51
|
Yang R, Liu Q, Pang W, Gao F, Liang H, Zhang W, Lin Y, Li M, Liu Z, Gao GF, Zhang L, Xiao H, Zheng Y, Huang Z, Jin X. Two immunogenic recombinant protein vaccine candidates showed disparate protective efficacy against Zika virus infection in rhesus macaques. Vaccine 2021; 39:915-925. [PMID: 33451779 DOI: 10.1016/j.vaccine.2020.12.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/27/2022]
Abstract
Zika virus (ZIKV) infection has caused major public health problems recently. To develop subunit vaccines for ZIKV, we have previously constructed recombinant ZIKV envelope protein domain III (EDIII), and the entire ectodomain (E80, which comprises EDI, EDII and EDIII), as vaccine candidates and showed both of them being immunogenic and protective in murine models. In this follow-up study, we compared these vaccine candidates in non-human primates. Both of them elicited neutralizing antibody responses, but only E80 immunization inhibited ZIKV infection in both peripheral blood and monkey tissues, whereas EDIII increased blood ZIKV RNA through possibly antibody-dependent enhancement. Further investigations revealed that the virion-binding antibody response in E80 immunized monkeys persisted longer and stronger than in EDIII immunized monkeys. These results demonstrate that E80 is superior to EDIII as a vaccine candidate, and that the magnitude, quality and durability of virion-binding neutralizing antibodies are correlates of protection.
Collapse
Affiliation(s)
- Ruoheng Yang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Qingwei Liu
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Pang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Fei Gao
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Huabin Liang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yalong Lin
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Min Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhihua Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - George F Gao
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Linqi Zhang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Hui Xiao
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yongtang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhong Huang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Xia Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|
52
|
Yang W, Wu YH, Liu SQ, Sheng ZY, Zhen ZD, Gao RQ, Cui XY, Fan DY, Qin ZH, Zheng AH, Wang PG, An J. S100A4+ macrophages facilitate zika virus invasion and persistence in the seminiferous tubules via interferon-gamma mediation. PLoS Pathog 2020; 16:e1009019. [PMID: 33315931 PMCID: PMC7769614 DOI: 10.1371/journal.ppat.1009019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/28/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Testicular invasion and persistence are features of Zika virus (ZIKV), but their mechanisms are still unknown. Here, we showed that S100A4+ macrophages, a myeloid macrophage subpopulation with susceptibility to ZIKV infection, facilitated ZIKV invasion and persistence in the seminiferous tubules. In ZIKV-infected mice, S100A4+ macrophages were specifically recruited into the interstitial space of testes and differentiated into interferon-γ-expressing M1 macrophages. With interferon-γ mediation, S100A4+ macrophages down-regulated Claudin-1 expression and induced its redistribution from the cytosol to nucleus, thus increasing the permeability of the blood-testis barrier which facilitated S100A4+ macrophages invasion into the seminiferous tubules. Intraluminal S100A4+ macrophages were segregated from CD8+ T cells and consequently helped ZIKV evade cellular immunity. As a result, ZIKV continued to replicate in intraluminal S100A4+ macrophages even when the spermatogenic cells disappeared. Deficiencies in S100A4 or interferon-γ signaling both reduced ZIKV infection in the seminiferous tubules. These results demonstrated crucial roles of S100A4+ macrophages in ZIKV infection in testes.
Collapse
Affiliation(s)
- Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan-Hua Wu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuang-Qing Liu
- Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | - Zi-Yang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zi-Da Zhen
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Rui-Qi Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Yun Cui
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Science and Technology, Capital Institute of Pediatrics, Beijing, China
| | - Dong-Ying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhi-Hai Qin
- Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | - Ai-Hua Zheng
- Institute of Zoology, Chinese Academy of Science, Beijing, China
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- * E-mail: (PGW); , (JA)
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
- * E-mail: (PGW); , (JA)
| |
Collapse
|
53
|
NS5 Sumoylation Directs Nuclear Responses That Permit Zika Virus To Persistently Infect Human Brain Microvascular Endothelial Cells. J Virol 2020; 94:JVI.01086-20. [PMID: 32699085 DOI: 10.1128/jvi.01086-20] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV) is cytopathic to neurons and persistently infects brain microvascular endothelial cells (hBMECs), which normally restrict viral access to neurons. Despite replicating in the cytoplasm, ZIKV and Dengue virus (DENV) polymerases, NS5 proteins, are predominantly trafficked to the nucleus. We found that a SUMO interaction motif in ZIKV and DENV NS5 proteins directs nuclear localization. However, ZIKV NS5 formed discrete punctate nuclear bodies (NBs), while DENV NS5 was uniformly dispersed in the nucleoplasm. Yet, mutating one DENV NS5 SUMO site (K546R) localized the NS5 mutant to discrete NBs, and NBs formed by the ZIKV NS5 SUMO mutant (K252R) were restructured into discrete protein complexes. In hBMECs, NBs formed by STAT2 and promyelocytic leukemia (PML) protein are present constitutively and enhance innate immunity. During ZIKV infection or NS5 expression, we found that ZIKV NS5 evicts PML from STAT2 NBs, forming NS5/STAT2 NBs that dramatically reduce PML expression in hBMECs and inhibit the transcription of interferon-stimulated genes (ISG). Expressing the ZIKV NS5 SUMO site mutant (K252R) resulted in NS5/STAT2/PML NBs that failed to degrade PML, reduce STAT2 expression, or inhibit ISG induction. Additionally, the K252 SUMOylation site and NS5 nuclear localization were required for ZIKV NS5 to regulate hBMEC cell cycle transcriptional responses. Our data reveal NS5 SUMO motifs as novel NB coordinating factors that distinguish flavivirus NS5 proteins. These findings establish SUMOylation of ZIKV NS5 as critical in the regulation of antiviral ISG and cell cycle responses that permit ZIKV to persistently infect hBMECs.IMPORTANCE ZIKV is a unique neurovirulent flavivirus that persistently infects human brain microvascular endothelial cells (hBMECs), the primary barrier that restricts viral access to neuronal compartments. Here, we demonstrate that flavivirus-specific SIM and SUMO sites determine the assembly of NS5 proteins into discrete nuclear bodies (NBs). We found that NS5 SIM sites are required for NS5 nuclear localization and that SUMO sites regulate NS5 NB complex constituents, assembly, and function. We reveal that ZIKV NS5 SUMO sites direct NS5 binding to STAT2, disrupt the formation of antiviral PML-STAT2 NBs, and direct PML degradation. ZIKV NS5 SUMO sites also transcriptionally regulate cell cycle and ISG responses that permit ZIKV to persistently infect hBMECs. Our findings demonstrate the function of SUMO sites in ZIKV NS5 NB formation and their importance in regulating nuclear responses that permit ZIKV to persistently infect hBMECs and thereby gain access to neurons.
Collapse
|
54
|
Rodrigues de Sousa J, Azevedo RDSDS, Quaresma JAS, Vasconcelos PFDC. The innate immune response in Zika virus infection. Rev Med Virol 2020; 31:e2166. [PMID: 32926478 DOI: 10.1002/rmv.2166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 11/06/2022]
Abstract
Zika virus (ZIKV; Flaviviridae, Flavivirus) was discovered in 1947 in Uganda, Africa, from the serum of a sentinel Rhesus monkey (Macaca mulatta). It is an enveloped, positive-sense, single-stranded RNA virus, which encodes a single polyprotein that is cleaved into 10 individual proteins. In 2015, the Zika-epidemic in Brazil was marked mainly by the exponential growth of microcephaly cases and other congenital defects. With regard to host-pathogen relationships, understanding the role of the immune response in the pathogenesis ZIKV infection is challenging. The innate immune response is the first-line immunological defence, in which pathogen-associated molecular patterns are recognized by pattern-recognition receptors that trigger macrophages, dendritic cells, natural killer cells and endothelial cells to produce several mediators, which modulate viral replication and immune evasion. In this review, we have summarized current knowledge on the innate immune response against ZIKV.
Collapse
Affiliation(s)
- Jorge Rodrigues de Sousa
- Departamento de Arbovirologia e Febres Hemorrágicas, Instituto Evandro Chagas, Ananindeua, Brazil.,Universidade do Estado do Pará, Belém, Brazil
| | | | - Juarez Antônio Simões Quaresma
- Universidade do Estado do Pará, Belém, Brazil.,Departamento de Patologia, Instituto Evandro Chagas, Ananindeua, Brazil.,Núcleo de Medicina Tropical, Universidade Federal do Pará, Belém, Brazil
| | - Pedro Fernando da Costa Vasconcelos
- Departamento de Arbovirologia e Febres Hemorrágicas, Instituto Evandro Chagas, Ananindeua, Brazil.,Universidade do Estado do Pará, Belém, Brazil
| |
Collapse
|
55
|
Abstract
The blood-brain barrier (BBB), which protects the CNS from pathogens, is composed of specialized brain microvascular endothelial cells (BMECs) joined by tight junctions and ensheathed by pericytes and astrocyte endfeet. The stability of the BBB structure and function is of great significance for the maintenance of brain homeostasis. When a neurotropic virus invades the CNS via a hematogenous or non-hematogenous route, it may cause structural and functional disorders of the BBB, and also activate the BBB anti-inflammatory or pro-inflammatory innate immune response. This article focuses on the structural and functional changes that occur in the three main components of the BBB (endothelial cells, astrocytes, and pericytes) in response to infection with neurotropic viruses transmitted by hematogenous routes, and also briefly describes the supportive effect of three cells on the BBB under normal physiological conditions. For example, all three types of cells express several PRRs, which can quickly sense the virus and make corresponding immune responses. The pro-inflammatory immune response will exacerbate the destruction of the BBB, while the anti-inflammatory immune response, based on type I IFN, consolidates the stability of the BBB. Exploring the details of the interaction between the host and the pathogen at the BBB during neurotropic virus infection will help to propose new treatments for viral encephalitis. Enhancing the defense function of the BBB, maintaining the integrity of the BBB, and suppressing the pro-inflammatory immune response of the BBB provide more ideas for limiting the neuroinvasion of neurotropic viruses. In the future, these new treatments are expected to cooperate with traditional antiviral methods to improve the therapeutic effect of viral encephalitis.
Collapse
Affiliation(s)
- Zhuangzhuang Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, People's Republic of China
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, People's Republic of China
| |
Collapse
|
56
|
Zika Virus Infection Promotes Local Inflammation, Cell Adhesion Molecule Upregulation, and Leukocyte Recruitment at the Blood-Brain Barrier. mBio 2020; 11:mBio.01183-20. [PMID: 32753493 PMCID: PMC7407083 DOI: 10.1128/mbio.01183-20] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The blood-brain barrier (BBB) largely prevents toxins and pathogens from accessing the brain. Some viruses have the ability to cross this barrier and replicate in the central nervous system (CNS). Zika virus (ZIKV) was responsible in 2015 to 2016 for a major epidemic in South America and was associated in some cases with neurological impairments. Here, we characterized some of the mechanisms behind its neuroinvasion using an innovative in vitro human BBB model. ZIKV efficiently replicated, was released on the BBB parenchyma side, and triggered subtle modulation of BBB integrity as well as an upregulation of inflammatory and cell adhesion molecules (CAMs), which in turn favored leukocyte recruitment. Finally, we showed that ZIKV-infected mouse models displayed similar CAM upregulation and that soluble CAMs were increased in plasma samples from ZIKV-infected patients. Our observations suggest a complex interplay between ZIKV and the BBB, which may trigger local inflammation, leukocyte recruitment, and possible cerebral vasculature impairment.IMPORTANCE Zika virus (ZIKV) can be associated with neurological impairment in children and adults. To reach the central nervous system, viruses have to cross the blood-brain barrier (BBB), a multicellular system allowing a tight separation between the bloodstream and the brain. Here, we show that ZIKV infects cells of the BBB and triggers a subtle change in its permeability. Moreover, ZIKV infection leads to the production of inflammatory molecules known to modulate BBB integrity and participate in immune cell attraction. The virus also led to the upregulation of cellular adhesion molecules (CAMs), which in turn favored immune cell binding to the BBB and potentially increased infiltration into the brain. These results were also observed in a mouse model of ZIKV infection. Furthermore, plasma samples from ZIKV-infected patients displayed an increase in CAMs, suggesting that this mechanism could be involved in neuroinflammation triggered by ZIKV.
Collapse
|
57
|
Panganiban AT, Blair RV, Hattler JB, Bohannon DG, Bonaldo MC, Schouest B, Maness NJ, Kim WK. A Zika virus primary isolate induces neuroinflammation, compromises the blood-brain barrier and upregulates CXCL12 in adult macaques. Brain Pathol 2020; 30:1017-1027. [PMID: 32585067 DOI: 10.1111/bpa.12873] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/29/2020] [Accepted: 07/21/2020] [Indexed: 12/23/2022] Open
Abstract
Zika virus (ZIKV) is a flavivirus that can cause neuropathogenesis in adults and fetal neurologic malformation following the infection of pregnant women. We used a nonhuman primate model, the Indian-origin Rhesus macaque (IRM), to gain insight into virus-associated hallmarks of ZIKV-induced adult neuropathology. We find that the virus causes prevalent acute and chronic neuroinflammation and chronic disruption of the blood-brain barrier (BBB) in adult animals. ZIKV infection resulted in specific short- and long-term augmented expression of the chemokine CXCL12 in the central nervous system (CNS)of adult IRMs. Moreover, CXCL12 expression persists long after the initial viral infection is apparently cleared. CXCL12 plays a key role both in regulating lymphocyte trafficking through the BBB to the CNS and in mediating repair of damaged neural tissue including remyelination. Understanding how CXCL12 expression is controlled will likely be of central importance in the definition of ZIKV-associated neuropathology in adults.
Collapse
Affiliation(s)
- Antonito T Panganiban
- Division of Microbiology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Robert V Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Julian B Hattler
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Diana G Bohannon
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Myrna C Bonaldo
- Laboratory of Flavivirus Molecular Biology, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Blake Schouest
- Division of Microbiology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Nicholas J Maness
- Division of Microbiology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| |
Collapse
|
58
|
Rastogi M, Singh SK. Zika virus NS1 affects the junctional integrity of human brain microvascular endothelial cells. Biochimie 2020; 176:52-61. [PMID: 32640279 DOI: 10.1016/j.biochi.2020.06.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022]
Abstract
Zika virus (ZIKV) infection leads to microcephaly in newborns. Flaviviruses are known to secrete NS1 protein extracellularly and its concentration in serum directly co-relate to disease severity. The presence of ZIKV-NS1 near the brain microvascular endothelial cells (BMVECs) affects blood-brain-barrier, which is composed of tight junctions (TJs) and adherens junctions (AJs). Viruses utilize different strategies to circumvent this barrier to enter in brain. The present study demonstrated the mechanism of junctional integrity disruption in BMVECs by ZIKV-NS1 protein exposure. The Transendothelial Electrical Resistance and sodium fluorescein migration assays revealed the endothelial barrier disruption in BMVECs exposed to ZIKV-NS1 at different time (12hr and 24hr) and doses (500 ng/mL, 1000 ng/mL and 1500 ng/mL). The exposure of ZIKV-NS1 on BMVECs led to the phosphorylation of AJs and suppression of TJs through secreted ZIKV-NS1 in a bystander fashion. The activation of NADPH dependent reactive oxygen species activity and redox sensitive tyrosine kinase further increased the phosphorylation of AJs. The reduced expression of the phosphatase led to the increased phosphorylation of the AJs. The treatment with Diphenyleneiodonium chloride rescued the phosphatase and TJs expression and suppressed the expression of kinase and AJs in BMVECs exposed to ZIKV-NS1.
Collapse
Affiliation(s)
- Meghana Rastogi
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, U.P, India
| | - Sunit K Singh
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, U.P, India.
| |
Collapse
|
59
|
Pryzdial ELG, Sutherland MR, Lin BH, Horwitz M. Antiviral anticoagulation. Res Pract Thromb Haemost 2020; 4:774-788. [PMID: 32685886 PMCID: PMC7354393 DOI: 10.1002/rth2.12406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel envelope virus that causes coronavirus disease 2019 (COVID-19). Hallmarks of COVID-19 are a puzzling form of thrombophilia that has elevated D-dimer but only modest effects on other parameters of coagulopathy. This is combined with severe inflammation, often leading to acute respiratory distress and possible lethality. Coagulopathy and inflammation are interconnected by the transmembrane receptor, tissue factor (TF), which initiates blood clotting as a cofactor for factor VIIa (FVIIa)-mediated factor Xa (FXa) generation. TF also functions from within the nascent TF/FVIIa/FXa complex to trigger profound changes via protease-activated receptors (PARs) in many cell types, including SARS-CoV-2-trophic cells. Therefore, aberrant expression of TF may be the underlying basis of COVID-19 symptoms. Evidence suggests a correlation between infection with many virus types and development of clotting-related symptoms, ranging from heart disease to bleeding, depending on the virus. Since numerous cell types express TF and can act as sites for virus replication, a model envelope virus, herpes simplex virus type 1 (HSV1), has been used to investigate the uptake of TF into the envelope. Indeed, HSV1 and other viruses harbor surface TF antigen, which retains clotting and PAR signaling function. Strikingly, envelope TF is essential for HSV1 infection in mice, and the FXa-directed oral anticoagulant apixaban had remarkable antiviral efficacy. SARS-CoV-2 replicates in TF-bearing epithelial and endothelial cells and may stimulate and integrate host cell TF, like HSV1 and other known coagulopathic viruses. Combined with this possibility, the features of COVID-19 suggest that it is a TFopathy, and the TF/FVIIa/FXa complex is a feasible therapeutic target.
Collapse
Affiliation(s)
- Edward L. G. Pryzdial
- Center for InnovationCanadian Blood ServicesVancouverBCCanada
- Centre for Blood Research and Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Michael R. Sutherland
- Center for InnovationCanadian Blood ServicesVancouverBCCanada
- Centre for Blood Research and Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Bryan H. Lin
- Center for InnovationCanadian Blood ServicesVancouverBCCanada
- Centre for Blood Research and Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Marc Horwitz
- Department of Microbiology and ImmunologyUniversity of British ColumbiaVancouverBCCanada
| |
Collapse
|
60
|
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
|
61
|
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
|
62
|
Human Metapneumovirus Establishes Persistent Infection in Lung Microvascular Endothelial Cells and Primes a Th2-Skewed Immune Response. Microorganisms 2020; 8:microorganisms8060824. [PMID: 32486193 PMCID: PMC7357125 DOI: 10.3390/microorganisms8060824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/28/2020] [Indexed: 01/15/2023] Open
Abstract
Human Metapneumovirus (HMPV) is a major cause of lower respiratory tract infections. HMPV infection has been hypothesized to alter dendritic cell (DC) immune response; however, many questions regarding HMPV pathogenesis within the infected lung remain unanswered. Here, we show that HMPV productively infects human lung microvascular endothelial cells (L-HMVECs). The release of infectious virus occurs for up to more than 30 days of culture without producing overt cytopathic effects and medium derived from persistently HMPV-infected L-HMVECs (secretome) induced monocyte-derived DCs to prime naïve CD4 T-cells toward a Th2 phenotype. Moreover, we demonstrated that infected secretomes trigger DCs to up-regulate OX40L expression and OX40L neutralization abolished the pro-Th2 effect that is induced by HMPV-secretome. We clarified secretome from HMPV by size exclusion and ultracentrifugation with the aim to characterize the role of viral particles in the observed pro-Th2 effect. In both cases, the percentage of IL-4-producing cells and expression of OX40L returned at basal levels. Finally, we showed that HMPV, per se, could reproduce the ability of secretome to prime pro-Th2 DCs. These results suggest that HMPV, persistently released by L-HMVECs, might take part in the development of a skewed, pro-Th2 lung microenvironment.
Collapse
|
63
|
Kim J, Alejandro B, Hetman M, Hattab EM, Joiner J, Schroten H, Ishikawa H, Chung DH. Zika virus infects pericytes in the choroid plexus and enters the central nervous system through the blood-cerebrospinal fluid barrier. PLoS Pathog 2020; 16:e1008204. [PMID: 32357162 PMCID: PMC7194358 DOI: 10.1371/journal.ppat.1008204] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV) can infect and cause microcephaly and Zika-associated neurological complications in the developing fetal and adult brains. In terms of pathogenesis, a critical question is how ZIKV overcomes the barriers separating the brain from the circulation and gains access to the central nervous system (CNS). Despite the importance of ZIKV pathogenesis, the route ZIKV utilizes to cross CNS barriers remains unclear. Here we show that in mouse models, ZIKV-infected cells initially appeared in the periventricular regions of the brain, including the choroid plexus and the meninges, prior to infection of the cortex. The appearance of ZIKV in cerebrospinal fluid (CSF) preceded infection of the brain parenchyma. Further the brain infection was significantly attenuated by neutralization of the virus in the CSF, indicating that ZIKV in the CSF at the early stage of infection might be responsible for establishing a lethal infection of the brain. We show that cells infected by ZIKV in the choroid plexus were pericytes. Using in vitro systems, we highlight the possibility that ZIKV crosses the blood-CSF barrier by disrupting the choroid plexus epithelial layer. Taken together, our results suggest that ZIKV might exploit the blood-CSF barrier rather than the blood-brain barrier to invade the CNS.
Collapse
Affiliation(s)
- Jihye Kim
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Kentucky, United States of America
| | - Brian Alejandro
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Kentucky, United States of America
| | - Michal Hetman
- Department of Neurological Surgery, School of Medicine, University of Louisville, Kentucky, United States of America
| | - Eyas M. Hattab
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Joshua Joiner
- Centre College, Danville, Kentucky, United States of America
| | - Horst Schroten
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Dong-Hoon Chung
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Kentucky, United States of America
- Center for Predictive Medicine, School of Medicine, University of Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
64
|
Roth H, Schneider L, Eberle R, Lausen J, Modlich U, Blümel J, Baylis SA. Zika virus infection studies with CD34 + hematopoietic and megakaryocyte-erythroid progenitors, red blood cells and platelets. Transfusion 2020; 60:561-574. [PMID: 32086956 DOI: 10.1111/trf.15692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/28/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND To date, several cases of transfusion-transmitted ZIKV infections have been confirmed. Multiple studies detected prolonged occurrence of ZIKV viral RNA in whole blood as compared to plasma samples indicating potential ZIKV interaction with hematopoietic cells. Also, infection of cells from the granulocyte/macrophage lineage has been demonstrated. Patients may develop severe thrombocytopenia, microcytic anemia, and a fatal course of disease occurred in a patient with sickle cell anemia suggesting additional interference of ZIKV with erythroid and megakaryocytic cells. Therefore, we analyzed whether ZIKV propagates in or compartmentalizes with hematopoietic progenitor, erythroid, and megakaryocytic cells. METHODS ZIKV RNA replication, protein translation and infectious particle formation in hematopoietic cell lines as well as primary CD34+ HSPCs and ex vivo differentiated erythroid and megakaryocytic cells was monitored using qRT-PCR, FACS, immunofluorescence analysis and infectivity assays. Distribution of ZIKV RNA and infectious particles in spiked red blood cell (RBC) units or platelet concentrates (PCs) was evaluated. RESULTS While subsets of K562 and KU812Ep6EPO cells supported ZIKV propagation, primary CD34+ HSPCs, MEP cells, RBCs, and platelets were non-permissive for ZIKV infection. In spiking studies, ZIKV RNA was detectable for 7 days in all fractions of RBC units and PCs, however, ZIKV infectious particles were not associated with erythrocytes or platelets. CONCLUSION Viral particles from plasma or contaminating leukocytes, rather than purified CD34+ HSPCs or the cellular component of RBC units or PCs, present the greatest risk for transfusion-transmitted ZIKV infections.
Collapse
Affiliation(s)
- Hanna Roth
- Division of Virology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Lucas Schneider
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Hessen, Germany
| | - Regina Eberle
- Division of Immunology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Jörn Lausen
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Hessen, Germany.,Department of Genetics of Eukaryotes, Institute of Industrial Genetics, Stuttgart, Baden-Württemberg, Germany
| | - Ute Modlich
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Johannes Blümel
- Division of Virology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| | - Sally A Baylis
- Division of Virology, Paul-Ehrlich-Institute, Langen, Hessen, Germany
| |
Collapse
|
65
|
Ventura CV, Gois AL, Freire BO, de Almeida DC, MacKeen LD, Ventura Filho MC, Berrocal AM, Paul Chan RV, Belfort R, Maia M, Ventura LO. Fluorescein Angiography Findings in Children With Congenital Zika Syndrome. Ophthalmic Surg Lasers Imaging Retina 2020; 50:702-708. [PMID: 31755969 DOI: 10.3928/23258160-20191031-05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/25/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND OBJECTIVE To evaluate the retinal and vasculature changes in infants with congenital Zika syndrome (CZS) using fluorescein angiography (FA). PATIENTS AND METHODS This consecutive case series included six infants with CZS. FA and color fundus imaging were performed under general anesthesia in both eyes of all infants using a contact widefield digital imaging system. All color fundus images were obtained using a 130° field of view lens, and the FA images were captured using either a 130° or 80° field of view lens. The immunoglobulin M antibody capture enzyme-linked immunosorbent assay was positive for Zika virus in the cerebrospinal fluid samples of all infants. Other congenital infections were ruled out. RESULTS The mean ± standard deviation age of the infants at the time of examination was 1.4 years ± 0.1 years (range: 1.3 years to 1.5 years). Contact fundus photographs showed macular abnormalities in seven eyes (58%) and retinal vasculature changes in two eyes (17%). FA detected macular abnormalities in all 12 eyes (100%) and retinal vasculature changes in five eyes (42%). The main retinal vasculature changes were peripheral avascularity in five eyes (42%) and microvasculature abnormalities in three eyes (25%). CONCLUSION FA may be an important tool for detecting subtle macular and retinal vasculature changes in CZS. [Ophthalmic Surg Lasers Imaging Retina. 2019;50:702-708.].
Collapse
|
66
|
Garcia G, Paul S, Beshara S, Ramanujan VK, Ramaiah A, Nielsen-Saines K, Li MMH, French SW, Morizono K, Kumar A, Arumugaswami V. Hippo Signaling Pathway Has a Critical Role in Zika Virus Replication and in the Pathogenesis of Neuroinflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:844-861. [PMID: 32035058 DOI: 10.1016/j.ajpath.2019.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/22/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Zika virus (ZIKV) is a reemerging human pathogen that causes congenital abnormalities, including microcephaly and eye disease. The cellular/molecular basis of ZIKV and host interactions inducing ocular and neuronal pathogenesis are unclear. Herein, we noted that the Hippo/Salvador-Warts-Hippo signaling pathway, which controls organ size through progenitor cell proliferation and differentiation, is dysregulated after ZIKV infection. In human fetal retinal pigment epithelial cells, there is an early induction of transcriptional coactivator, Yes-associated protein (YAP), which is later degraded with a corresponding activation of the TANK binding kinase 1/interferon regulatory factor 3 type I interferon pathway. YAP/transcriptional co-activator with a PDZ-binding domain (TAZ) silencing results in reduced ZIKV replication, indicating a direct role of Hippo pathway in regulating ZIKV infection. Using an in vivo Ifnar1-/- knockout mouse model, ZIKV infection was found to reduce YAP/TAZ protein levels while increasing phosphorylated YAP Ser127 in the retina and brain. Hippo pathway is activated in major cellular components of the blood-brain barrier, including endothelial cells and astrocytes. In addition, this result suggests AMP-activated protein kinase signaling pathway's role in regulating YAP/TAZ in ZIKV-infected cells. These data demonstrate that ZIKV infection might initiate a cross talk among AMP-activated protein kinase-Hippo-TBK1 pathways, which could regulate antiviral and energy stress responses during oculoneuronal inflammation.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | - Sayan Paul
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California; Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, India
| | - Sara Beshara
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | | | - Arunachalam Ramaiah
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California
| | - Karin Nielsen-Saines
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Melody M H Li
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California
| | - Samuel W French
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Kouki Morizono
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California; UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, Michigan
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California.
| |
Collapse
|
67
|
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
|
68
|
Poland GA, Ovsyannikova IG, Kennedy RB. Zika Vaccine Development: Current Status. Mayo Clin Proc 2019; 94:2572-2586. [PMID: 31806107 PMCID: PMC7094556 DOI: 10.1016/j.mayocp.2019.05.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/22/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022]
Abstract
Zika virus outbreaks have been explosive and unpredictable and have led to significant adverse health effects-as well as considerable public anxiety. Significant scientific work has resulted in multiple candidate vaccines that are now undergoing further clinical development, with several vaccines now in phase 2 clinical trials. In this review, we survey current vaccine efforts, preclinical and clinical results, and ethical and other concerns that directly bear on vaccine development. It is clear that the world needs safe and effective vaccines to protect against Zika virus infection. Whether such vaccines can be developed through to licensure and public availability absent significant financial investment by countries, and other barriers discussed within this article, remains uncertain.
Collapse
Key Words
- ade, antibody-dependent enhancement
- c, capsid
- denv, dengue virus
- e, envelope
- gbs, guillain-barré syndrome
- ifn, interferon
- irf, ifn response factor
- mrna, messenger rna
- prm, premembrane/membrane
- who, world health organization
- zikv, zika virus
Collapse
Affiliation(s)
- Gregory A Poland
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN.
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
69
|
Apari P, Bajer K, Brooks DR, Molnar O. Hiding in plain sight: an evolutionary approach to the South American Zika outbreak and its future consequences. ZOOLOGIA 2019. [DOI: 10.3897/zoologia.36.e36272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Emerging Infectious Diseases (EID) pose a world-wide health and socio-economic threat. Accelerating climate change and globalization are exposing unforeseen ways that pathogens cope with their surroundings. The 2015 Zika virus (ZIKV) outbreak was an example of expansion into previously inaccessible fitness spaces, causing a sudden epidemic. Recent studies indicating the subsequent decrease in symptomatic cases means the virus is in remission, currently poses little threat, and therefore can be ignored. We present an evolutionary scenario derived from the Stockholm Paradigm, of oscillating phases of expansion and isolation, accompanied by changes in transmission, persistence, virulence, and pathology. Chief among these is the likelihood that asymptomatic strains are constantly transmitted sexually. This suggests that the currently quiescent virus retains capacities to reemerge abruptly and spread rapidly in an arena of changing opportunity.
Collapse
|
70
|
Su KY, Balasubramaniam VRMT. Zika Virus as Oncolytic Therapy for Brain Cancer: Myth or Reality? Front Microbiol 2019; 10:2715. [PMID: 31824472 PMCID: PMC6879458 DOI: 10.3389/fmicb.2019.02715] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
The ability of self-replicating oncolytic viruses (OVs) to preferentially infect and lyse cancer cells while stimulating anti-tumor immunity of the host strongly indicates its value as a new field of cancer therapeutics to be further explored. The emergence of Zika virus (ZIKV) as a global health threat due to its recent outbreak in Brazil has caught the attention of the scientific community and led to the discovery of its oncolytic potential for the treatment of glioblastoma multiforme (GBM), the most common and fatal brain tumor with poor prognosis. Herein, we evaluate the neurotropism of ZIKV relative to the receptor tyrosine kinase AXL and its ligand Gas6 in viral entry and the RNA-binding protein Musashi-1 (MSI1) in replication which are also overexpressed in GBM, suggesting its potential for specific targeting of the tumor. Additionally, this review discusses genetic modifications performed to enhance safety and efficacy of ZIKV as well as speculates future directions for the OV therapy.
Collapse
Affiliation(s)
- Kar Yan Su
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia.,School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | | |
Collapse
|
71
|
Ayala-Nunez NV, Follain G, Delalande F, Hirschler A, Partiot E, Hale GL, Bollweg BC, Roels J, Chazal M, Bakoa F, Carocci M, Bourdoulous S, Faklaris O, Zaki SR, Eckly A, Uring-Lambert B, Doussau F, Cianferani S, Carapito C, Jacobs FMJ, Jouvenet N, Goetz JG, Gaudin R. Zika virus enhances monocyte adhesion and transmigration favoring viral dissemination to neural cells. Nat Commun 2019; 10:4430. [PMID: 31562326 PMCID: PMC6764950 DOI: 10.1038/s41467-019-12408-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Zika virus (ZIKV) invades and persists in the central nervous system (CNS), causing severe neurological diseases. However the virus journey, from the bloodstream to tissues through a mature endothelium, remains unclear. Here, we show that ZIKV-infected monocytes represent suitable carriers for viral dissemination to the CNS using human primary monocytes, cerebral organoids derived from embryonic stem cells, organotypic mouse cerebellar slices, a xenotypic human-zebrafish model, and human fetus brain samples. We find that ZIKV-exposed monocytes exhibit higher expression of adhesion molecules, and higher abilities to attach onto the vessel wall and transmigrate across endothelia. This phenotype is associated to enhanced monocyte-mediated ZIKV dissemination to neural cells. Together, our data show that ZIKV manipulates the monocyte adhesive properties and enhances monocyte transmigration and viral dissemination to neural cells. Monocyte transmigration may represent an important mechanism required for viral tissue invasion and persistence that could be specifically targeted for therapeutic intervention. Zika virus (ZIKV) can infect the central nervous system, but it is not clear how it reaches the brain. Here, Ayala-Nunez et al. show in ex vivo and in vivo models that ZIKV can hitch a ride in monocytes in a Trojan Horse manner to cross the endothelium and disseminate the virus.
Collapse
Affiliation(s)
- Nilda Vanesa Ayala-Nunez
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, 34293, Montpellier, France.,Université de Strasbourg, INSERM, 67000, Strasbourg, France
| | | | - François Delalande
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Aurélie Hirschler
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Emma Partiot
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, 34293, Montpellier, France
| | - Gillian L Hale
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centers for Disease Control and Prevention, 1600 Clifton Rd NE, MS: G32, Atlanta, GA, 30329-4027, USA
| | - Brigid C Bollweg
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centers for Disease Control and Prevention, 1600 Clifton Rd NE, MS: G32, Atlanta, GA, 30329-4027, USA
| | - Judith Roels
- University of Amsterdam, Swammerdam Institute for Life Sciences, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Maxime Chazal
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology Department, Institut Pasteur, 75015, Paris, France
| | - Florian Bakoa
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology Department, Institut Pasteur, 75015, Paris, France
| | - Margot Carocci
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S1255, FMTS, 67000, Strasbourg, France
| | - Sandrine Bourdoulous
- INSERM U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Paris, France
| | - Orestis Faklaris
- MRI Core facility, Biocampus, CNRS UMS 3426, 34293, Montpellier, France
| | - Sherif R Zaki
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Centers for Disease Control and Prevention, 1600 Clifton Rd NE, MS: G32, Atlanta, GA, 30329-4027, USA
| | - Anita Eckly
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S1255, FMTS, 67000, Strasbourg, France
| | - Béatrice Uring-Lambert
- Hôpitaux universitaires de Strasbourg, laboratoire central d'immunologie, 67000, Strasbourg, France
| | - Frédéric Doussau
- Institut des Neurosciences Cellulaires et Intégratives, CNRS, Université de Strasbourg, 67000, Strasbourg, France
| | - Sarah Cianferani
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC, UMR 7178, CNRS-Université de Strasbourg, ECPM, 67087, Strasbourg, France
| | - Frank M J Jacobs
- University of Amsterdam, Swammerdam Institute for Life Sciences, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Nolwenn Jouvenet
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology Department, Institut Pasteur, 75015, Paris, France
| | | | - Raphael Gaudin
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, Université de Montpellier, 34293, Montpellier, France. .,Université de Strasbourg, INSERM, 67000, Strasbourg, France.
| |
Collapse
|
72
|
Leda AR, Bertrand L, Andras IE, El-Hage N, Nair M, Toborek M. Selective Disruption of the Blood-Brain Barrier by Zika Virus. Front Microbiol 2019; 10:2158. [PMID: 31620112 PMCID: PMC6759472 DOI: 10.3389/fmicb.2019.02158] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022] Open
Abstract
The blood–brain barrier (BBB) selectively regulates the cellular exchange of macromolecules between the circulation and the central nervous system (CNS). Here, we hypothesize that Zika virus (ZIKV) infects the brain via a disrupted BBB and altered expression of tight junction (TJ) proteins, which are structural components of the BBB. To assess this hypothesis, in vitro and in vivo studies were performed using three different strains of ZIKV: Honduras (ZIKV-H), Puerto Rico (ZIKV-PR), and Uganda (ZIKV-U). Primary human brain microvascular endothelial cells (BMECs) were productively infected by all studied ZIKV strains at MOI 0.01, and were analyzed by plaque assay, immunofluorescence for NS1 protein, and qRT-PCR at 2 and 6 days post-infection (dpi). Compared to mock-infected controls, expression level of ZO-1 was significantly upregulated in ZIKV-H-infected BMECs, while occludin and claudin-5 levels were significantly downregulated in BMECs infected by all three studied viral strains. Interestingly, BMEC permeability was not disturbed by ZIKV infection, even in the presence of a very high viral load (MOI 10). All studied ZIKV strains productively infected wild-type C57BL/J mice after intravenous infection with 107 PFU. Viral load was detected in the plasma, spleen, and brain from 1 to 8 dpi. Peak brain infection was observed at 2 dpi; therefore, TJ protein expression was assessed at this time point. Claudin-5 was significantly downregulated in ZIKV-U-infected animals and the BBB integrity was significantly disturbed in ZIKV-H-infected animals. Our results suggest that ZIKV penetrates the brain parenchyma early after infection with concurrent alterations of TJ protein expression and disruption of the BBB permeability in a strain-dependent manner.
Collapse
Affiliation(s)
- Ana Rachel Leda
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Luc Bertrand
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ibolya Edit Andras
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Nazira El-Hage
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
73
|
Tamhankar M, Patterson JL. Directional entry and release of Zika virus from polarized epithelial cells. Virol J 2019; 16:99. [PMID: 31395061 PMCID: PMC6688342 DOI: 10.1186/s12985-019-1200-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/18/2019] [Indexed: 03/09/2023] Open
Abstract
Background Both vector borne and sexual transmission of Zika virus (ZIKV) involve infection of epithelial cells in the initial stages of infection. Epithelial cells are unique in their ability to form polarized monolayers and their barrier function. Cell polarity induces an asymmetry in the epithelial monolayer, which is maintained by tight junctions and specialized sorting machinery. This differential localization can have a potential impact of virus infection. Asymmetrical distribution of a viral receptor can restrict virus entry to a particular membrane while polarized sorting can lead to a directional release of virions. The present study examined the impact of cell polarity on ZIKV infection and release. Methods A polarized Caco-2 cell model we described previously was used to assess ZIKV infection. Transepithelial resistance (TEER) was used to assess epithelial cell polarity, and virus infection was measured by immunofluorescence microscopy and qRT-PCR. Cell permeability was measured using a fluorescein leakage assay. Statistical significance was calculated using one-way ANOVA and significance was set at p < 0.05. Results Using the Caco-2 cell model for polarized epithelial cells, we report that Zika virus preferentially infects polarized cells from the apical route and is released vectorially through the basolateral route. Our data also indicates that release occurs without disruption of cell permeability. Conclusions Our results show that ZIKV has directional infection and egress in a polarized cell system. This mechanism of directional infection may be one of the mechanisms that enables the cross the epithelial barrier effectively without a disruption in cell monolayer integrity. Elucidation of entry and release characteristics of Zika virus in polarized epithelial cells can lead to better understanding of virus dissemination in the host, and can help in developing effective therapeutic interventions. Electronic supplementary material The online version of this article (10.1186/s12985-019-1200-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manasi Tamhankar
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA.,Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jean L Patterson
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|
74
|
Wang C, Puerta-Guardo H, Biering SB, Glasner DR, Tran EB, Patana M, Gomberg TA, Malvar C, Lo NTN, Espinosa DA, Harris E. Endocytosis of flavivirus NS1 is required for NS1-mediated endothelial hyperpermeability and is abolished by a single N-glycosylation site mutation. PLoS Pathog 2019; 15:e1007938. [PMID: 31356638 PMCID: PMC6687192 DOI: 10.1371/journal.ppat.1007938] [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: 10/01/2018] [Revised: 08/08/2019] [Accepted: 06/22/2019] [Indexed: 12/22/2022] Open
Abstract
Arthropod-borne flaviviruses cause life-threatening diseases associated with endothelial hyperpermeability and vascular leak. We recently found that vascular leak can be triggered by dengue virus (DENV) non-structural protein 1 (NS1) via the disruption of the endothelial glycocalyx-like layer (EGL). However, the molecular determinants of NS1 required to trigger EGL disruption and the cellular pathway(s) involved remain unknown. Here we report that mutation of a single glycosylated residue of NS1 (N207Q) abolishes the ability of NS1 to trigger EGL disruption and induce endothelial hyperpermeability. Intriguingly, while this mutant bound to the surface of endothelial cells comparably to wild-type NS1, it was no longer internalized, suggesting that NS1 binding and internalization are distinct steps. Using endocytic pathway inhibitors and gene-specific siRNAs, we determined that NS1 was endocytosed into endothelial cells in a dynamin- and clathrin-dependent manner, which was required to trigger endothelial dysfunction in vitro and vascular leak in vivo. Finally, we found that the N207 glycosylation site is highly conserved among flaviviruses and is also essential for West Nile and Zika virus NS1 to trigger endothelial hyperpermeability via clathrin-mediated endocytosis. These data provide critical mechanistic insight into flavivirus NS1-induced pathogenesis, presenting novel therapeutic and vaccine targets for flaviviral diseases. Vascular leak is a hallmark of severe dengue disease. Recently, our group revealed a critical role for NS1 in induction of endothelial hyperpermeability and vascular leakage in an endothelial cell-intrinsic manner. However, the upstream pathway triggered by NS1, as well as the molecular determinants of NS1 required for this phenomenon, remain obscure. Gaining insight into this endothelial cell-intrinsic pathway is critical for understanding dengue pathogenesis, developing novel antiviral therapies, and developing NS1-based vaccine approaches that pose a minimal risk of antibody-dependent enhancement. Our current study expands our knowledge of this novel pathway not only by identifying the requirement of internalization of secreted NS1 via clathrin-mediated endocytosis, but also by pinpointing the NS1 molecular determinant (N207) required to trigger vascular leak. Further, our work identifies N207 as a residue conserved among multiple flaviviruses (Zika virus and West Nile virus, in addition to DENV), which is critical for NS1-mediated vascular leak in biologically relevant human endothelial cells modeling interstitial compartments in the lung or the blood-brain barrier. Thus, our study identifies endocytosis and a single amino acid (N207) of the NS1 viral toxin as critical for pan-flavivirus pathogenesis, representing a novel target for anti-flaviviral therapy and vaccine development.
Collapse
Affiliation(s)
- Chunling Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Dustin R. Glasner
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Edwina B. Tran
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Mark Patana
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Trent A. Gomberg
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Carmel Malvar
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Nicholas T. N. Lo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Diego A. Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
75
|
Arévalo Romero H, Vargas Pavía TA, Velázquez Cervantes MA, Flores Pliego A, Helguera Repetto AC, León Juárez M. The Dual Role of the Immune Response in Reproductive Organs During Zika Virus Infection. Front Immunol 2019; 10:1617. [PMID: 31354746 PMCID: PMC6637308 DOI: 10.3389/fimmu.2019.01617] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/28/2019] [Indexed: 12/16/2022] Open
Abstract
Zika virus is a mosquito-borne viral disease that emerged as a significant health problem in the Americas after an epidemic in 2015. Especially concerning are cases where Zika is linked to the development of brain abnormalities in newborns. Unlike other flaviviruses, Zika can be transmitted sexually, increasing the potential for intraspecies infection. Several reports show that the virus can persist for months in the testis of males after clearance of viremia, and that females are highly susceptible to infection via sexual transmission. The most common route of sexual transmission is male-to-female, which suggests that the mechanism driving persistence of Zika in the testis is essential for dissemination. The immune system plays an essential role in Zika infection. In females, a robust response inhibits the virus to control the infection. In males, however, the immunological response to Zika infection correlates with viral persistence. Thus, the immune system may have a dual role in sexually transmitted pathogenesis. The mechanism by which the immune system allows the virus to enter an immune-privileged site while continuing to disseminate is unclear. In this mini-review, we highlight advances in our knowledge of sexually transmitted Zika virus pathogenesis and the possible mechanisms mounted by the immune system that control or exacerbate the infection.
Collapse
Affiliation(s)
- Haruki Arévalo Romero
- Laboratory of Immunology and Molecular Microbiology, Multidisciplinary Academic Division of Jalpa de Méndez, Department of Genomics, University Juárez Autonomous of Tabasco, Jalpa de Méndez, Mexico
| | - Tania A Vargas Pavía
- Laboratory of Perinatal Virology, Department of Immuno-Biochemistry, National Institution of Perinatology "Isidro Espinosa de los Reyes", Mexico City, Mexico
| | - Manuel A Velázquez Cervantes
- Laboratory of Perinatal Virology, Department of Immuno-Biochemistry, National Institution of Perinatology "Isidro Espinosa de los Reyes", Mexico City, Mexico
| | - Arturo Flores Pliego
- Laboratory of Perinatal Virology, Department of Immuno-Biochemistry, National Institution of Perinatology "Isidro Espinosa de los Reyes", Mexico City, Mexico
| | - Addy C Helguera Repetto
- Laboratory of Perinatal Virology, Department of Immuno-Biochemistry, National Institution of Perinatology "Isidro Espinosa de los Reyes", Mexico City, Mexico
| | - Moises León Juárez
- Laboratory of Perinatal Virology, Department of Immuno-Biochemistry, National Institution of Perinatology "Isidro Espinosa de los Reyes", Mexico City, Mexico
| |
Collapse
|
76
|
Khaiboullina SF, Ribeiro FM, Uppal T, Martynova EV, Rizvanov AA, Verma SC. Zika Virus Transmission Through Blood Tissue Barriers. Front Microbiol 2019; 10:1465. [PMID: 31333605 PMCID: PMC6621930 DOI: 10.3389/fmicb.2019.01465] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 06/11/2019] [Indexed: 01/12/2023] Open
Abstract
The recent Zika virus (ZIKV) epidemic in the Americas and the Caribbean revealed a new deadly strain of the mosquito-borne virus, which has never been associated with previous outbreaks in Asia. For the first time, widespread ZIKV infection was shown to cause microcephaly and death of newborns, which was most likely due to the mutation acquired during the large outbreak recorded in French Polynesia in 2013–2014. Productive ZIKV replication and persistence has been demonstrated in placenta and fetal brains. Possible association between ZIKV and microcephaly and fetal death has been confirmed using immunocompetent mouse models in vitro and in vivo. Having crossed the placenta, ZIKV directly targets neural progenitor cells (NPCs) in developing human fetus and triggers apoptosis. The embryonic endothelial cells are exceptionally susceptible to ZIKV infection, which causes cell death and tissue necrosis. On the contrary, ZIKV infection does not affect the adult brain microvascular cell morphology and blood–brain barrier function. ZIKV is transmitted primarily by Aedes mosquito bite and is introduced into the placenta/blood through replication at the site of the entry. Also, virus can be transmitted through unprotected sex. Although, multiple possible routes of virus infection have been identified, the exact mechanism(s) utilized by ZIKV to cross the placenta still remain largely unknown. In this review, the current understanding of ZIKV infection and transmission through the placental and brain barriers is summarized.
Collapse
Affiliation(s)
- Svetlana F Khaiboullina
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States.,Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Fabiola M Ribeiro
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Timsy Uppal
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States
| | - Ekaterina V Martynova
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Subhash C Verma
- Department of Microbiology and Immunology, Reno School of Medicine, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
77
|
Anfasa F, Goeijenbier M, Widagdo W, Siegers JY, Mumtaz N, Okba N, van Riel D, Rockx B, Koopmans MPG, Meijers JCM, Martina BEE. Zika Virus Infection Induces Elevation of Tissue Factor Production and Apoptosis on Human Umbilical Vein Endothelial Cells. Front Microbiol 2019; 10:817. [PMID: 31068911 PMCID: PMC6491739 DOI: 10.3389/fmicb.2019.00817] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 04/01/2019] [Indexed: 12/22/2022] Open
Abstract
Zika virus (ZIKV) infection is typically characterized by a mild disease presenting with fever, maculopapular rash, headache, fatigue, myalgia, and arthralgia. A recent animal study found that ZIKV-infected pregnant Ifnar -/-mice developed vascular damage in the placenta and reduced amount of fetal capillaries. Moreover, ZIKV infection causes segmental thrombosis in the umbilical cord of pregnant rhesus macaques. Furthermore, several case reports suggest that ZIKV infection cause coagulation disorders. These results suggest that ZIKV could cause an alteration in the host hemostatic response, however, the mechanism has not been investigated thus far. This paper aims to determine whether ZIKV infection on HUVECs induces apoptosis and elevation of tissue factor (TF) that leads to activation of secondary hemostasis. We infected HUVECs with two ZIKV strains and performed virus titration, immunostaining, and flow cytometry to confirm and quantify infection. We measured TF concentrations with flow cytometry and performed thrombin generation test (TGT) as a functional assay to assess secondary hemostasis. Furthermore, we determined the amount of cell death using flow cytometry. We also performed enzyme-linked immunosorbent assay (ELISA) to determine interleukin (IL)-6 and IL-8 production and conducted blocking experiments to associate these cytokines with TF expression. Both ZIKV strains infected and replicated to high titers in HUVECs. We found that infection induced elevation of TF expressions. We also showed that increased TF expression led to shortened TGT time. Moreover, the data revealed that infection induced apoptosis. In addition, there was a significant increase of IL-6 and IL-8 production in infected cells. Here we provide in vitro evidence that infection of HUVECs with ZIKV induces apoptosis and elevation of TF expression that leads to activation of secondary hemostasis.
Collapse
Affiliation(s)
- Fatih Anfasa
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Marco Goeijenbier
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Widagdo Widagdo
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jurre Y Siegers
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Noreen Mumtaz
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nisreen Okba
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Debby van Riel
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Joost C M Meijers
- Department of Plasma Proteins, Sanquin Research, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Byron E E Martina
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Artemis One Health Research Institute, Delft, Netherlands
| |
Collapse
|
78
|
Mustafá YM, Meuren LM, Coelho SVA, de Arruda LB. Pathways Exploited by Flaviviruses to Counteract the Blood-Brain Barrier and Invade the Central Nervous System. Front Microbiol 2019; 10:525. [PMID: 30984122 PMCID: PMC6447710 DOI: 10.3389/fmicb.2019.00525] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
Human infection by different flaviviruses may cause severe neurologic syndromes, through pathogenic mechanisms that are still largely unknown. Japanese encephalitis virus (JEV), West Nile virus (WNV), Zika virus (ZIKV), yellow fever virus (YFV), dengue virus (DENV), and tick-borne encephalitis virus (TBEV) are believed to reach the central nervous system by a hematogenous route, upon crossing the blood-brain barrier. Although the disruption of BBB during flavivirus infection has been largely evidenced in experimental models, the relevance of BBB breakdown for virus entering the brain was not completely elucidated. In vitro models of BBB had demonstrated that these viruses replicated in brain microvascular endothelial cells (BMECs), which induced downregulation of tight junction proteins and increased the permeability of the barrier. Other reports demonstrated that infection of BMECs allowed the basolateral release of infectious particles, without a remarkable cytopathic effect, what might be sufficient for virus invasion. Virus replication and activation of other cells associated to the BBB, mostly astrocytes and microglia, were also reported to affect the endothelial barrier permeability. This event might occur simultaneously or after BMECs infection, being a secondary effect leading to BBB disruption. Importantly, activation of BMECs, astrocytes, and microglia by flaviviruses was associated to the expression and secretion of inflammatory mediators, which are believed to recruit leukocytes to the CNS. The leukocyte infiltrate could further mediate viral invasion through a Trojan horse mechanism and might contribute to BBB breakdown and to neurological alterations. This review discussed the previous studies regarding in vitro and in vivo models of JEV, WNV, ZIKV, YFV, DENV, and TBEV infection and addressed the pathways for BBB overcome and invasion of the CNS described for each virus infection, aiming to increment the knowledge and stimulate further discussion about the role of BBB in the neuropathogenesis of flavivirus infection.
Collapse
Affiliation(s)
- Yasmin Mucunã Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lana Monteiro Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Sharton Vinícius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
79
|
Ventura CV, Bandstra ES, Fernandez MP, Cooper JM, Saigal GM, Bauer CR, Hofheimer JA, Berkovits MD, Fifer RC, Pensirikul AD, Gonzalez IA, Curry CL, Andreansky S, Younis RT, Liu XZ, Banker TP, Dubovy SR, Langer SM, Berrocal AM. First Locally Acquired Congenital Zika Syndrome Case in the United States: Neonatal Clinical Manifestations. Ophthalmic Surg Lasers Imaging Retina 2019; 49:e93-e98. [PMID: 30222826 DOI: 10.3928/23258160-20180907-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/27/2018] [Indexed: 11/20/2022]
Abstract
In the spring of 2017, a full-term infant with microcephaly was delivered in South Florida. During first trimester, the mother presented with fever, nausea, and vomiting. She reported no foreign travel for herself or her partner. The infant's neurologic, ophthalmologic, neuroradiologic, and audiologic findings were highly suggestive of congenital Zika syndrome (CZS), confirmed by IgM antibodies and plaque reduction neutralization test. New observations, including peripheral temporal retinal avascularity and peripapillary retinal nerve fiber layer thinning, are presented from this first known case of non-travel-associated CZS in the United States. [Ophthalmic Surg Lasers Imaging Retina. 2018;49:e93-e98.].
Collapse
|
80
|
Pardy RD, Valbon SF, Richer MJ. Running interference: Interplay between Zika virus and the host interferon response. Cytokine 2019; 119:7-15. [PMID: 30856603 DOI: 10.1016/j.cyto.2019.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/07/2019] [Accepted: 02/10/2019] [Indexed: 12/11/2022]
Abstract
The interferon (IFN) family of cytokines is a crucial part of the host's ability to mount an effective immune response against viral infections. In addition to establishing an antiviral state within cells, IFNs also support the optimal activation of other key immune cell types. The ability of members of the Flaviviridae family to suppress type I IFN responses has been well-described. Of these viruses, Zika virus (ZIKV) has recently attracted international attention due to a series of major outbreaks that featured the novel association of neurological symptoms with infection. Researchers have begun to investigate the strategies ZIKV uses to evade type I IFNs, and the impact this has on the host. However, a unique feature of ZIKV infection compared to other flaviviruses is its capacity to be transmitted sexually, as well as its ability to infect and persist within reproductive tissues. As such, this raises the question of a potential role for type III IFN during ZIKV infection. In this review, we will discuss the interplay between these two classes of IFN with ZIKV, models that have been used to interrogate these interactions, and the effect this interplay has on infection and infection outcomes. We will also consider the intriguing possibility of whether ZIKV has evolved improved evasion mechanisms to suppress the IFN response in recent outbreaks.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Stefanie F Valbon
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Martin J Richer
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
81
|
Sutherland MR, Simon AY, Shanina I, Horwitz MS, Ruf W, Pryzdial ELG. Virus envelope tissue factor promotes infection in mice. J Thromb Haemost 2019; 17:482-491. [PMID: 30659719 PMCID: PMC6397068 DOI: 10.1111/jth.14389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Indexed: 01/04/2023]
Abstract
Essentials The coagulation initiator, tissue factor (TF), is on the herpes simplex virus 1 (HSV1) surface. HSV1 surface TF was examined in mice as an antiviral target since it enhances infection in vitro. HSV1 surface TF facilitated infection of all organs evaluated and anticoagulants were antiviral. Protease activated receptor 2 inhibited infection in vivo and its pre-activation was antiviral. SUMMARY: Background Tissue factor (TF) is the essential cell surface initiator of coagulation, and mediates cell signaling through protease-activated receptor (PAR) 2. Having a diverse cellular distribution, TF is involved in many biological pathways and pathologies. Our earlier work identified host cell-derived TF on the envelope covering several viruses, and showed its involvement in enhanced cell infection in vitro. Objective In the current study, we evaluated the in vivo effects of virus surface TF on infection and on the related modulator of infection PAR2. Methods With the use of herpes simplex virus type 1 (HSV1) as a model enveloped virus, purified HSV1 was generated with or without envelope TF through propagation in a TF-inducible cell line. Infection was studied after intravenous inoculation of BALB/c, C57BL/6J or C57BL/6J PAR2 knockout mice with 5 × 105 plaque-forming units of HSV1, mimicking viremia. Three days after inoculation, organs were processed, and virus was quantified with plaque-forming assays and quantitative real-time PCR. Results Infection of brain, lung, heart, spinal cord and liver by HSV1 required viral TF. Demonstrating promise as a therapeutic target, virus-specific anti-TF mAbs or small-molecule inhibitors of coagulation inhibited infection. PAR2 modulates HSV1 in vivo as demonstrated with PAR2 knockout mice and PAR2 agonist peptide. Conclusion TF is a constituent of many permissive host cell types. Therefore, the results presented here may explain why many viruses are correlated with hemostatic abnormalities, and indicate that TF is a novel pan-specific envelope antiviral target.
Collapse
MESH Headings
- Animals
- Anticoagulants/pharmacology
- Antiviral Agents/pharmacology
- Disease Models, Animal
- Female
- Herpes Simplex/blood
- Herpes Simplex/drug therapy
- Herpes Simplex/immunology
- Herpes Simplex/virology
- Herpesvirus 1, Human/drug effects
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/metabolism
- Host-Pathogen Interactions
- Injections, Intravenous
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, PAR-2/genetics
- Receptor, PAR-2/metabolism
- Th1 Cells/immunology
- Th1 Cells/virology
- Thromboplastin/administration & dosage
- Thromboplastin/metabolism
- Viral Envelope Proteins/administration & dosage
- Viral Envelope Proteins/metabolism
Collapse
Affiliation(s)
- Michael R Sutherland
- Canadian Blood Services, Center for Innovation, Vancouver, Canada
- Centre for Blood Research and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Ayo Y Simon
- Canadian Blood Services, Center for Innovation, Vancouver, Canada
- Centre for Blood Research and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- African Centre of Excellence on Neglected Tropical Diseases and Forensic Biotechnology and Veterinary Teaching Hospital, Ahmadu Bello University, Zaria, Nigeria
- Preclinical Research and Development, Emergent BioSolutions, Winnipeg, Manitoba, Canada
| | - Iryna Shanina
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Marc S Horwitz
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Wolfram Ruf
- Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA, USA
- Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany
| | - Edward L G Pryzdial
- Canadian Blood Services, Center for Innovation, Vancouver, Canada
- Centre for Blood Research and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
82
|
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
|
83
|
Zika virus: lessons learned in Brazil. Microbes Infect 2018; 20:661-669. [DOI: 10.1016/j.micinf.2018.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/15/2018] [Accepted: 02/21/2018] [Indexed: 01/07/2023]
|
84
|
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
|
85
|
Beaufrère A, Bessières B, Bonnière M, Driessen M, Alfano C, Couderc T, Thiry M, Thelen N, Lecuit M, Attié-Bitach T, Vekemans M, Ville Y, Nguyen L, Leruez-Ville M, Encha-Razavi F. A clinical and histopathological study of malformations observed in fetuses infected by the Zika virus. Brain Pathol 2018; 29:114-125. [PMID: 30020561 DOI: 10.1111/bpa.12644] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/20/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The recent outbreak of Zika virus (ZIKV) infection and the associated increased prevalence of microcephaly in Brazil underline the impact of viral infections on embryo fetal development. The aim of the present study is to provide a detailed clinical and histopathological study of the fetal disruption caused by the ZIKV, with a special focus on the associated neuropathological findings. METHODS A detailed feto-placental examination, as well as neuropathological and neurobiological studies were performed on three fetuses collected after pregnancy termination between 22 and 25 weeks of gestation (WG), because brain malformations associated with a maternal and fetal ZIKV infection was diagnosed. RESULTS In all three cases, the maternal infection occurred during the first trimester of pregnancy. A small head was observed on the ultrasound examination of the second trimester of pregnancy and led to the diagnosis of ZIKV fetopathy and pregnancy termination. The fetal histopathological examination was unremarkable on the viscera but showed on the testis an interstitial lymphocytic infiltrate. The placenta contained a Hofbauer cells hyperplasia with signs of inflammation. Neuropathological findings included a meningoencephalitis and an ex vacuo hydrocephalus. Immunohistochemical studies showed the presence of T lymphocytic and histiocytic meningitis associated with an abundant cerebral astroglial and macrophagic reaction. In situ hybridization demonstrated, abundant ZIKV particles within the cerebral parenchyma mainly in the ventricular/subventricular zone and in the cortical plate. In addition massive cells death and endoplasmic reticulum damage were present. CONCLUSION The present study reports on the clinical and histopathological findings observed in three fetuses infected by the ZIKV. It emphasizes the severity of brain damages and the minimal visceral and placental changes observed upon ZIKV infection. This confirms the selective neurotropism of ZIKV. Finally, it allows us to describe the cascade of multifactorial developmental defects leading to microcephaly.
Collapse
Affiliation(s)
- Aurélie Beaufrère
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France
| | - Bettina Bessières
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France
| | - Maryse Bonnière
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France
| | - Marine Driessen
- Service de Gynécologie-Obstétrique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France
| | | | - Thérèse Couderc
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Inserm U1117, Paris, France
| | - Marc Thiry
- GIGA-Neurosciences, Université de Liège, Liège, Belgique
| | - Nicolas Thelen
- GIGA-Neurosciences, Université de Liège, Liège, Belgique
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Inserm U1117, Paris, France.,Paris-Descartes University, Sorbonne Paris Cité, Centre d'Infectiologie Necker-Pasteur, Necker-Enfants Malades, Paris, France
| | - Tania Attié-Bitach
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France.,INSERM U-1163, Institut Imagine, Paris, France.,Université Paris Descartes, Paris, France
| | - Michel Vekemans
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France.,INSERM U-1163, Institut Imagine, Paris, France.,Université Paris Descartes, Paris, France
| | - Yves Ville
- Service de Gynécologie-Obstétrique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France.,Université Paris Descartes, Paris, France
| | - Laurent Nguyen
- GIGA-Neurosciences, Université de Liège, Liège, Belgique
| | - Marianne Leruez-Ville
- Université Paris Descartes, Paris, France.,Laboratoire de Virologie, Hôpital Universitaire Necker Enfants Malades, APHP, Paris, France
| | - Férechté Encha-Razavi
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Universitaire Necker-Enfants Malades, APHP, Paris, France
| |
Collapse
|
86
|
de Oliveira Dias JR, Ventura CV, de Paula Freitas B, Prazeres J, Ventura LO, Bravo-Filho V, Aleman T, Ko AI, Zin A, Belfort R, Maia M. Zika and the Eye: Pieces of a Puzzle. Prog Retin Eye Res 2018; 66:85-106. [PMID: 29698814 DOI: 10.1016/j.preteyeres.2018.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/05/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022]
Abstract
Zika virus (ZIKV) is an arbovirus mainly transmitted to humans by mosquitoes from Aedes genus. Other ways of transmission include the perinatal and sexual routes, blood transfusion, and laboratory exposure. Although the first human cases were registered in 1952 in African countries, outbreaks were only reported since 2007, when entire Pacific islands were affected. In March 2015, the first cases of ZIKV acute infection were notified in Brazil and, to date, 48 countries and territories in the Americas have confirmed local mosquito-borne transmission of ZIKV. Until 2015, ZIKV infection was thought to only cause asymptomatic or mild exanthematous febrile infections. However, after explosive ZIKV outbreaks in Polynesia and Latin American countries, it was confirmed that ZIKV could also lead to Guillain-Barré syndrome and congenital birth abnormalities. These abnormalities, which can include neurologic, ophthalmologic, audiologic, and skeletal findings, are now considered congenital Zika syndrome (CZS). Brain abnormalities in CZS include cerebral calcifications, malformations of cortical development, ventriculomegaly, lissencephaly, hypoplasia of the cerebellum and brainstem. The ocular findings, which are present in up to 70% of infants with CZS, include iris coloboma, lens subluxation, cataract, congenital glaucoma, and especially posterior segment findings. Loss of retinal pigment epithelium, the presence of a thin choroid, a perivascular choroidal inflammatory infiltrate, and atrophic changes within the optic nerve were seen in histologic analyses of eyes from deceased fetuses. To date, there is no ZIKV licensed vaccines or antiviral therapies are available for treatment. Preventive measures include individual protection from mosquito bites, control of mosquito populations and the use of barriers measures such as condoms during sexual intercourse or sexual abstinence for couples either at risk or after confirmed infection. A literature review based on studies that analyzed ocular findings in mothers and infants with CZS, with or without microcephaly, was conducted and a theoretical pathophysiologic explanation for ZIKV-ocular abnormalities was formulated.
Collapse
Affiliation(s)
- João Rafael de Oliveira Dias
- Vision Institute, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil
| | - Camila V Ventura
- Vision Institute, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil; Altino Ventura Foundation, Recife, Pernambuco, Brazil; HOPE Eye Hospital, Recife, Pernambuco, Brazil
| | - Bruno de Paula Freitas
- Vision Institute, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil; Department of Ophthalmology, Roberto Santos General Hospital, Salvador, Brazil
| | - Juliana Prazeres
- Vision Institute, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil
| | - Liana O Ventura
- Vision Institute, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil; Altino Ventura Foundation, Recife, Pernambuco, Brazil; HOPE Eye Hospital, Recife, Pernambuco, Brazil
| | - Vasco Bravo-Filho
- Altino Ventura Foundation, Recife, Pernambuco, Brazil; HOPE Eye Hospital, Recife, Pernambuco, Brazil
| | - Tomas Aleman
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, Perelman School of Medicine, Department of Ophthalmology, University of Pennsylvania, Philadelphia, United States
| | - Albert Icksang Ko
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation, Salvador, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Andréa Zin
- Clinical Research Unit, Fernandes Figueira Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Rubens Belfort
- Vision Institute, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil
| | - Mauricio Maia
- Vision Institute, Department of Ophthalmology, Paulista Medical School, Federal University of São Paulo, São Paulo, Brazil; Brazilian Institute of Fight Against Blindness, Assis and Presidente Prudente, São Paulo, Brazil.
| |
Collapse
|
87
|
Abstract
Recent Zika virus outbreaks have been associated with severe outcomes, especially during pregnancy. A great deal of effort has been put toward understanding this virus, particularly the immune mechanisms responsible for rapid viral control in the majority of infections. Identifying and understanding the key mechanisms of immune control will provide the foundation for the development of effective vaccines and antiviral therapy. Here, we outline a mathematical modeling approach for analyzing the within-host dynamics of Zika virus, and we describe how these models can be used to understand key aspects of the viral life cycle and to predict antiviral efficacy.
Collapse
Affiliation(s)
- Katharine Best
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| |
Collapse
|
88
|
Garcez PP, Stolp HB, Sravanam S, Christoff RR, Ferreira JCCG, Dias AA, Pezzuto P, Higa LM, Barbeito-Andrés J, Ferreira RO, Andrade CBV, Siqueira M, Santos TMP, Drumond J, Hoerder-Suabedissen A, de Lima CVF, Tovar-Moll F, Lopes RT, Fragel-Madeira L, Lent R, Ortiga-Carvalho TM, Stipursky J, Bellio M, Tanuri A, Molnár Z. Zika virus impairs the development of blood vessels in a mouse model of congenital infection. Sci Rep 2018; 8:12774. [PMID: 30143723 PMCID: PMC6109170 DOI: 10.1038/s41598-018-31149-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/13/2018] [Indexed: 01/28/2023] Open
Abstract
Zika virus (ZIKV) is associated with brain development abnormalities such as primary microcephaly, a severe reduction in brain growth. Here we demonstrated in vivo the impact of congenital ZIKV infection in blood vessel development, a crucial step in organogenesis. ZIKV was injected intravenously in the pregnant type 2 interferon (IFN)-deficient mouse at embryonic day (E) 12.5. The embryos were collected at E15.5 and postnatal day (P)2. Immunohistochemistry for cortical progenitors and neuronal markers at E15.5 showed the reduction of both populations as a result of ZIKV infection. Using confocal 3D imaging, we found that ZIKV infected brain sections displayed a reduction in the vasculature density and vessel branching compared to mocks at E15.5; altogether, cortical vessels presented a comparatively immature pattern in the infected tissue. These impaired vascular patterns were also apparent in the placenta and retina. Moreover, proteomic analysis has shown that angiogenesis proteins are deregulated in the infected brains compared to controls. At P2, the cortical size and brain weight were reduced in comparison to mock-infected animals. In sum, our results indicate that ZIKV impairs angiogenesis in addition to neurogenesis during development. The vasculature defects represent a limitation for general brain growth but also could regulate neurogenesis directly.
Collapse
Affiliation(s)
- P P Garcez
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - H B Stolp
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
- Centre for the Developing Brain, Department of Perinatal Imaging & Health, King's College London, London, UK.
| | - S Sravanam
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - R R Christoff
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J C C G Ferreira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A A Dias
- Microbiology Institute Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - P Pezzuto
- Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - L M Higa
- Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J Barbeito-Andrés
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - R O Ferreira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - C B V Andrade
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - M Siqueira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - T M P Santos
- Nuclear Instrumentation Laboratory, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J Drumond
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - C V F de Lima
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - F Tovar-Moll
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - R T Lopes
- Nuclear Instrumentation Laboratory, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - L Fragel-Madeira
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - R Lent
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - T M Ortiga-Carvalho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J Stipursky
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - M Bellio
- Microbiology Institute Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A Tanuri
- Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Z Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
89
|
Matusali G, Houzet L, Satie AP, Mahé D, Aubry F, Couderc T, Frouard J, Bourgeau S, Bensalah K, Lavoué S, Joguet G, Bujan L, Cabié A, Avelar G, Lecuit M, Le Tortorec A, Dejucq-Rainsford N. Zika virus infects human testicular tissue and germ cells. J Clin Invest 2018; 128:4697-4710. [PMID: 30063220 DOI: 10.1172/jci121735] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV) is a teratogenic mosquito-borne flavivirus that can be sexually transmitted from man to woman. The finding of high viral loads and prolonged viral shedding in semen suggests that ZIKV replicates within the human male genital tract, but its target organs are unknown. Using ex vivo infection of organotypic cultures, we demonstrated here that ZIKV replicates in human testicular tissue and infects a broad range of cell types, including germ cells, which we also identified as infected in semen from ZIKV-infected donors. ZIKV had no major deleterious effect on the morphology and hormonal production of the human testis explants. Infection induced a broad antiviral response but no IFN upregulation and minimal proinflammatory response in testis explants, with no cytopathic effect. Finally, we studied ZIKV infection in mouse testis and compared it to human infection. This study provides key insights into how ZIKV may persist in semen and alter semen parameters, as well as a valuable tool for testing antiviral agents.
Collapse
Affiliation(s)
- Giulia Matusali
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Laurent Houzet
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Anne-Pascale Satie
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Dominique Mahé
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Florence Aubry
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Thérèse Couderc
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Inserm U1117, Paris, France
| | - Julie Frouard
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Salomé Bourgeau
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Karim Bensalah
- Service d'Urologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Sylvain Lavoué
- Unité de coordination hospitalière des prélèvements d'organes et de tissus, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Guillaume Joguet
- Centre Caribéen de Médecine de la Reproduction-CECOS CHU de Pointe-à-Pitre, Pointe-à-Pitre, France
| | - Louis Bujan
- Research Group on Human Fertility EA 3694, University Paul Sabatier Toulouse III - CECOS, Hôpital Paule de Viguier, CHU Toulouse, Toulouse, France
| | - André Cabié
- Inserm Centre d'Investigation Clinique 1424, Centre Hospitalier Universitaire de Martinique, and Service de maladies infectieuses, Centre Hospitalier Universitaire de Martinique, Fort de France, France
| | - Gleide Avelar
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Inserm U1117, Paris, France.,Paris-Descartes University, Department of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, Paris, France
| | - Anna Le Tortorec
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- Université de Rennes, Inserm, École des hautes études en santé publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) - UMR_S1085, Rennes, France
| |
Collapse
|
90
|
Beaver JT, Lelutiu N, Habib R, Skountzou I. Evolution of Two Major Zika Virus Lineages: Implications for Pathology, Immune Response, and Vaccine Development. Front Immunol 2018; 9:1640. [PMID: 30072993 PMCID: PMC6058022 DOI: 10.3389/fimmu.2018.01640] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/03/2018] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) became a public health emergency of global concern in 2015 due to its rapid expansion from French Polynesia to Brazil, spreading quickly throughout the Americas. Its unexpected correlation to neurological impairments and defects, now known as congenital Zika syndrome, brought on an urgency to characterize the pathology and develop safe, effective vaccines. ZIKV genetic analyses have identified two major lineages, Asian and African, which have undergone substantial changes during the past 50 years. Although ZIKV infections have been circulating throughout Africa and Asia for the later part of the 20th century, the symptoms were mild and not associated with serious pathology until now. ZIKV evolution also took the form of novel modes of transmission, including maternal-fetal transmission, sexual transmission, and transmission through the eye. The African and Asian lineages have demonstrated differential pathogenesis and molecular responses in vitro and in vivo. The limited number of human infections prior to the 21st century restricted ZIKV research to in vitro studies, but current animal studies utilize mice deficient in type I interferon (IFN) signaling in order to invoke enhanced viral pathogenesis. This review examines ZIKV strain differences from an evolutionary perspective, discussing how these differentially impact pathogenesis via host immune responses that modulate IFN signaling, and how these differential effects dictate the future of ZIKV vaccine candidates.
Collapse
Affiliation(s)
| | | | | | - Ioanna Skountzou
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
91
|
Qadir A, Riaz M, Saeed M, Shahzad-Ul-Hussan S. Potential targets for therapeutic intervention and structure based vaccine design against Zika virus. Eur J Med Chem 2018; 156:444-460. [PMID: 30015077 DOI: 10.1016/j.ejmech.2018.07.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023]
Abstract
Continuously increasing number of reports of Zika virus (ZIKV) infections and associated severe clinical manifestations, including autoimmune abnormalities and neurological disorders such as neonatal microcephaly and Guillain-Barré syndrome have created alarming situation in various countries. To date, no specific antiviral therapy or vaccine is available against ZIKV. This review provides a comprehensive insight into the potential therapeutic targets and describes viral epitopes of broadly neutralizing antibodies (bNAbs) in vaccine design perspective. Interactions between ZIKV envelope glycoprotein E and cellular receptors mediate the viral fusion and entry to the target cell. Blocking these interactions by targeting cellular receptors or viral structural proteins mediating these interactions or viral surface glycans can inhibit viral entry to the cell. Similarly, different non-structural proteins of ZIKV and un-translated regions (UTRs) of its RNA play essential roles in viral replication cycle and potentiate for therapeutic interventions. Structure based vaccine design requires identity and structural description of the epitopes of bNAbs. We have described different conserved bNAb epitopes present in the ZIKV envelope as potential targets for structure based vaccine design. This review also highlights successes, unanswered questions and future perspectives in relation to therapeutic and vaccine development against ZIKV.
Collapse
Affiliation(s)
- Amina Qadir
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Muhammad Riaz
- Department of Chemistry, University of Azad Jammu & Kashmir, Muzaffarabad, Pakistan
| | - Muhammad Saeed
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan.
| | - Syed Shahzad-Ul-Hussan
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, 54792, Pakistan.
| |
Collapse
|
92
|
Lang J, Cheng Y, Rolfe A, Hammack C, Vera D, Kyle K, Wang J, Meissner TB, Ren Y, Cowan C, Tang H. An hPSC-Derived Tissue-Resident Macrophage Model Reveals Differential Responses of Macrophages to ZIKV and DENV Infection. Stem Cell Reports 2018; 11:348-362. [PMID: 29983385 PMCID: PMC6092684 DOI: 10.1016/j.stemcr.2018.06.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/14/2022] Open
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are two closely related flaviviruses that lead to different clinical outcomes. The mechanism for the distinct pathogenesis of ZIKV and DENV is poorly understood. Here, we investigate ZIKV and DENV infection of macrophages using a human pluripotent stem cell (hPSC)-derived macrophage model and discover key virus-specific responses. ZIKV and DENV productively infect hPSC-derived macrophages. DENV, but not ZIKV, infection of macrophages strongly activates macrophage migration inhibitory factor (MIF) secretion and decreases macrophage migration. Neutralization of MIF leads to improved migratory ability of DENV-infected macrophages. In contrast, ZIKV-infected macrophages exhibit prolonged migration and express low levels of pro-inflammatory cytokines and chemokines. Mechanistically, ZIKV disrupts the nuclear factor κB (NF-κB)-MIF positive feedback loop by inhibiting the NF-κB signaling pathway. Our results demonstrate the utility of hPSC-derived macrophages in infectious disease modeling and suggest that the distinct impact of ZIKV and DENV on macrophage immune response may underlie different pathogenesis of Zika and dengue diseases. An hPSC-derived tissue-resident macrophage model for ZIKV and DENV infection ZIKV-, but not DENV-, infected macrophages maintain migratory capacity ZIKV, but not DENV, inhibits pro-inflammatory cytokines and chemokines expression ZIKV disrupts NF-κB-MIF positive feedback loop by inhibiting NF-κB pathway
Collapse
Affiliation(s)
- Jianshe Lang
- Department of Biological Science, Florida State University, 319 Stadium Dr., Tallahassee, FL 32306-4295, USA
| | - Yichen Cheng
- Department of Biological Science, Florida State University, 319 Stadium Dr., Tallahassee, FL 32306-4295, USA
| | - Alyssa Rolfe
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Christy Hammack
- Department of Biological Science, Florida State University, 319 Stadium Dr., Tallahassee, FL 32306-4295, USA
| | - Daniel Vera
- Center for Genomics and Personalized Medicine, Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Kathleen Kyle
- Center for Genomics and Personalized Medicine, Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Jingying Wang
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Torsten B Meissner
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Yi Ren
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Chad Cowan
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, 319 Stadium Dr., Tallahassee, FL 32306-4295, USA.
| |
Collapse
|
93
|
Marsakatla P, Suneetha S, Lee J, Swaminathan PD, Vasudevan L, Supriya R, Suneetha LM. Insights from the sequence similarity of Zika virus proteins with the Human nerve proteins. Bioinformation 2018; 14:194-200. [PMID: 30108415 PMCID: PMC6077820 DOI: 10.6026/97320630014194] [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: 04/10/2018] [Revised: 04/23/2018] [Accepted: 04/30/2018] [Indexed: 11/23/2022] Open
Abstract
Massive peptide sharing between the Zika virus polyprotein and host tissue proteins could elicit significant host-pathogen interactions and cross-reactions leading to autoimmune diseases. This study found similarities in the Zika V proteins and human nerve tissue proteins. 63 human nerve proteins were screened for similarities with the Zika V of which Neuromodulin, Nestin, Galanin, Bombesin, Calcium-binding protein were found to have similarities to the Zika V poly protein C at different sequence regions. These sequence similarities could be significant in regulating pathogenic interactions/autoimmunity, as Polyprotein C is known to be a virulent factor.
Collapse
Affiliation(s)
| | - Sujai Suneetha
- CODEWEL Nireekshana ACET, Narayanaguda, Hyderabad -500029, Telangana, India
| | - Joshua Lee
- York University, Department of Science, 4700 Keele St, Toronto, ON M3J 1P3, Canada
| | | | | | - Rachael Supriya
- CODEWEL Nireekshana ACET, Narayanaguda, Hyderabad -500029, Telangana, India
| | | |
Collapse
|
94
|
Alimonti JB, Ribecco-Lutkiewicz M, Sodja C, Jezierski A, Stanimirovic DB, Liu Q, Haqqani AS, Conlan W, Bani-Yaghoub M. Zika virus crosses an in vitro human blood brain barrier model. Fluids Barriers CNS 2018; 15:15. [PMID: 29759080 PMCID: PMC5952854 DOI: 10.1186/s12987-018-0100-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) is a flavivirus that is highly neurotropic causing congenital abnormalities and neurological damage to the central nervous systems (CNS). In this study, we used a human induced pluripotent stem cell (iPSC)-derived blood brain barrier (BBB) model to demonstrate that ZIKV can infect brain endothelial cells (i-BECs) without compromising the BBB barrier integrity or permeability. Although no disruption to the BBB was observed post-infection, ZIKV particles were released on the abluminal side of the BBB model and infected underlying iPSC-derived neural progenitor cells (i-NPs). AXL, a putative ZIKV cellular entry receptor, was also highly expressed in ZIKV-susceptible i-BEC and i-NPs. This iPSC-derived BBB model can help elucidate the mechanism by which ZIKV can infect BECs, cross the BBB and gain access to the CNS.
Collapse
Affiliation(s)
- Judie B. Alimonti
- Human Health Therapeutics Research Center, National Research Council of Canada, 100 Sussex Dr., Ottawa, ON Canada
| | - Maria Ribecco-Lutkiewicz
- Human Health Therapeutics Research Center, National Research Council of Canada, Bldg M54-1200 Montreal Rd., Ottawa, ON K1A 0R6 Canada
| | - Caroline Sodja
- Human Health Therapeutics Research Center, National Research Council of Canada, Bldg M54-1200 Montreal Rd., Ottawa, ON K1A 0R6 Canada
| | - Anna Jezierski
- Human Health Therapeutics Research Center, National Research Council of Canada, Bldg M54-1200 Montreal Rd., Ottawa, ON K1A 0R6 Canada
| | - Danica B. Stanimirovic
- Human Health Therapeutics Research Center, National Research Council of Canada, Bldg M54-1200 Montreal Rd., Ottawa, ON K1A 0R6 Canada
| | - Qing Liu
- Human Health Therapeutics Research Center, National Research Council of Canada, Bldg M54-1200 Montreal Rd., Ottawa, ON K1A 0R6 Canada
| | - Arsalan S. Haqqani
- Human Health Therapeutics Research Center, National Research Council of Canada, 100 Sussex Dr., Ottawa, ON Canada
| | - Wayne Conlan
- Human Health Therapeutics Research Center, National Research Council of Canada, 100 Sussex Dr., Ottawa, ON Canada
| | - Mahmud Bani-Yaghoub
- Human Health Therapeutics Research Center, National Research Council of Canada, Bldg M54-1200 Montreal Rd., Ottawa, ON K1A 0R6 Canada
| |
Collapse
|
95
|
da Silva LRC. Zika Virus Trafficking and Interactions in the Human Male Reproductive Tract. Pathogens 2018; 7:E51. [PMID: 29751638 PMCID: PMC6027493 DOI: 10.3390/pathogens7020051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/28/2022] Open
Abstract
Sexual transmission of Zika virus (ZIKV) is a matter of great concern. Infectious viral particles can be shed in semen for as long as six months after infection and can be transferred to male and female sexual partners during unprotected sexual intercourse. The virus can be found inside spermatozoa and could be directly transferred to the oocyte during fertilization. Sexual transmission of ZIKV can contribute to the rise in number of infected individuals in endemic areas as well as in countries where the mosquito vector does not thrive. There is also the possibility, as has been demonstrated in mouse models, that the vaginal deposition of ZIKV particles present in semen could lead to congenital syndrome. In this paper, we review the current literature to understand ZIKV trafficking from the bloodstream to the human male reproductive tract and viral interactions with host cells in interstitial spaces, tubule walls, annexed glands and semen. We hope to highlight gaps to be filled by future research and potential routes for vaccine and antiviral development.
Collapse
|
96
|
Lee I, Bos S, Li G, Wang S, Gadea G, Desprès P, Zhao RY. Probing Molecular Insights into Zika Virus⁻Host Interactions. Viruses 2018; 10:v10050233. [PMID: 29724036 PMCID: PMC5977226 DOI: 10.3390/v10050233] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 12/13/2022] Open
Abstract
The recent Zika virus (ZIKV) outbreak in the Americas surprised all of us because of its rapid spread and association with neurologic disorders including fetal microcephaly, brain and ocular anomalies, and Guillain–Barré syndrome. In response to this global health crisis, unprecedented and world-wide efforts are taking place to study the ZIKV-related human diseases. Much has been learned about this virus in the areas of epidemiology, genetic diversity, protein structures, and clinical manifestations, such as consequences of ZIKV infection on fetal brain development. However, progress on understanding the molecular mechanism underlying ZIKV-associated neurologic disorders remains elusive. To date, we still lack a good understanding of; (1) what virologic factors are involved in the ZIKV-associated human diseases; (2) which ZIKV protein(s) contributes to the enhanced viral pathogenicity; and (3) how do the newly adapted and pandemic ZIKV strains alter their interactions with the host cells leading to neurologic defects? The goal of this review is to explore the molecular insights into the ZIKV–host interactions with an emphasis on host cell receptor usage for viral entry, cell innate immunity to ZIKV, and the ability of ZIKV to subvert antiviral responses and to cause cytopathic effects. We hope this literature review will inspire additional molecular studies focusing on ZIKV–host Interactions.
Collapse
Affiliation(s)
- Ina Lee
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Sandra Bos
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France.
| | - Ge Li
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Shusheng Wang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Gilles Gadea
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France.
| | - Philippe Desprès
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France.
| | - Richard Y Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
97
|
Zeng M, Chen S, Zhang J, Wu Z, Wang M, Jia R, Zhu D, Liu M, Sun K, Yang Q, Wu Y, Zhao X, Cheng A. Molecular identification of goose (Anser cygnoide) suppressor ubiquitin-specific protease 18 (USP18) and the effects of goose IFN and TMUV on its comparative transcripts. Poult Sci 2018; 97:1022-1031. [PMID: 29267974 DOI: 10.3382/ps/pex322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 10/11/2017] [Indexed: 11/20/2022] Open
Abstract
Ubiquitin-specific protease 18 (USP18) is known as an inhibition factor and has been associated with the innate immune response to pathogens. USP18 is the only deconjugating protease with specificity for interferon-stimulated gene 15 (ISG15), which is supposed to be missing in birds. To analyze the efficacy of goose USP18 (goUSP18) against Tembusu virus (TMUV) infection, we first cloned USP18 homologous cDNA from TMUV infected geese. The coding sequence was 1131 bp, and the deduced amino acid sequence shared conserved motifs with its homologues. Tissue-specific expression has shown that goUSP18 transcripts are strongly expressed in the spleen and liver of adult geese, as well as in the pancreas of goslings. Moreover, the goUSP18 transcripts were induced by goose interferons (goIFN) in goose embryo fibroblasts (GEF) and by TLR ligands in peripheral blood mononuclear cells (PBMC). Notably, goUSP18 transcripts were highly up-regulated by TMUV infection compared to the basal level in uninfected birds. Taken together, these results suggested that goUSP18 was involved in host innate immunity against TMUV infection.
Collapse
Affiliation(s)
- M Zeng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - S Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - J Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Z Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - M Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - R Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - D Zhu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - M Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - K Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Q Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Y Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - X Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - A Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
98
|
Papa MP, Meuren LM, Coelho SVA, Lucas CGDO, Mustafá YM, Lemos Matassoli F, Silveira PP, Frost PS, Pezzuto P, Ribeiro MR, Tanuri A, Nogueira ML, Campanati L, Bozza MT, Paula Neto HA, Pimentel-Coelho PM, Figueiredo CP, de Aguiar RS, de Arruda LB. Zika Virus Infects, Activates, and Crosses Brain Microvascular Endothelial Cells, without Barrier Disruption. Front Microbiol 2017; 8:2557. [PMID: 29312238 PMCID: PMC5743735 DOI: 10.3389/fmicb.2017.02557] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 12/08/2017] [Indexed: 12/19/2022] Open
Abstract
Zika virus (ZIKV) has been associated to central nervous system (CNS) harm, and virus was detected in the brain and cerebrospinal fluids of microcephaly and meningoencephalitis cases. However, the mechanism by which the virus reaches the CNS is unclear. Here, we addressed the effects of ZIKV replication in human brain microvascular endothelial cells (HBMECs), as an in vitro model of blood brain barrier (BBB), and evaluated virus extravasation and BBB integrity in an in vivo mouse experimental model. HBMECs were productively infected by African and Brazilian ZIKV strains (ZIKVMR766 and ZIKVPE243), which induce increased production of type I and type III IFN, inflammatory cytokines and chemokines. Infection with ZIKVMR766 promoted earlier cellular death, in comparison to ZIKVPE243, but infection with either strain did not result in enhanced endothelial permeability. Despite the maintenance of endothelial integrity, infectious virus particles crossed the monolayer by endocytosis/exocytosis-dependent replication pathway or by transcytosis. Remarkably, both viruses' strains infected IFNAR deficient mice, with high viral load being detected in the brains, without BBB disruption, which was only detected at later time points after infection. These data suggest that ZIKV infects and activates endothelial cells, and might reach the CNS through basolateral release, transcytosis or transinfection processes. These findings further improve the current knowledge regarding ZIKV dissemination pathways.
Collapse
Affiliation(s)
- Michelle P. Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lana M. Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sharton V. A. Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina G. de Oliveira Lucas
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yasmin M. Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavio Lemos Matassoli
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola P. Silveira
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula S. Frost
- Núcleo de Neurociências da Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Pezzuto
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milene R. Ribeiro
- Laboratório de Pesquisas em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauricio L. Nogueira
- Laboratório de Pesquisas em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
| | - Loraine Campanati
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor A. Paula Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro M. Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia P. Figueiredo
- Núcleo de Neurociências da Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato S. de Aguiar
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana B. de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
99
|
Medina MT, Medina-Montoya M. New spectrum of the neurologic consequences of Zika. J Neurol Sci 2017; 383:214-215. [DOI: 10.1016/j.jns.2017.10.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 02/02/2023]
|
100
|
Kalkeri R, Murthy KK. Zika virus reservoirs: Implications for transmission, future outbreaks, drug and vaccine development. F1000Res 2017; 6:1850. [PMID: 29225778 PMCID: PMC5710464 DOI: 10.12688/f1000research.12695.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/12/2017] [Indexed: 12/23/2022] Open
Abstract
Zika virus (ZIKV) was recently declared as a 'Global Health Emergency' by the World Health Organization. Various tissue reservoirs of ZIKV in infected humans and animals models have been observed, the implications of which are not known. Compared to other Flaviviruses, sexual transmission and persistence in the genitourinary tract seem to be unique to ZIKV. ZIKV persistence and shedding in bodily secretions (e.g. saliva, semen) is a concern for potential disease spread and could pose challenges in diagnosis, regulatory guidelines and drug/vaccine development. Murine and non-human primate models could be useful to study the role of tissue reservoirs in the development of prophylactic or therapeutic strategies. There is a need for meta-analysis of the ZIKV infection and virus shedding data from infected patients and ZIKV animal models, and additional research is needed to fully comprehend the long term implications of tissue reservoirs on ZIKV disease pathogenesis and biology.
Collapse
Affiliation(s)
- Raj Kalkeri
- Infectious Diseases Research, Southern Research, Frederick, MD, 21701, USA
| | | |
Collapse
|