DHA's neuroprotective proprieties could have an influence on ZIKV-induced neurotoxicity in SH-SY5Y cells. Our data showed that pre-treatment of SH-SY5Y cells with DHA increased the cell viability and proliferation in ZIKV-infected cells. Moreover, DHA triggered an anti-inflammatory response in those infected cells. Besides, DHA was capable of restoring mitochondria function and number in ZIKV-infected SH-SY5Y cells. In addition, cells pre-treated with DHA prior to ZIKV infection presented a lower viral load at different times of infection. Taking together, these results demonstrated that DHA has a potential anti-inflammatory and neuroprotective effect against ZIKV infection in these neuron-like cells and could be a useful tool in the treatment against this virus.
Collapse
Affiliation(s)
- Heloísa Antoniella Braz-De-Melo
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília (UnB), 70910-900, Brasilia, Brazil
| | | | - Rafael Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília (UnB), 70910-900, Brasilia, Brazil
| | - Raquel das Neves Almeida
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília (UnB), 70910-900, Brasilia, Brazil
| | - Igor de Oliveira Santos
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília (UnB), 70910-900, Brasilia, Brazil
| | - Paulo Sousa Prado
- Central Laboratory of Federal District (LACEN), 70830-010, Brasilia, Brazil
| | - Victor Picolo
- Department of Physiological Sciences, University of Brasília (UnB), 70910-900, Brasilia, Brazil
| | - Andreza Fabro de Bem
- Department of Physiological Sciences, University of Brasília (UnB), 70910-900, Brasilia, Brazil
| | - Nathalia Pizato
- Department of Nutrition, University of Brasília (UnB), 70910-900, Brasilia, Brazil
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasília (UnB), 70910-900, Brasilia, Brazil.
| |
Collapse
|
42
|
Pokorna Formanova P, Palus M, Salat J, Hönig V, Stefanik M, Svoboda P, Ruzek D. Changes in cytokine and chemokine profiles in mouse serum and brain, and in human neural cells, upon tick-borne encephalitis virus infection. J Neuroinflammation 2019; 16:205. [PMID: 31699097 PMCID: PMC6839073 DOI: 10.1186/s12974-019-1596-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022] Open
Abstract
Background Tick-borne encephalitis (TBE) is a severe neuropathological disorder caused by tick-borne encephalitis virus (TBEV). Brain TBEV infection is characterized by extensive pathological neuroinflammation. The mechanism by which TBEV causes CNS destruction remains unclear, but growing evidence suggests that it involves both direct neuronal damage by the virus infection and indirect damage caused by the immune response. Here, we aimed to examine the TBEV-infection-induced innate immune response in mice and in human neural cells. We also compared cytokine/chemokine communication between naïve and infected neuronal cells and astrocytes. Methods We used a multiplexed Luminex system to measure multiple cytokines/chemokines and growth factors in mouse serum samples and brain tissue, and in human neuroblastoma cells (SK-N-SH) and primary cortical astrocytes (HBCA), which were infected with the highly pathogenic TBEV strain Hypr. We also investigated changes in cytokine/chemokine production in naïve HBCA cells treated with virus-free supernatants from TBEV-infected SK-N-SH cells and in naïve SK-N-SH cells treated with virus-free supernatants from TBEV-infected HBCA cells. Additionally, a plaque assay was performed to assess how cytokine/chemokine treatment influenced viral growth following TBEV infection. Results TBEV-infected mice exhibited time-dependent increases in serum and brain tissue concentrations of multiple cytokines/chemokines (mainly CXCL10/IP-10, and also CXCL1, G-CSF, IL-6, and others). TBEV-infected SK-N-SH cells exhibited increased production of IL-8 and RANTES and downregulated MCP-1 and HGF. TBEV infection of HBCA cells activated production of a broad spectrum of pro-inflammatory cytokines, chemokines, and growth factors (mainly IL-6, IL-8, CXCL10, RANTES, and G-CSF) and downregulated the expression of VEGF. Treatment of SK-N-SH with supernatants from infected HBCA induced expression of a variety of chemokines and pro-inflammatory cytokines, reduced SK-N-SH mortality after TBEV infection, and decreased virus growth in these cells. Treatment of HBCA with supernatants from infected SK-N-SH had little effect on cytokine/chemokine/growth factor expression but reduced TBEV growth in these cells after infection. Conclusions Our results indicated that both neurons and astrocytes are potential sources of pro-inflammatory cytokines in TBEV-infected brain tissue. Infected/activated astrocytes produce cytokines/chemokines that stimulate the innate neuronal immune response, limiting virus replication, and increasing survival of infected neurons.
Collapse
Affiliation(s)
- Petra Pokorna Formanova
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic
| | - Martin Palus
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005, Ceske Budejovice, Czech Republic
| | - Jiri Salat
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic
| | - Vaclav Hönig
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005, Ceske Budejovice, Czech Republic
| | - Michal Stefanik
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic.,Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-61300, Brno, Czech Republic
| | - Pavel Svoboda
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic
| | - Daniel Ruzek
- Department of Virology, Veterinary Research Institute, Hudcova 70, CZ-62100, Brno, Czech Republic. .,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005, Ceske Budejovice, Czech Republic.
| |
Collapse
|
43
|
Salgado DM, Vega R, Rodríguez JA, Niño Á, Rodríguez R, Ortiz Á, DeLaura I, Bosch I, Narváez CF. Clinical, laboratory and immune aspects of Zika virus-associated encephalitis in children. Int J Infect Dis 2019; 90:104-110. [PMID: 31678190 DOI: 10.1016/j.ijid.2019.10.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE To evaluate the clinical, laboratory, and immune characteristics of Zika virus (ZIKV)-associated encephalitis in pediatric patients after the epidemic in Huila, southern Colombia. METHODS A pediatric neuro-surveillance hospital study was conducted in a referral health center in southern Colombia, from October 2016 to October 2017. Cases of encephalitis were confirmed by nucleic acid amplification tests and serological methods in cerebrospinal fluid (CSF), plasma, and/or urine. Levels of six cytokines were evaluated by flow cytometry. Patients underwent daily clinical and laboratory follow-up. RESULTS Twenty children with probable encephalitis were included for further studies and 16 of them were confirmed. Four cases of bacterial meningoencephalitis (Streptococcus pneumoniae, group B Streptococcus, Staphylococcus epidermidis, and Escherichia coli) and 12 cases of viral encephalitis were identified, six of them associated with ZIKV infection. Other viral encephalitis cases were caused by herpes viruses (n=3), enterovirus (n=2), and dengue virus type 2 (DENV-2; n=1) infections. ZIKV-associated encephalitis symptoms subsided faster than those of patients with encephalitis caused by other agents. CSF analysis revealed lymphocytic pleocytosis. Compared to healthy controls, children with ZIKV-associated encephalitis presented modest plasma interleukin (IL)-10 but not IL-2, IL-4, IL-6, interferon gamma (IFN-γ), or tumor necrosis factor alpha (TNF-α). Cytokine expression was differentially regulated, as dramatically elevated IL-6, IL-10, and IFN-γ levels were observed in CSF but not in paired plasma samples in one of the patients with ZIKV detectable in CSF. CONCLUSIONS This study provides evidence that ZIKV is responsible for pediatric encephalitis in endemic areas, and the local presence of the virus may induce cephalic but not systemic expression of cytokines.
Collapse
Affiliation(s)
- Doris M Salgado
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Departamento de Pediatría, Hospital Universitario de Neiva, Neiva, Huila, Colombia; Especialización Médica en Pediatría, Postgrados Clínicos, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia.
| | - Rocío Vega
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Departamento de Pediatría, Hospital Universitario de Neiva, Neiva, Huila, Colombia; Especialización Médica en Pediatría, Postgrados Clínicos, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Jairo Antonio Rodríguez
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Departamento de Pediatría, Hospital Universitario de Neiva, Neiva, Huila, Colombia; Especialización Médica en Pediatría, Postgrados Clínicos, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Ángela Niño
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Departamento de Pediatría, Hospital Universitario de Neiva, Neiva, Huila, Colombia; Especialización Médica en Pediatría, Postgrados Clínicos, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Rocío Rodríguez
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Departamento de Pediatría, Hospital Universitario de Neiva, Neiva, Huila, Colombia; Especialización Médica en Pediatría, Postgrados Clínicos, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Ángela Ortiz
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Departamento de Pediatría, Hospital Universitario de Neiva, Neiva, Huila, Colombia; Especialización Médica en Pediatría, Postgrados Clínicos, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Isabel DeLaura
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Harvard College, Cambridge, MA 02138, USA
| | - Irene Bosch
- E25Bio Inc., The Engine of MIT, 501 Massachusetts Ave., Cambridge, MA 02139, USA; Mount Sinai School of Medicine, Department of Medicine, New York, NY 10029-6500, USA
| | - Carlos F Narváez
- Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia; Especialización Médica en Pediatría, Postgrados Clínicos, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia.
| |
Collapse
|
44
|
ZIKV Strains Differentially Affect Survival of Human Fetal Astrocytes versus Neurons and Traffic of ZIKV-Laden Endocytotic Compartments. Sci Rep 2019; 9:8069. [PMID: 31147629 PMCID: PMC6542792 DOI: 10.1038/s41598-019-44559-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 04/23/2019] [Indexed: 01/05/2023] Open
Abstract
Malformations of the fetal CNS, known as microcephaly, have been linked to Zika virus (ZIKV) infection. Here, the responses of mammalian and mosquito cell lines, in addition to primary human fetal astrocytes and neurons were studied following infection by ZIKV strains Brazil 2016 (ZIKV-BR), French Polynesia 2013 (ZIKV-FP), and Uganda #976 1947 (ZIKV-UG). Viral production, cell viability, infectivity rate, and mobility of endocytotic ZIKV-laden vesicles were compared. All cell types (SK-N-SH, Vero E6, C6/36, human fetal astrocytes and human fetal neurons) released productive virus. Among primary cells, astrocytes were more susceptible to ZIKV infection than neurons, released more progeny virus and tolerated higher virus loads than neurons. In general, the infection rate of ZIKV-UG strain was the highest. All ZIKV strains elicited differences in trafficking of ZIKV-laden endocytotic vesicles in the majority of cell types, including astrocytes and neurons, except in mosquito cells, where ZIKV infection failed to induce cell death. These results represent a thorough screening of cell viability, infection and production of three ZIKV strains in five different cell types and demonstrate that ZIKV affects vesicle mobility in all but mosquito cells.
Collapse
|
45
|
Alfano C, Gladwyn-Ng I, Couderc T, Lecuit M, Nguyen L. The Unfolded Protein Response: A Key Player in Zika Virus-Associated Congenital Microcephaly. Front Cell Neurosci 2019; 13:94. [PMID: 30971894 PMCID: PMC6445045 DOI: 10.3389/fncel.2019.00094] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/26/2019] [Indexed: 01/08/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that belongs to the Flaviviridae family, together with dengue, yellow fever, and West Nile viruses. In the wake of its emergence in the French Polynesia and in the Americas, ZIKV has been shown to cause congenital microcephaly. It is the first arbovirus which has been proven to be teratogenic and sexually transmissible. Confronted with this major public health challenge, the scientific and medical communities teamed up to precisely characterize the clinical features of congenital ZIKV syndrome and its underlying pathophysiological mechanisms. This review focuses on the critical impact of the unfolded protein response (UPR) on ZIKV-associated congenital microcephaly. ZIKV infection of cortical neuron progenitors leads to high endoplasmic reticulum (ER) stress. This results in both the stalling of indirect neurogenesis, and UPR-dependent neuronal apoptotic death, and leads to cortical microcephaly. In line with these results, the administration of molecules inhibiting UPR prevents ZIKV-induced cortical microcephaly. The discovery of the link between ZIKV infection and UPR activation has a broader relevance, since this pathway plays a crucial role in many distinct cellular processes and its induction by ZIKV may account for several reported ZIKV-associated defects.
Collapse
Affiliation(s)
- Christian Alfano
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège, Belgium
| | - Ivan Gladwyn-Ng
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège, Belgium
| | - Thérèse Couderc
- Institut Pasteur, Biology of Infection Unit, Paris, France.,INSERM U1117, Biologie des Infections, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France.,INSERM U1117, Biologie des Infections, Paris, France.,Paris Descartes University, Division of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades Hospital, Institut Imagine, Sorbonne Paris Cité, Paris, France
| | - Laurent Nguyen
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Liège, Belgium
| |
Collapse
|
46
|
Lee JK, Shin OS. Advances in Zika Virus⁻Host Cell Interaction: Current Knowledge and Future Perspectives. Int J Mol Sci 2019; 20:ijms20051101. [PMID: 30836648 PMCID: PMC6429326 DOI: 10.3390/ijms20051101] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 12/12/2022] Open
Abstract
Emerging mosquito-transmitted RNA viruses, such as Zika virus (ZIKV) and Chikungunya represent human pathogens of an immense global health problem. In particular, ZIKV has emerged explosively since 2007 to cause a series of epidemics in the South Pacific and most recently in the Americas. Although typical ZIKV infections are asymptomatic, ZIKV infection during pregnancy is increasingly associated with microcephaly and other fetal developmental abnormalities. In the last few years, genomic and molecular investigations have established a remarkable progress on the pathogenic mechanisms of ZIKV infection using in vitro and in vivo models. Here, we highlight recent advances in ZIKV-host cell interaction studies, including cellular targets of ZIKV, ZIKV-mediated cell death mechanisms, host cell restriction factors that limit ZIKV replication, and immune evasion mechanisms utilized by ZIKV. Understanding of the mechanisms of ZIKV⁻host interaction at the cellular level will contribute crucial insights into the development of ZIKV therapeutics and vaccines.
Collapse
Affiliation(s)
- Jae Kyung Lee
- Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Korea.
| | - Ok Sarah Shin
- Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Korea.
| |
Collapse
|
47
|
Ojha CR, Rodriguez M, Karuppan MKM, Lapierre J, Kashanchi F, El-Hage N. Toll-like receptor 3 regulates Zika virus infection and associated host inflammatory response in primary human astrocytes. PLoS One 2019; 14:e0208543. [PMID: 30735502 PMCID: PMC6368285 DOI: 10.1371/journal.pone.0208543] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
The connection between Zika virus (ZIKV) and neurodevelopmental defects is widely recognized, although the mechanisms underlying the infectivity and pathology in primary human glial cells are poorly understood. Here we show that three isolated strains of ZIKV, an African strain MR766 (Uganda) and two closely related Asian strains R103451 (Honduras) and PRVABC59 (Puerto Rico) productively infect primary human astrocytes, although Asian strains showed a higher infectivity rate and increased cell death when compared to the African strain. Inhibition of AXL receptor significantly attenuated viral entry of MR766 and PRVABC59 and to a lesser extend R103451, suggesting an important role of TAM receptors in ZIKV cell entry, irrespective of lineage. Infection by PRVABC59 elicited the highest release of inflammatory molecules, with a 8-fold increase in the release of RANTES, 10-fold increase in secretion of IP-10 secretion and a 12-fold increase in IFN-β secretion when compared to un-infected human astrocytes. Minor changes in the release of several growth factors, endoplasmic reticulum (ER)-stress response factors and the transcription factor, NF-κB were detected with the Asian strains, while significant increases in FOXO6, MAPK10 and JNK were detected with the African strain. Activation of the autophagy pathway was evident with increased expression of the autophagy related proteins Beclin1, LC3B and p62/SQSTM1 with all three strains of ZIKV. Pharmacological inhibition of the autophagy pathway and genetic inhibition of the Beclin1 showed minimal effects on ZIKV replication. The expression of toll-like receptor 3 (TLR3) was significantly increased with all three strains of ZIKV; pharmacological and genetic inhibition of TLR3 caused a decrease in viral titers and in viral-induced inflammatory response in infected astrocytes. We conclude that TLR3 plays a vital role in both ZIKV replication and viral-induced inflammatory responses, irrespective of the strains, while the autophagy protein Beclin1 influences host inflammatory responses.
Collapse
Affiliation(s)
- Chet Raj Ojha
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Myosotys Rodriguez
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Mohan Kumar Muthu Karuppan
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Jessica Lapierre
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Fatah Kashanchi
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Nazira El-Hage
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| |
Collapse
|
48
|
Potokar M, Jorgačevski J, Zorec R. Astrocytes in Flavivirus Infections. Int J Mol Sci 2019; 20:ijms20030691. [PMID: 30736273 PMCID: PMC6386967 DOI: 10.3390/ijms20030691] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
Virus infections of the central nervous system (CNS) can manifest in various forms of inflammation, including that of the brain (encephalitis) and spinal cord (myelitis), all of which may have long-lasting deleterious consequences. Although the knowledge of how different viruses affect neural cells is increasing, understanding of the mechanisms by which cells respond to neurotropic viruses remains fragmented. Several virus types have the ability to infect neural tissue, and astrocytes, an abundant and heterogeneous neuroglial cell type and a key element providing CNS homeostasis, are one of the first CNS cell types to get infected. Astrocytes are morphologically closely aligned with neuronal synapses, blood vessels, and ventricle cavities, and thereby have the capacity to functionally interact with neurons and endothelial cells. In this review, we focus on the responses of astrocytes to infection by neurotropic flaviviruses, including tick-borne encephalitis virus (TBEV), Zika virus (ZIKV), West Nile virus (WNV), and Japanese encephalitis virus (JEV), which have all been confirmed to infect astrocytes and cause multiple CNS defects. Understanding these mechanisms may help design new strategies to better contain and mitigate virus- and astrocyte-dependent neuroinflammation.
Collapse
Affiliation(s)
- Maja Potokar
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| |
Collapse
|
49
|
Bubak AN, Como CN, Blackmon AM, Jones D, Nagel MA. Varicella zoster virus differentially alters morphology and suppresses proinflammatory cytokines in primary human spinal cord and hippocampal astrocytes. J Neuroinflammation 2018; 15:318. [PMID: 30442152 PMCID: PMC6236967 DOI: 10.1186/s12974-018-1360-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/05/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Varicella zoster virus (VZV) is a ubiquitous alphaherpesvirus that produces varicella and zoster. VZV can infect multiple cell types in the spinal cord and brain, including astrocytes, producing myelopathy and encephalopathy. While studies of VZV-astrocyte interactions are sparse, a recent report showed that quiescent primary human spinal cord astrocytes (qHA-sps) did not appear activated morphologically during VZV infection. Since astrocytes play a critical role in host defenses during viral infections of the central nervous system, we examined the cytokine responses of qHA-sps and quiescent primary human hippocampal astrocytes (qHA-hps) to VZV infection in vitro, as well as the ability of conditioned supernatant to recruit immune cells. METHODS At 3 days post-infection, mock- and VZV-infected qHA-sps and qHA-hps were examined for morphological changes by immunofluorescence antibody assay using antibodies directed against glial fibrillary acidic protein and VZV. Conditioned supernatants were analyzed for proinflammatory cytokines [interleukin (IL)-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, interferon-gamma, and tumor necrosis factor-α] using the Meso Scale Discovery multiplex ELISA platform. Finally, the ability of conditioned supernatants to attract peripheral blood mononuclear cells (PBMCs) was determined using a chemotaxis assay. Quiescent primary human perineurial cells (qHPNCs) served as a control for VZV-induced cytokine production and PBMC migration. To confirm that the astrocytes have the ability to increase cytokine secretion, qHA-sps and qHA-hps were treated with IL-1β and examined for morphological changes and IL-6 secretion. RESULTS VZV-infected qHA-sps displayed extensive cellular processes, whereas VZV-infected qHA-hps became swollen and clustered together. Astrocytes had the capacity to secrete IL-6 in response to IL-1β. Compared to mock-infected cells, VZV-infected qHA-sps showed significantly reduced secretion of IL-2, IL-4, IL-6, IL-12p70, and IL-13, while VZV-infected qHA-hps showed significantly reduced IL-8 secretion. In contrast, levels of all 10 cytokines examined were significantly increased in VZV-infected qHPNCs. Consistent with these results, conditioned supernatant from VZV-infected qHPNCs, but not that from VZV-infected qHA-sps and qHA-hps, recruited PBMCs. CONCLUSIONS VZV-infected qHA-sps and qHA-hps have distinct morphological alterations and patterns of proinflammatory cytokine suppression that could contribute to ineffective viral clearance in VZV myelopathy and encephalopathy, respectively.
Collapse
Affiliation(s)
- Andrew N. Bubak
- Department of Neurology, University of Colorado School of Medicine, 4200 E. 19th Avenue, Mail Stop B182, Aurora, CO 80045 USA
| | - Christina N. Como
- Department of Neurology, University of Colorado School of Medicine, 4200 E. 19th Avenue, Mail Stop B182, Aurora, CO 80045 USA
| | - Anna M. Blackmon
- Department of Neurology, University of Colorado School of Medicine, 4200 E. 19th Avenue, Mail Stop B182, Aurora, CO 80045 USA
| | - Dallas Jones
- Department of Neurology, University of Colorado School of Medicine, 4200 E. 19th Avenue, Mail Stop B182, Aurora, CO 80045 USA
| | - Maria A. Nagel
- Department of Neurology, University of Colorado School of Medicine, 4200 E. 19th Avenue, Mail Stop B182, Aurora, CO 80045 USA
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045 USA
| |
Collapse
|
50
|
Alves MP, Vielle NJ, Thiel V, Pfaender S. Research Models and Tools for the Identification of Antivirals and Therapeutics against Zika Virus Infection. Viruses 2018; 10:v10110593. [PMID: 30380760 PMCID: PMC6265910 DOI: 10.3390/v10110593] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022] Open
Abstract
Zika virus recently re-emerged and caused global outbreaks mainly in Central Africa, Southeast Asia, the Pacific Islands and in Central and South America. Even though there is a declining trend, the virus continues to spread throughout different geographical regions of the world. Since its re-emergence in 2015, massive advances have been made regarding our understanding of clinical manifestations, epidemiology, genetic diversity, genomic structure and potential therapeutic intervention strategies. Nevertheless, treatment remains a challenge as there is no licensed effective therapy available. This review focuses on the recent advances regarding research models, as well as available experimental tools that can be used for the identification and characterization of potential antiviral targets and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Marco P Alves
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Nathalie J Vielle
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland.
| | - Volker Thiel
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Stephanie Pfaender
- Institute of Virology and Immunology, 3012 Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| |
Collapse
|