1
|
Cenci Dietrich V, Costa JMC, Oliveira MMGL, Aguiar CEO, Silva LGDO, Luz MS, Lemos FFB, de Melo FF. Pathogenesis and clinical management of arboviral diseases. World J Virol 2025; 14:100489. [DOI: 10.5501/wjv.v14.i1.100489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Arboviral diseases are viral infections transmitted to humans through the bites of arthropods, such as mosquitoes, often causing a variety of pathologies associated with high levels of morbidity and mortality. Over the past decades, these infections have proven to be a significant challenge to health systems worldwide, particularly following the considerable geographic expansion of the dengue virus (DENV) and its most recent outbreak in Latin America as well as the difficult-to-control outbreaks of yellow fever virus (YFV), chikungunya virus (CHIKV), and Zika virus (ZIKV), leaving behind a substantial portion of the population with complications related to these infections. Currently, the world is experiencing a period of intense globalization, which, combined with global warming, directly contributes to wider dissemination of arbovirus vectors across the globe. Consequently, all continents remain on high alert for potential new outbreaks. Thus, this review aims to provide a comprehensive understanding of the pathogenesis of the four main arboviruses today (DENV, ZIKV, YFV, and CHIKV) discussing their viral characteristics, immune responses, and mechanisms of viral evasion, as well as important clinical aspects for patient management. This includes associated symptoms, laboratory tests, treatments, existing or developing vaccines and the main associated complications, thus integrating a broad historical, scientific and clinical approach.
Collapse
Affiliation(s)
- Victoria Cenci Dietrich
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Juan Marcos Caram Costa
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | | | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
2
|
Shinde DP, Plante JA, Scharton D, Mitchell B, Walker J, Azar SR, Campos RK, Sacchetto L, Drumond BP, Vasilakis N, Plante KS, Weaver SC. Potential role of heterologous flavivirus immunity in preventing urban transmission of yellow fever virus. Nat Commun 2024; 15:9728. [PMID: 39523371 PMCID: PMC11551182 DOI: 10.1038/s41467-024-54146-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
During the recent yellow fever (YF) epidemics in Brazil, human cases were attributed to spillover infections via sylvatic mosquito transmission. Despite YF virus (YFV) transmission in major urban centers with insufficient vaccination coverage and abundant populations of the domestic vector, Aedes aegypti, there was no evidence of human-amplified transmission. Furthermore, the historic absence of YF in Asia, despite abundant Ae. aegypti and an immunologically naive human population, is unexplained. We tested the hypothesis that pre-existing, heterologous flavivirus immunity, specifically from dengue (DENV) and Zika (ZIKV) viruses, limits YFV viremia and transmission by Ae. aegypti. We infected cynomolgus macaques with DENV or ZIKV, then challenged them 6-9 months later with YFV. We then measured viremia and disease and allowed Ae. aegypti mosquitoes to feed during peak macaque viremia. Although prior heterologous immunity had variable effects on disease, DENV and ZIKV immunity consistently suppressed YFV viremia. Despite no statistical difference due to a small sample size, the suppression in viremia led to a significant reduction in Ae. aegypti infection and a lack of transmission potential. These results support the hypothesis that, in DENV- and ZIKV-endemic regions such as South America and Asia, human flavivirus immunity suppresses YFV human amplification potential, reducing the risk of urban outbreaks.
Collapse
Affiliation(s)
- Divya P Shinde
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Center for Vector-borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Jessica A Plante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Dionna Scharton
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Brooke Mitchell
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Jordyn Walker
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Sasha R Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Tissue Engineering, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Rafael K Campos
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lívia Sacchetto
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Betânia P Drumond
- Deparment of Microbiology-Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nikos Vasilakis
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Center for Vector-borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Kenneth S Plante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| | - Scott C Weaver
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA.
- Center for Vector-borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
3
|
Kim N, Choi H, Kim U, Kim S, Kim YB, Shin HY. Sustained Microglial Activation Promotes Synaptic Loss and Neuronal Dysfunction after Recovery from ZIKV Infection. Int J Mol Sci 2024; 25:9451. [PMID: 39273400 PMCID: PMC11394746 DOI: 10.3390/ijms25179451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Zika virus (ZIKV), transmitted by Aedes mosquitoes, has been a global health concern since 2007. It primarily causes fetal microcephaly and neuronal defects through maternal transmission and induces neurological complications in adults. Recent studies report elevated proinflammatory cytokines and persistent neurological alterations post recovery, but the in vivo mechanisms remain unclear. In our study, viral RNA loads in the brains of mice infected with ZIKV peaked at 7 days post infection and returned to baseline by day 21, indicating recovery. RNA sequencing of the cerebral cortex at 7 and 21 days revealed upregulated genes related to neuroinflammation and microglial activation. Histological analyses indicated neuronal cell death and altered neurite morphology owing to severe neuroinflammation. Additionally, sustained microglial activation was associated with increased phospho-Tau levels, constituting a marker of neurodegeneration. These findings highlight how persistent microglial activation leads to neuronal dysfunction post ZIKV recovery, providing insights into the molecular pathogenesis of ZIKV-induced brain abnormalities.
Collapse
Affiliation(s)
- Nahyun Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Hanul Choi
- Department of Bio-Industrial Technologies, Konkuk University, Seoul 05029, Republic of Korea
| | - Uijin Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Suyeon Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
- Department of Bio-Industrial Technologies, Konkuk University, Seoul 05029, Republic of Korea
| | - Ha Youn Shin
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
4
|
Hernández-Sarmiento LJ, Valdés-López JF, Urcuqui-Inchima S. Zika virus infection suppresses CYP24A1 and CAMP expression in human monocytes. Arch Virol 2024; 169:135. [PMID: 38839691 PMCID: PMC11153301 DOI: 10.1007/s00705-024-06050-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/27/2024] [Indexed: 06/07/2024]
Abstract
Monocytes are the primary targets of Zika virus (ZIKV) and are associated with ZIKV pathogenesis. Currently, there is no effective treatment for ZIKV infection. It is known that 1,25-dihydroxy vitamin D3 (VitD3) has strong antiviral activity in dengue virus-infected macrophages, but it is unknown whether VitD3 inhibits ZIKV infection in monocytes. We investigated the relationship between ZIKV infection and the expression of genes of the VitD3 pathway, as well as the inflammatory response of infected monocytes in vitro. ZIKV replication was evaluated using a plaque assay, and VitD3 pathway gene expression was analyzed by RT-qPCR. Pro-inflammatory cytokines/chemokines were quantified using ELISA. We found that VitD3 did not suppress ZIKV replication. The results showed a significant decrease in the expression of vitamin D3 receptor (VDR), cytochrome P450 family 24 subfamily A member 1 (CYP24A1), and cathelicidin antimicrobial peptide (CAMP) genes upon ZIKV infection. Treatment with VitD3 was unable to down-modulate production of pro-inflammatory cytokines, except TNF-α, and chemokines. This suggests that ZIKV infection inhibits the expression of VitD3 pathway genes, thereby preventing VitD3-dependent inhibition of viral replication and the inflammatory response. This is the first study to examine the effects of VitD3 in the context of ZIKV infection, and it has important implications for the role of VitD3 in the control of viral replication and inflammatory responses during monocyte infection.
Collapse
Affiliation(s)
| | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
5
|
Shinde DP, Plante JA, Scharton D, Mitchell B, Walker J, Azar SR, Campos RK, Sacchetto L, Drumond BP, Vasilakis N, Plante KS, Weaver SC. Yellow Fever Emergence: Role of Heterologous Flavivirus Immunity in Preventing Urban Transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583168. [PMID: 38463973 PMCID: PMC10925309 DOI: 10.1101/2024.03.03.583168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
During major, recent yellow fever (YF) epidemics in Brazil, human cases were attributed only to spillover infections from sylvatic transmission with no evidence of human amplification. Furthermore, the historic absence of YF in Asia, despite abundant peridomestic Aedes aegypti and naive human populations, represents a longstanding enigma. We tested the hypothesis that immunity from dengue (DENV) and Zika (ZIKV) flaviviruses limits YF virus (YFV) viremia and transmission by Ae. aegypti . Prior DENV and ZIKV immunity consistently suppressed YFV viremia in experimentally infected macaques, leading to reductions in Ae. aegypti infection when mosquitoes were fed on infected animals. These results indicate that, in DENV- and ZIKV-endemic regions such as South America and Asia, flavivirus immunity suppresses YFV human amplification potential, reducing the risk of urban outbreaks. One-Sentence Summary Immunity from dengue and Zika viruses suppresses yellow fever viremia, preventing infection of mosquitoes and reducing the risk of epidemics.
Collapse
|
6
|
de Sales-Neto JM, Madruga Carvalho DC, Arruda Magalhães DW, Araujo Medeiros AB, Soares MM, Rodrigues-Mascarenhas S. Zika virus: Antiviral immune response, inflammation, and cardiotonic steroids as antiviral agents. Int Immunopharmacol 2024; 127:111368. [PMID: 38103408 DOI: 10.1016/j.intimp.2023.111368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne virus first reported from humans in Nigeria in 1954. The first outbreak occurred in Micronesia followed by an outbreak in French Polynesia and another in Brazil when the virus was associated with numerous cases of severe neurological manifestations such as Guillain-Barre syndrome in adults and congenital zika syndrome in fetuses, particularly congenital microcephaly. Innate immunity is the first line of defense against ZIKV through triggering an antiviral immune response. Along with innate immune responses, a sufficient balance between anti- and pro-inflammatory cytokines and the amount of these cytokines are triggered to enhance the antiviral responses. Here, we reviewed the complex interplay between the mediators and signal pathways that coordinate antiviral immune response and inflammation as a key to understanding the development of the underlying diseases triggered by ZIKV. In addition, we summarize current and new therapeutic strategies for ZIKV infection, highlighting cardiotonic steroids as antiviral drugs for the development of this agent.
Collapse
Affiliation(s)
- José Marreiro de Sales-Neto
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | | | | | - Mariana Mendonça Soares
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Sandra Rodrigues-Mascarenhas
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil.
| |
Collapse
|
7
|
Yue Z, Zhang X, Gu Y, Liu Y, Lan LM, Liu Y, Li Y, Yang G, Wan P, Chen X. Regulation and functions of the NLRP3 inflammasome in RNA virus infection. Front Cell Infect Microbiol 2024; 13:1309128. [PMID: 38249297 PMCID: PMC10796458 DOI: 10.3389/fcimb.2023.1309128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Virus infection is one of the greatest threats to human life and health. In response to viral infection, the host's innate immune system triggers an antiviral immune response mostly mediated by inflammatory processes. Among the many pathways involved, the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome has received wide attention in the context of viral infection. The NLRP3 inflammasome is an intracellular sensor composed of three components, including the innate immune receptor NLRP3, adaptor apoptosis-associated speck-like protein containing CARD (ASC), and the cysteine protease caspase-1. After being assembled, the NLRP3 inflammasome can trigger caspase-1 to induce gasdermin D (GSDMD)-dependent pyroptosis, promoting the maturation and secretion of proinflammatory cytokines such as interleukin-1 (IL-1β) and interleukin-18 (IL-18). Recent studies have revealed that a variety of viruses activate or inhibit the NLRP3 inflammasome via viral particles, proteins, and nucleic acids. In this review, we present a variety of regulatory mechanisms and functions of the NLRP3 inflammasome upon RNA viral infection and demonstrate multiple therapeutic strategies that target the NLRP3 inflammasome for anti-inflammatory effects in viral infection.
Collapse
Affiliation(s)
- Zhaoyang Yue
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Xuelong Zhang
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yu Gu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ying Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Lin-Miaoshen Lan
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yilin Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yongkui Li
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Pin Wan
- Foshan Institute of Medical Microbiology, Foshan, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Xin Chen
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| |
Collapse
|
8
|
Connors J, Cusimano G, Mege N, Woloszczuk K, Konopka E, Bell M, Joyner D, Marcy J, Tardif V, Kutzler MA, Muir R, Haddad EK. Using the power of innate immunoprofiling to understand vaccine design, infection, and immunity. Hum Vaccin Immunother 2023; 19:2267295. [PMID: 37885158 PMCID: PMC10760375 DOI: 10.1080/21645515.2023.2267295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
In the field of immunology, a systems biology approach is crucial to understanding the immune response to infection and vaccination considering the complex interplay between genetic, epigenetic, and environmental factors. Significant progress has been made in understanding the innate immune response, including cell players and critical signaling pathways, but many questions remain unanswered, including how the innate immune response dictates host/pathogen responses and responses to vaccines. To complicate things further, it is becoming increasingly clear that the innate immune response is not a linear pathway but is formed from complex networks and interactions. To further our understanding of the crosstalk and complexities, systems-level analyses and expanded experimental technologies are now needed. In this review, we discuss the most recent immunoprofiling techniques and discuss systems approaches to studying the global innate immune landscape which will inform on the development of personalized medicine and innovative vaccine strategies.
Collapse
Affiliation(s)
- Jennifer Connors
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Gina Cusimano
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Nathan Mege
- Tower Health, Reading Hospital, West Reading, PA, USA
| | - Kyra Woloszczuk
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Emily Konopka
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Matthew Bell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - David Joyner
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Molecular and Cellular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jennifer Marcy
- Department of Molecular and Cellular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Virginie Tardif
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michele A. Kutzler
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Roshell Muir
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Family, Community, and Preventative Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Elias K. Haddad
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
9
|
Ravindran S, Lahon A. Tropism and immune response of chikungunya and zika viruses: An overview. Cytokine 2023; 170:156327. [PMID: 37579710 DOI: 10.1016/j.cyto.2023.156327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023]
Abstract
Zika virus (ZIKV) and chikungunya virus (CHIKV) are two medically important vector-borne viruses responsible for causing significant disease burden in humans, including neurological sequelae/complications. Besides sharing some common clinical features, ZIKV has major shares in causing microcephaly and brain malformations in developing foetus, whereas CHIKV causes chronic joint pain/swelling in infected individuals. Both viruses have a common route of entry to the host body. i.e., dermal site of inoculation through the bite of an infected mosquito and later taken up by different immune cells for further dissemination to other areas of the host body that lead to a range of immune responses via different pathways. The immune responses generated by both viruses have similar characteristics with varying degrees of inflammation and activation of immune cells. However, the overall response of immune cells is not fully explored in the context of ZIKV and CHIKV infection. The knowledge of cellular tropism and the immune response is the key to understanding the mechanisms of viral immunity and pathogenesis, which may allow to develop novel therapeutic strategies for these viral infections. This review aims to discuss recent advancements and identify the knowledge gaps in understanding the mechanism of cellular tropism and immune response of CHIKV and ZIKV.
Collapse
Affiliation(s)
- Shilpa Ravindran
- Institute of Advanced Virology, Thiruvananthapuram, Kerala 695317, India
| | - Anismrita Lahon
- Institute of Advanced Virology, Thiruvananthapuram, Kerala 695317, India.
| |
Collapse
|
10
|
da Costa HHM, Bielavsky M, Orts DJB, Araujo S, Adriani PP, Nogueira JS, Astray RM, Pandey RP, Lancellotti M, Cunha-Junior JP, Prudencio CR. Production of Recombinant Zika Virus Envelope Protein by Airlift Bioreactor as a New Subunit Vaccine Platform. Int J Mol Sci 2023; 24:13955. [PMID: 37762254 PMCID: PMC10531330 DOI: 10.3390/ijms241813955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 09/29/2023] Open
Abstract
The Zika Virus (ZIKV) is an emerging arbovirus of great public health concern, particularly in the Americas after its last outbreak in 2015. There are still major challenges regarding disease control, and there is no ZIKV vaccine currently approved for human use. Among many different vaccine platforms currently under study, the recombinant envelope protein from Zika Virus (rEZIKV) constitutes an alternative option for vaccine development and has great potential for monitoring ZIKV infection and antibody response. This study describes a method to obtain a bioactive and functional rEZIKV using an E. coli expression system, with the aid of a 5-L airlift bioreactor and following an automated fast protein liquid chromatography (FPLC) protocol, capable of obtaining high yields of approximately 20 mg of recombinant protein per liter of bacterium cultures. The purified rEZIKV presented preserved antigenicity and immunogenicity. Our results show that the use of an airlift bioreactor for the production of rEZIKV is ideal for establishing protocols and further research on ZIKV vaccines bioprocess, representing a promising system for the production of a ZIKV envelope recombinant protein-based vaccine candidate.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Monica Bielavsky
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Diego J. B. Orts
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Laboratory of Cardiobiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, São Paulo 04023-062, Brazil
| | - Sergio Araujo
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Patrícia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Renato M. Astray
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
- Multi-Purpose Laboratory Butantan Institute, São Paulo 05503-900, Brazil
| | - Ramendra P. Pandey
- School of Health Sciences and Technology, UPES University, Dehradun 248007, Uttarakhand, India
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences—FCF, University of Campinas—UNICAMP, Campinas 13083-871, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
11
|
de Jesús López Medina Y, Tamayo-Molina YS, Valdés-López JF, Urcuqui-Inchima S. Protective Effects of Caffeine on Chikungunya and Zika Virus Infections: An in Vitro and in Silico Study. Chem Biodivers 2023; 20:e202300192. [PMID: 37489706 DOI: 10.1002/cbdv.202300192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 07/26/2023]
Abstract
Infection by viruses Chikungunya (CHIKV) and Zika (ZIKV) continue to be serious problems in tropical and subtropical areas of the world. Here, we evaluated the antiviral and virucidal activity of caffeine against CHIKV and ZIKV in Vero, A549, and Huh-7 cell lines. Results showed that caffeine displays antiviral properties against both viruses. By pre-and post-infection treatment, caffeine significantly inhibited CHIKV and ZIKV replication in a dose-dependent manner. Furthermore, caffeine showed a virucidal effect against ZIKV. Molecular docking suggests the possible binding of caffeine with envelope protein and RNA-dependent RNA polymerase of CHIKV and ZIKV. This is the first study that showed an antiviral effect of caffeine against CHIKV and ZIKV. Although further studies are needed to better understand the mechanism of caffeine-mediated repression of viral replication, caffeine appears to be a promising compound that could be used for in vivo studies, perhaps in synergy with other compounds present in daily beverages.
Collapse
Affiliation(s)
| | | | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| |
Collapse
|
12
|
Silva AJD, de Jesus ALS, Leal LRS, de Macêdo LS, da Silva Barros BR, de Sousa GF, da Paz Leôncio Alves S, Pena LJ, de Melo CML, de Freitas AC. Whole Yeast Vaccine Displaying ZIKV B and T Cell Epitopes Induces Cellular Immune Responses in the Murine Model. Pharmaceutics 2023; 15:1898. [PMID: 37514084 PMCID: PMC10385271 DOI: 10.3390/pharmaceutics15071898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Improving antigen presentation is crucial for the success of immunization strategies. Yeasts are classically used as biofactories to produce recombinant proteins and are efficient vehicles for antigen delivery, in addition to their adjuvant properties. Despite the absence of epidemic outbreaks, several vaccine approaches continue to be developed for Zika virus infection. The development of these prophylactic strategies is fundamental given the severity of clinical manifestations, mainly due to viral neurotropism. The present study aimed to evaluate in vivo the immune response induced by P. pastoris recombinant strains displaying epitopes of the envelope (ENV) and NS1 ZIKV proteins. Intramuscular immunization with heat-attenuated yeast enhanced the secretion of IL-6, TNF-α, and IFN-γ, in addition to the activation of CD4+ and CD8+ T cells, in BALB/c mice. P. pastoris displaying ENV epitopes induced a more robust immune response, increasing immunoglobulin production, especially IgG isotypes. Both proposed vaccines showed the potential to induce immune responses without adverse effects, confirming the safety of administering P. pastoris as a vaccine vehicle. Here, we demonstrated, for the first time, the evaluation of a vaccine against ZIKV based on a multiepitope construct using yeast as a delivery system and reinforcing the applicability of P. pastoris as a whole-cell vaccine.
Collapse
Affiliation(s)
- Anna Jéssica Duarte Silva
- Laboratory of Molecular Studies and Experimental Therapy-LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | | | - Lígia Rosa Sales Leal
- Laboratory of Molecular Studies and Experimental Therapy-LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Larissa Silva de Macêdo
- Laboratory of Molecular Studies and Experimental Therapy-LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| | | | | | | | - Lindomar José Pena
- Department of Virology and Experimental Therapy, Instituto Aggeu Magalhães, Oswaldo Cruz Foundation, Recife 50670-901, Brazil
| | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy-LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil
| |
Collapse
|
13
|
Petphong V, Kosoltanapiwat N, Limkittikul K, Maneekan P, Chatchen S, Jittmittraphap A, Sriburin P, Chattanadee S, Leaungwutiwong P. Detection of Anti-ZIKV NS1 IgA, IgM, and Combined IgA/IgM and Identification of IL-4 and IL-10 as Potential Biomarkers for Early ZIKV and DENV Infections in Hyperendemic Regions, Thailand. Trop Med Infect Dis 2023; 8:tropicalmed8050284. [PMID: 37235332 DOI: 10.3390/tropicalmed8050284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The frequency of Zika virus (ZIKV)-specific IgA and IgM and the cytokine expression profile of ZIKV-infected patients in hyperendemic areas remain unclear. This study investigated the rates of ZIKV non-structural protein 1 (NS1)-specific IgA and IgM and evaluated serum cytokine levels of ZIKV and Dengue virus (DENV) cases in Thailand to identify potential diagnostic biomarkers, elucidate the immunity against ZIKV and DENV, and investigate the association between cytokine levels and ZIKV symptoms. Low rates of positivity for ZIKV NS1-specific IgA and IgM were detected in our study. ZIKV NS1 IgA/M (11%, 11/101) in combination was more frequently detected than ZIKV NS1 IgM (2%, 2/101) or ZIKV NS1 IgA (4%, 4/96) alone, especially in acute ZIKV cases with previous DENV exposure (14%, 10/72). Cytokine analysis showed that both ZIKV and DENV infections induced polyfunctional immunity, and the latter triggered more prolonged responses. The existence of significant differences in IL-4 and IL-10 levels between acute ZIKV and acute DENV cases suggested that IL-4 (p = 0.0176) and IL-10 (p = 0.0003) may represent biomarkers for acute ZIKV and acute DENV infections, respectively. Analysis of the association between increased cytokine levels and ZIKV symptoms indicated that CXCL10 (p = 0.0029) was associated with exanthema, while IL-5 (p = 0.0496) was linked to headache. The detection of ZIKV NS1 IgA and IgM in combination may enhance the diagnosis of early ZIKV infection, particularly when levels of IgM or IgA alone are low or undetectable. IL-4 and IL-10 may serve as targets for the development of diagnostic tools to detect ZIKV and DENV infections early, respectively, in flavivirus-endemic regions.
Collapse
Affiliation(s)
- Vajee Petphong
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Nathamon Kosoltanapiwat
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Kriengsak Limkittikul
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Pannamas Maneekan
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Supawat Chatchen
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Akanitt Jittmittraphap
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Pimolpachr Sriburin
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Siriporn Chattanadee
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Pornsawan Leaungwutiwong
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
14
|
Fernandez GJ, Ramírez-Mejía JM, Castillo JA, Urcuqui-Inchima S. Vitamin D modulates expression of antimicrobial peptides and proinflammatory cytokines to restrict Zika virus infection in macrophages. Int Immunopharmacol 2023; 119:110232. [PMID: 37150017 DOI: 10.1016/j.intimp.2023.110232] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/09/2023]
Abstract
Although the impact of Zika virus (ZIKV) infection on human health has been well documented, we still have no vaccine or effective treatment. This fact highlights the importance of searching for alternative therapy for treating ZIKV. To search for ZIKV antivirals, we examined the effect of vitamin D in monocyte-derived macrophages (MDMs) differentiated in the presence of vitamin D (D3-MDM) and explored the molecular mechanisms by analyzing transcriptional profiles. Our data show the restriction of ZIKV infection in D3-MDMs as compared to MDMs. Transcriptional profiles show that vitamin D alters about 19% of Zika response genes (8.2% diminished and 10.8% potentiated). Among the genes with diminished expression levels, we found proinflammatory cytokines and chemokines such as IL6, TNF, IL1A, IL1B, and IL12B, CCL1, CCL4, CCL7, CXCL3, CXCL6, and CXCL8. On the other hand, genes with potentiated expression were related to degranulation such as Lysozyme, cathelicidin (CAMP), and Serglycin. Since the CAMP gene encodes the antimicrobial peptide LL-37, we treated MDMs with LL-37 and infected them with ZIKV. The results showed a decrease in the proportion of infected cells. Our data provide new insights into the role of vitamin D in restricting ZIKV infection in macrophages that are mediated by induction of cathelicidin/LL-37 expression and downregulation of proinflammatory genes. Results highlight the biological relevance of vitamin D-inducible peptides as an antiviral treatment for Zika fever.
Collapse
Affiliation(s)
- Geysson Javier Fernandez
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia; Grupo Biología y Control de Enfermedades Infecciosas, Universidad de Antioquia UdeA, Medellín, Colombia.
| | - Julieta M Ramírez-Mejía
- CIBIOP Group, Department of Applied Sciences and Engineering, Universidad EAFIT, Medellín, Antioquia, Colombia.
| | - Jorge Andrés Castillo
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia; Grupo de enfermedades infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia.
| |
Collapse
|
15
|
Hernández-Sarmiento LJ, Valdés-López JF, Urcuqui-Inchima S. American-Asian- and African lineages of Zika virus induce differential pro-inflammatory and Interleukin 27-dependent antiviral responses in human monocytes. Virus Res 2023; 325:199040. [PMID: 36610657 PMCID: PMC10194209 DOI: 10.1016/j.virusres.2023.199040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/06/2023]
Abstract
Zika virus (ZIKV) is an arbovirus that belongs to the Flaviviridae family and inflammatory responses play a critical role in ZIKV pathogenesis. As a first-line defense, monocytes are key components of innate immunity and host response to viruses. Monocytes are considered the earliest blood cell type to be infected by ZIKV and have been shown to be associated with ZIKV pathogenesis. The first ZIKV epidemic was reported in Africa and Asia although, it is less well known whether African- and Asian- lineages of ZIKV have different impacts on host immune response. We studied the pro-inflammatory and antiviral response of ZIKV-infected monocytes using publicly available RNA-seq analysis (GSE103114). We compared the transcriptomic profiles of human monocytes infected with ZIKV Puerto Rico strain (PRVABC59), American-Asian lineage, and ZIKV Nigeria strain (IBH30656), African lineage. We validated RNA-seq results by ELISA or RT-qPCR, in human monocytes infected with a clinical isolate of ZIKV from Colombia (American-Asian lineage), or with ZIKV from Dakar (African lineage). The transcriptomic analysis showed that ZIKV Puerto Rico strain promotes a higher pro-inflammatory response through TLR2 signaling and NF-kB activation and induces a strong IL27-dependent antiviral activity than ZIKV Nigeria strain. Furthermore, human monocytes are more susceptible to infection with ZIKV from Colombia than ZIKV from Dakar. Likewise, Colombian ZIKV isolate activated IL27 signaling and induced a robust antiviral response in an IFN-independent manner. Moreover, we show that treatment of monocytes with IL27 results in decreased release of ZIKV particles in a dose-dependent manner with an EC50 =2.870 ng/mL for ZIKV from Colombia and EC50 =10.23 ng/mL to ZIKV from Dakar. These findings highlight the differential inflammatory response and antiviral activity of monocytes infected with different lineages of ZIKV and may help better management of ZIKV-infected patients.
Collapse
Affiliation(s)
| | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
16
|
Wikan N, Potikanond S, Hankittichai P, Thaklaewphan P, Monkaew S, Smith DR, Nimlamool W. Alpinetin Suppresses Zika Virus-Induced Interleukin-1β Production and Secretion in Human Macrophages. Pharmaceutics 2022; 14:pharmaceutics14122800. [PMID: 36559293 PMCID: PMC9782830 DOI: 10.3390/pharmaceutics14122800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/16/2022] Open
Abstract
Zika virus (ZIKV) infection has been recognized to cause adverse sequelae in the developing fetus. Specially, this virus activates the excessive release of IL-1β causing inflammation and altered physiological functions in multiple organs. Although many attempts have been invested to develop vaccine, antiviral, and antibody therapies, development of agents focusing on limiting ZIKV-induced IL-1β release have not gained much attention. We aimed to study the effects of alpinetin (AP) on IL-1β production in human macrophage upon exposure to ZIKV. Our study demonstrated that ZIKV stimulated IL-1β release in the culture supernatant of ZIKV-infected cells, and AP could effectively reduce the level of this cytokine. AP exhibited no virucidal activities against ZIKV nor caused alteration in viral production. Instead, AP greatly inhibited intracellular IL-1β synthesis. Surprisingly, this compound did not inhibit ZIKV-induced activation of NF-κB and its nuclear translocation. However, AP could significantly inhibit ZIKV-induced p38 MAPK activation without affecting the phosphorylation status of ERK1/2 and JNK. These observations suggest the possibility that AP may reduce IL-1β production, in part, through suppressing p38 MAPK signaling. Our current study sheds light on the possibility of using AP as an alternative agent for treating complications caused by ZIKV infection-induced IL-1β secretion.
Collapse
Affiliation(s)
- Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center for Development of Local Lanna Rice and Rice Products, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phateep Hankittichai
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sathit Monkaew
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Duncan R. Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
- Correspondence: (D.R.S.); (W.N.); Tel.: +66-53-934597 (W.N.)
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center for Development of Local Lanna Rice and Rice Products, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: (D.R.S.); (W.N.); Tel.: +66-53-934597 (W.N.)
| |
Collapse
|
17
|
Sekaran SD, Ismail AA, Thergarajan G, Chandramathi S, Rahman SKH, Mani RR, Jusof FF, Lim YAL, Manikam R. Host immune response against DENV and ZIKV infections. Front Cell Infect Microbiol 2022; 12:975222. [PMID: 36159640 PMCID: PMC9492869 DOI: 10.3389/fcimb.2022.975222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 11/15/2022] Open
Abstract
Dengue is a major public health concern, affecting almost 400 million people worldwide, with about 70% of the global burden of disease in Asia. Despite revised clinical classifications of dengue infections by the World Health Organization, the wide spectrum of the manifestations of dengue illness continues to pose challenges in diagnosis and patient management for clinicians. When the Zika epidemic spread through the American continent and then later to Africa and Asia in 2015, researchers compared the characteristics of the Zika infection to Dengue, considering both these viruses were transmitted primarily through the same vector, the Aedes aegypti female mosquitoes. An important difference to note, however, was that the Zika epidemic diffused in a shorter time span compared to the persisting feature of Dengue infections, which is endemic in many Asian countries. As the pathogenesis of viral illnesses is affected by host immune responses, various immune modulators have been proposed as biomarkers to predict the risk of the disease progression to a severe form, at a much earlier stage of the illness. However, the findings for most biomarkers are highly discrepant between studies. Meanwhile, the cross-reactivity of CD8+ and CD4+ T cells response to Dengue and Zika viruses provide important clues for further development of potential treatments. This review discusses similarities between Dengue and Zika infections, comparing their disease transmissions and vectors involved, and both the innate and adaptive immune responses in these infections. Consideration of the genetic identity of both the Dengue and Zika flaviviruses as well as the cross-reactivity of relevant T cells along with the actions of CD4+ cytotoxic cells in these infections are also presented. Finally, a summary of the immune biomarkers that have been reported for dengue and Zika viral infections are discussed which may be useful indicators for future anti-viral targets or predictors for disease severity. Together, this information appraises the current understanding of both Zika and Dengue infections, providing insights for future vaccine design approaches against both viruses.
Collapse
Affiliation(s)
| | - Amni Adilah Ismail
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Gaythri Thergarajan
- Faculty of Medical & Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Samudi Chandramathi
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - S. K. Hanan Rahman
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ravishankar Ram Mani
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Felicita Fedelis Jusof
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yvonne A. L. Lim
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Rishya Manikam
- Department of Trauma and Emergency Medicine, University Malaya Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
18
|
Polonio CM, da Silva P, Russo FB, Hyppolito BRN, Zanluqui NG, Benazzato C, Beltrão-Braga PCB, Muxel SM, Peron JPS. microRNAs Control Antiviral Immune Response, Cell Death and Chemotaxis Pathways in Human Neuronal Precursor Cells (NPCs) during Zika Virus Infection. Int J Mol Sci 2022; 23:ijms231810282. [PMID: 36142200 PMCID: PMC9499039 DOI: 10.3390/ijms231810282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Viral infections have always been a serious burden to public health, increasing morbidity and mortality rates worldwide. Zika virus (ZIKV) is a flavivirus transmitted by the Aedes aegypti vector and the causative agent of severe fetal neuropathogenesis and microcephaly. The virus crosses the placenta and reaches the fetal brain, mainly causing the death of neuronal precursor cells (NPCs), glial inflammation, and subsequent tissue damage. Genetic differences, mainly related to the antiviral immune response and cell death pathways greatly influence the susceptibility to infection. These components are modulated by many factors, including microRNAs (miRNAs). MiRNAs are small noncoding RNAs that regulate post-transcriptionally the overall gene expression, including genes for the neurodevelopment and the formation of neural circuits. In this context, we investigated the pathways and target genes of miRNAs modulated in NPCs infected with ZIKV. We observed downregulation of miR-302b, miR-302c and miR-194, whereas miR-30c was upregulated in ZIKV infected human NPCs in vitro. The analysis of a public dataset of ZIKV-infected human NPCs evidenced 262 upregulated and 3 downregulated genes, of which 142 were the target of the aforementioned miRNAs. Further, we confirmed a correlation between miRNA and target genes affecting pathways related to antiviral immune response, cell death and immune cells chemotaxis, all of which could contribute to the establishment of microcephaly and brain lesions. Here, we suggest that miRNAs target gene expression in infected NPCs, directly contributing to the pathogenesis of fetal microcephaly.
Collapse
Affiliation(s)
- Carolina M. Polonio
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil
- Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-000, Brazil
| | - Patrick da Silva
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil
- Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-000, Brazil
| | - Fabiele B. Russo
- Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-000, Brazil
- Disease Modeling Laboratory at Department of Microbiology, Institute of Biomedical Sciences, São Paulo 05508-000, Brazil
| | - Brendo R. N. Hyppolito
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil
- Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo, São Paulo 05508-000, Brazil
| | - Nagela G. Zanluqui
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil
- Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-000, Brazil
- Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo, São Paulo 05508-000, Brazil
| | - Cecília Benazzato
- Disease Modeling Laboratory at Department of Microbiology, Institute of Biomedical Sciences, São Paulo 05508-000, Brazil
| | - Patrícia C. B. Beltrão-Braga
- Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-000, Brazil
- Disease Modeling Laboratory at Department of Microbiology, Institute of Biomedical Sciences, São Paulo 05508-000, Brazil
| | - Sandra M. Muxel
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil
- Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-000, Brazil
- Correspondence: (S.M.M.); (J.P.S.P.)
| | - Jean Pierre S. Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil
- Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-000, Brazil
- Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo, São Paulo 05508-000, Brazil
- Correspondence: (S.M.M.); (J.P.S.P.)
| |
Collapse
|
19
|
Zhao Z, Li Q, Ashraf U, Yang M, Zhu W, Gu J, Chen Z, Gu C, Si Y, Cao S, Ye J. Zika virus causes placental pyroptosis and associated adverse fetal outcomes by activating GSDME. eLife 2022; 11:73792. [PMID: 35972780 PMCID: PMC9381041 DOI: 10.7554/elife.73792] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 07/27/2022] [Indexed: 11/24/2022] Open
Abstract
Zika virus (ZIKV) can be transmitted from mother to fetus during pregnancy, causing adverse fetal outcomes. Several studies have indicated that ZIKV can damage the fetal brain directly; however, whether the ZIKV-induced maternal placental injury contributes to adverse fetal outcomes is sparsely defined. Here, we demonstrated that ZIKV causes the pyroptosis of placental cells by activating the executor gasdermin E (GSDME) in vitro and in vivo. Mechanistically, TNF-α release is induced upon the recognition of viral genomic RNA by RIG-I, followed by activation of caspase-8 and caspase-3 to ultimately escalate the GSDME cleavage. Further analyses revealed that the ablation of GSDME or treatment with TNF-α receptor antagonist in ZIKV-infected pregnant mice attenuates placental pyroptosis, which consequently confers protection against adverse fetal outcomes. In conclusion, our study unveils a novel mechanism of ZIKV-induced adverse fetal outcomes via causing placental cell pyroptosis, which provides new clues for developing therapies for ZIKV-associated diseases.
Collapse
Affiliation(s)
- Zikai Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qi Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Usama Ashraf
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mengjie Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wenjing Zhu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jun Gu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zheng Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changqin Gu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Youhui Si
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shengbo Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jing Ye
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
20
|
Chen C, Chen A, Yang Y. A diversified role for γδT cells in vector-borne diseases. Front Immunol 2022; 13:965503. [PMID: 36052077 PMCID: PMC9424759 DOI: 10.3389/fimmu.2022.965503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
Vector-borne diseases have high morbidity and mortality and are major health threats worldwide. γδT cells represent a small but essential subpopulation of T cells. They reside in most human tissues and exert important functions in both natural and adaptive immune responses. Emerging evidence have shown that the activation and expansion of γδT cells invoked by pathogens play a diversified role in the regulation of host-pathogen interactions and disease progression. A better understanding of such a role for γδT cells may contribute significantly to developing novel preventative and therapeutic strategies. Herein, we summarize recent exciting findings in the field, with a focus on the role of γδT cells in the infection of vector-borne pathogens.
Collapse
Affiliation(s)
- Chen Chen
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Chen Chen, ; Yanan Yang,
| | - Aibao Chen
- Department of Cell Biology, School of Life Sciences, Anhui Medical University, Hefei, China
| | - Yanan Yang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Chen Chen, ; Yanan Yang,
| |
Collapse
|
21
|
Jaylet T, Quintens R, Benotmane MA, Luukkonen J, Tanaka IB, Ibanez C, Durand C, Sachana M, Azimzadeh O, Adam-Guillermin C, Tollefsen KE, Laurent O, Audouze K, Armant O. Development of an Adverse Outcome Pathway for radiation-induced microcephaly via expert consultation and machine learning. Int J Radiat Biol 2022; 98:1752-1762. [PMID: 35947014 DOI: 10.1080/09553002.2022.2110312] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Brain development during embryogenesis and in early postnatal life is particularly complex and involves the interplay of many cellular processes and molecular mechanisms, making it extremely vulnerable to exogenous insults, including ionizing radiation (IR). Microcephaly is one of the most frequent neurodevelopmental abnormalities that is characterized by small brain size, and is often associated with intellectual deficiency. Decades of research span from epidemiological data on in utero exposure of the A-bomb survivors, to studies on animal and cellular models that allowed deciphering the most prominent molecular mechanisms leading to microcephaly. The Adverse Outcome Pathway (AOP) framework is used to organize, evaluate and portray the scientific knowledge of toxicological effects spanning different biological levels of organizations, from the initial interaction with molecular targets to the occurrence of a disease or adversity. In the present study, the framework was used in an attempt to organize the current scientific knowledge on microcephaly progression in the context of ionizing radiation (IR) exposure. This work was performed by a group of experts formed during a recent workshop organized jointly by the Multidisciplinary European Low Dose Initiative (MELODI) and the European Radioecology Alliance (ALLIANCE) associations to present the AOP approach and tools. Here we report on the development of a putative AOP for congenital microcephaly resulting from IR exposure based on discussions of the working group and we emphasize the use of a novel machine-learning approach to assist in the screening of the available literature to develop AOPs. CONCLUSION The expert consultation led to the identification of crucial biological events for the progression of microcephaly upon exposure to IR, and highlighted current knowledge gaps. The machine learning approach was successfully used to screen the existing knowledge and helped to rapidly screen the body of evidence and in particular the epidemiological data. This systematic review approach also ensured that the analysis was sufficiently comprehensive to identify the most relevant data and facilitate rapid and consistent AOP development. We anticipate that as machine learning approaches become more user-friendly through easy-to-use web interface, this would allow AOP development to become more efficient and less time consuming.
Collapse
Affiliation(s)
- Thomas Jaylet
- Université Paris Cité, T3S, Inserm UMRS 1124, Paris, France
| | - Roel Quintens
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK-CEN, Mol, Belgium
| | | | - Jukka Luukkonen
- University of Eastern Finland, Kuopio Campus, Department of Environmental and Biological Sciences, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Ignacia Braga Tanaka
- Department of Radiobiology, Institute for Environmental Sciences, 1-7 lenomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Chrystelle Ibanez
- PSE-SANTE/SESANE/LRTOX Institut de Radioprotection et de Sûreté Nucléaire (IRSN), F-92262, Fontenay-aux-Roses, France
| | - Christelle Durand
- PSE-SANTE/SESANE/LRTOX Institut de Radioprotection et de Sûreté Nucléaire (IRSN), F-92262, Fontenay-aux-Roses, France
| | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), Environment Health and Safety Division, 75775 CEDEX 16 Paris, France
| | - Omid Azimzadeh
- Federal Office for Radiation Protection (Bfs), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Christelle Adam-Guillermin
- PSE-SANTE/SDOS/LMDN, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Cadarache, 13115 Saint-Paul-Lez-Durance, France
| | - Knut Erik Tollefsen
- Norwegian Institute for Water Research (NIVA), Økernveien 94, N-0579, Oslo, Norway.,Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway.,Centre for Environmental Radioactivity, Norwegian University of Life Sciences (NMBU), Post box 5003, N-1432 Ås, Norway
| | - Olivier Laurent
- PSE-SANTE/SESANE/LEPID, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), F-92262, Fontenay-aux-Roses, France
| | - Karine Audouze
- Université Paris Cité, T3S, Inserm UMRS 1124, Paris, France
| | - Olivier Armant
- PSE-ENV/SRTE/LECO, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Cadarache, 13115 Saint-Paul-Lez-Durance, France
| |
Collapse
|
22
|
Zika Virus Replication in a Mast Cell Model is Augmented by Dengue Virus Antibody-Dependent Enhancement and Features a Selective Immune Mediator Secretory Profile. Microbiol Spectr 2022; 10:e0177222. [PMID: 35862953 PMCID: PMC9431662 DOI: 10.1128/spectrum.01772-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibodies generated against one dengue serotype can enhance infection of another by a phenomenon called antibody-dependent enhancement (ADE). Additionally, antigenic similarities between Zika and dengue viruses can promote Zika virus infection by way of ADE
in vitro
using these very same anti-dengue antibodies.
Collapse
|
23
|
Paiva IA, Familiar-Macedo D, Badolato-Corrêa J, Carvalho FR, Dias HG, Pauvolid-Corrêa A, dos Santos CF, Silva AA, de Azeredo EL, Vianna RADO, Cardoso CAA, Grifoni A, Sette A, Weiskopf D, de-Oliveira-Pinto LM. Involvement of Th1Th17 Cell Subpopulations in the Immune Responses of Mothers Who Gave Birth to Children with Congenital Zika Syndrome (CZS). Viruses 2022; 14:v14020250. [PMID: 35215843 PMCID: PMC8879837 DOI: 10.3390/v14020250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
High levels of T helper 17 cell (Th17)-related cytokines have been shown in acute Zika virus (ZIKV) infection. We hypothesized that the high levels of Th17-related cytokines, associated with a regulatory environment during pregnancy, create a favorable milieu for the differentiation of CD4+Th17 cells. We present data from a cross-sectional study on mothers who confirmed ZIKV infection by qRT-PCR and their children. We also recruited non-pregnant women infected with ZIKV in the same period. ZIKV infection occurred between 2015 and 2017. We collected samples for this study between 2018 and 2019, years after the initial infection. We highlight that, after in vitro stimulation with ZIKV CD4 megapool (ZIKV MP), we found a lower frequency of IL-17-producing CD4+ T cells (Th17), especially in the mothers, confirmed by the decrease in IL-17 production in the supernatant. However, a higher frequency of CD4+ IL-17+ IFN-γ+ T cells (Th1Th17) responding to the ZIKV MP was observed in the cells of the mothers and children but not in those of the non-pregnant women. Our data indicate that the priming of CD4 T cells of the Th1Th17 phenotype occurred preferentially in the mothers who gave birth to children with CZS and in the children.
Collapse
Affiliation(s)
- Iury Amancio Paiva
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Débora Familiar-Macedo
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Jéssica Badolato-Corrêa
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Fabiana Rabe Carvalho
- Multiuser Laboratory for Research in Nephrology and Medical Science, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil; (F.R.C.); (A.A.S.); (C.A.A.C.)
| | - Helver Gonçalves Dias
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Alex Pauvolid-Corrêa
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA;
- Laboratory of Respiratory Viruses and Measles, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Caroline Fernandes dos Santos
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | - Andréa Alice Silva
- Multiuser Laboratory for Research in Nephrology and Medical Science, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil; (F.R.C.); (A.A.S.); (C.A.A.C.)
| | - Elzinandes Leal de Azeredo
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
| | | | - Claudete Aparecida Araújo Cardoso
- Multiuser Laboratory for Research in Nephrology and Medical Science, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil; (F.R.C.); (A.A.S.); (C.A.A.C.)
- Department of Maternal and Child, School of Medicine, Universidade Federal Fluminense, Niterói 24033-900, Brazil;
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), San Diego, CA 92037, USA; (A.G.); (A.S.); (D.W.)
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), San Diego, CA 92037, USA; (A.G.); (A.S.); (D.W.)
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), San Diego, CA 92037, USA; (A.G.); (A.S.); (D.W.)
| | - Luzia Maria de-Oliveira-Pinto
- Laboratory of Viral Immunology, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (I.A.P.); (D.F.-M.); (J.B.-C.); (H.G.D.); (C.F.d.S.); (E.L.d.A.)
- Correspondence:
| |
Collapse
|
24
|
Mapalagamage M, Weiskopf D, Sette A, De Silva AD. Current Understanding of the Role of T Cells in Chikungunya, Dengue and Zika Infections. Viruses 2022; 14:v14020242. [PMID: 35215836 PMCID: PMC8878350 DOI: 10.3390/v14020242] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 02/06/2023] Open
Abstract
Arboviral infections such as Chikungunya (CHIKV), Dengue (DENV) and Zika (ZIKV) are a major disease burden in tropical and sub-tropical countries, and there are no effective vaccinations or therapeutic drugs available at this time. Understanding the role of the T cell response is very important when designing effective vaccines. Currently, comprehensive identification of T cell epitopes during a DENV infection shows that CD8 and CD4 T cells and their specific phenotypes play protective and pathogenic roles. The protective role of CD8 T cells in DENV is carried out through the killing of infected cells and the production of proinflammatory cytokines, as CD4 T cells enhance B cell and CD8 T cell activities. A limited number of studies attempted to identify the involvement of T cells in CHIKV and ZIKV infection. The identification of human immunodominant ZIKV viral epitopes responsive to specific T cells is scarce, and none have been identified for CHIKV. In CHIKV infection, CD8 T cells are activated during the acute phase in the lymph nodes/blood, and CD4 T cells are activated during the chronic phase in the joints/muscles. Studies on the role of T cells in ZIKV-neuropathogenesis are limited and need to be explored. Many studies have shown the modulating actions of T cells due to cross-reactivity between DENV-ZIKV co-infections and have repeated heterologous/homologous DENV infection, which is an important factor to consider when developing an effective vaccine.
Collapse
Affiliation(s)
- Maheshi Mapalagamage
- Department of Zoology and Environment Sciences, Faculty of Science, University of Colombo, Colombo 00700, Sri Lanka;
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Aruna Dharshan De Silva
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (D.W.); (A.S.)
- Department of Paraclinical Sciences, Faculty of Medicine, General Sir John Kotelawala Defence University, Colombo 10390, Sri Lanka
- Correspondence:
| |
Collapse
|
25
|
Guerra-Gomes IC, Gois BM, Peixoto RF, Palmeira PHDS, Dias CNDS, Csordas BG, Araújo JMG, Veras RC, de Medeiros IA, de Azevedo FDLAA, Boyton RJ, Altmann DM, Keesen TSL. Phenotypical characterization of regulatory T cells in acute Zika infection. Cytokine 2021; 146:155651. [PMID: 34325119 PMCID: PMC8405058 DOI: 10.1016/j.cyto.2021.155651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
Zika virus (ZIKV), alongside Dengue virus (DENV), Chikungunya virus (CHIKV), and Yellow Fever Virus (YFV) are prevalent arboviruses in the Americas. Each of these infections is associated with the development of associated disease immunopathology. Immunopathological processes are an outcome of counter-balancing impacts between effector and regulatory immune mechanisms. In this context, regulatory T cells (Tregs) are key in modulating the immune response and, therefore, in tissue damage control. However, to date, Treg phenotypes and mechanisms during acute infection of the ZIKV in humans have not been fully investigated. The main aim of this work was to characterize Tregs and their immunological profile related to cytokine production and molecules that are capable of controlling the exacerbated inflammatory profile in acute Zika infected patients. Using whole blood analyses of infected patients, an ex vivo phenotypical characterization of Tregs, circulating during acute Zika virus infection, was conducted by flow cytometry. We found that though there are no differences in absolute Treg frequency between infected and healthy control groups. However, pro-inflammatory cytokine up-regulation such as IFN-γ and LAP was observed in the acute disease. Furthermore, acute ZIKV patients expressed increased levels of CD39/CD73, perforin/granzyme B, PD-1, and CTLA-4, all markers involved in mechanisms used by Tregs to attempt to control strong inflammatory responses. Thus, the data indicates a potential contribution of Tregs during the inflammatory ZIKV infection response.
Collapse
Affiliation(s)
- Isabel Cristina Guerra-Gomes
- Immunology of Infectious Diseases Laboratory of Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Bruna Macêdo Gois
- Immunology of Infectious Diseases Laboratory of Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Rephany Fonseca Peixoto
- Immunology of Infectious Diseases Laboratory of Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Pedro Henrique de Sousa Palmeira
- Immunology of Infectious Diseases Laboratory of Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Cínthia Nóbrega de Sousa Dias
- Immunology of Infectious Diseases Laboratory of Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Bárbara Guimarães Csordas
- Immunology of Infectious Diseases Laboratory of Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Josélio Maria Galvão Araújo
- Molecular Biology of Cancer and Infectious Diseases Laboratory of Post-Graduation Program on Parasite Biology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte 58078-970, Brazil
| | - Robson Cavalcante Veras
- Research Institute for Drugs and Medicines, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Isac Almeida de Medeiros
- Research Institute for Drugs and Medicines, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | | | - Rosemary Jane Boyton
- Department of Infectious Disease, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Daniel Martin Altmann
- Department of Immunology and Inflammation, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Tatjana Souza Lima Keesen
- Immunology of Infectious Diseases Laboratory of Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil.
| |
Collapse
|
26
|
Shmeleva EV, Colucci F. Maternal natural killer cells at the intersection between reproduction and mucosal immunity. Mucosal Immunol 2021; 14:991-1005. [PMID: 33903735 PMCID: PMC8071844 DOI: 10.1038/s41385-020-00374-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/24/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Many maternal immune cells populate the decidua, which is the mucosal lining of the uterus transformed during pregnancy. Here, abundant natural killer (NK) cells and macrophages help the uterine vasculature adapt to fetal demands for gas and nutrients, thereby supporting fetal growth. Fetal trophoblast cells budding off the forming placenta and invading deep into maternal tissues come into contact with these and other immune cells. Besides their homeostatic functions, decidual NK cells can respond to pathogens during infection, but in doing so, they may become conflicted between destroying the invader and sustaining fetoplacental growth. We review how maternal NK cells balance their double duty both in the local microenvironment of the uterus and systemically, during toxoplasmosis, influenza, cytomegalovirus, malaria and other infections that threat pregnancy. We also discuss recent developments in the understanding of NK-cell responses to SARS-Cov-2 infection and the possible dangers of COVID-19 during pregnancy.
Collapse
Affiliation(s)
- Evgeniya V Shmeleva
- Department of Obstetrics & Gynaecology, University of Cambridge, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Francesco Colucci
- Department of Obstetrics & Gynaecology, University of Cambridge, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Filgueiras IS, Torrentes de Carvalho A, Cunha DP, Mathias da Fonseca DL, El Khawanky N, Freire PP, Cabral-Miranda G, Schimke LF, Camara NOS, Ochs HD, Peron JPS, Cabral-Marques O, de Vasconcelos ZFM. The clinical spectrum and immunopathological mechanisms underlying ZIKV-induced neurological manifestations. PLoS Negl Trop Dis 2021; 15:e0009575. [PMID: 34351896 PMCID: PMC8341629 DOI: 10.1371/journal.pntd.0009575] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Since the 2015 to 2016 outbreak in America, Zika virus (ZIKV) infected almost 900,000 patients. This international public health emergency was mainly associated with a significant increase in the number of newborns with congenital microcephaly and abnormal neurologic development, known as congenital Zika syndrome (CZS). Furthermore, Guillain-Barré syndrome (GBS), a neuroimmune disorder of adults, has also been associated with ZIKV infection. Currently, the number of ZIKV-infected patients has decreased, and most of the cases recently reported present as a mild and self-limiting febrile illness. However, based on its natural history of a typical example of reemerging pathogen and the lack of specific therapeutic options against ZIKV infection, new outbreaks can occur worldwide, demanding the attention of researchers and government authorities. Here, we discuss the clinical spectrum and immunopathological mechanisms underlying ZIKV-induced neurological manifestations. Several studies have confirmed the tropism of ZIKV for neural progenitor stem cells by demonstrating the presence of ZIKV in the central nervous system (CNS) during fetal development, eliciting a deleterious inflammatory response that compromises neurogenesis and brain formation. Of note, while the neuropathology of CZS can be due to a direct viral neuropathic effect, adults may develop neuroimmune manifestations such as GBS due to poorly understood mechanisms. Antiganglioside autoantibodies have been detected in multiple patients with ZIKV infection-associated GBS, suggesting a molecular mimicry. However, further additional immunopathological mechanisms remain to be uncovered, paving the way for new therapeutic strategies.
Collapse
Affiliation(s)
- Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences of University of São Paulo, São Paulo, Brazil
| | - Amanda Torrentes de Carvalho
- Department of Immunobiology, Institute of Biology of Federal University of Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Daniela Prado Cunha
- Department of Clinical Research, Instituto Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | | | - Nadia El Khawanky
- Department of Hematology and Oncology, Faculty of Medicine, the University of Freiburg, Freiburg, Germany
| | - Paula Paccielli Freire
- Department of Immunology, Institute of Biomedical Sciences of University of São Paulo, São Paulo, Brazil
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences of University of São Paulo, São Paulo, Brazil
| | - Lena F. Schimke
- Department of Immunology, Institute of Biomedical Sciences of University of São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Camara
- Department of Immunology, Institute of Biomedical Sciences of University of São Paulo, São Paulo, Brazil
| | - Hans D. Ochs
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | | | - Otávio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences of University of São Paulo, São Paulo, Brazil
- Department of Clinical Analyses and Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, Brazil
| | | |
Collapse
|
28
|
Teng Y, Xie W, Tao X, Liu N, Yu Y, Huang Y, Xu D, Fan Y. Infection-provoked psoriasis: Induced or aggravated (Review). Exp Ther Med 2021; 21:567. [PMID: 33850539 PMCID: PMC8027725 DOI: 10.3892/etm.2021.9999] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Psoriasis is a common chronic, immune-mediated, inflammatory skin disorder, with a reported prevalence of 0.0-2.1% among children and 0.91-8.50% among adults, worldwide. Psoriasis is induced by several environmental factors, including infection, alcohol consumption, drugs, trauma, acute withdrawal of systemic or potent topical corticosteroids, body mass index and endocrine disorders. Increasing evidence suggest that a variety of microorganisms play key roles in the induction and exacerbation of psoriasis. Pathogens, such as streptococci and staphylococci are considered causal factors, presumably via superantigen activation of skin-seeking T cells. In addition, fungal pathogens, such as Candida and Malassezia, and viral agents, such as human immunodeficiency virus, hepatitis C virus infection and human papillomavirus, are also closely associated with psoriasis. Recently, several types of pathogens, such as Helicobacter pylori infection, Zika virus and scabies, have been reported to potentially trigger psoriasis. The present review discusses the underlying molecular mechanisms by which these infections influence psoriasis to provide a better understanding of the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Yan Teng
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Wenqing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaohua Tao
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Na Liu
- Department of Dermatology, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yong Yu
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Youming Huang
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Danfeng Xu
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yibin Fan
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
29
|
Paiva IA, Badolato-Corrêa J, Familiar-Macedo D, de-Oliveira-Pinto LM. Th17 Cells in Viral Infections-Friend or Foe? Cells 2021; 10:cells10051159. [PMID: 34064728 PMCID: PMC8151546 DOI: 10.3390/cells10051159] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Th17 cells are recognized as indispensable in inducing protective immunity against bacteria and fungi, as they promote the integrity of mucosal epithelial barriers. It is believed that Th17 cells also play a central role in the induction of autoimmune diseases. Recent advances have evaluated Th17 effector functions during viral infections, including their critical role in the production and induction of pro-inflammatory cytokines and in the recruitment and activation of other immune cells. Thus, Th17 is involved in the induction both of pathogenicity and immunoprotective mechanisms seen in the host's immune response against viruses. However, certain Th17 cells can also modulate immune responses, since they can secrete immunosuppressive factors, such as IL-10; these cells are called non-pathogenic Th17 cells. Here, we present a brief review of Th17 cells and highlight their involvement in some virus infections. We cover these notions by highlighting the role of Th17 cells in regulating the protective and pathogenic immune response in the context of viral infections. In addition, we will be describing myocarditis and multiple sclerosis as examples of immune diseases triggered by viral infections, in which we will discuss further the roles of Th17 cells in the induction of tissue damage.
Collapse
|
30
|
Nascimento-Carvalho GC, Nascimento-Carvalho EC, Ramos CL, Vilas-Boas AL, Moreno-Carvalho OA, Vinhaes CL, Barreto-Duarte B, Queiroz ATL, Andrade BB, Nascimento-Carvalho CM. Zika-exposed microcephalic neonates exhibit higher degree of inflammatory imbalance in cerebrospinal fluid. Sci Rep 2021; 11:8474. [PMID: 33875756 PMCID: PMC8055905 DOI: 10.1038/s41598-021-87895-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 04/05/2021] [Indexed: 11/09/2022] Open
Abstract
Not every neonate with congenital Zika virus (ZIKV) infection (CZI) is born with microcephaly. We compared inflammation mediators in CSF (cerebrospinal fluid obtained from lumbar puncture) between ZIKV-exposed neonates with/without microcephaly (cases) and controls. In Brazil, in the same laboratory, we identified 14 ZIKV-exposed neonates during the ZIKV epidemic (2015-2016), 7(50%) with and 7(50%) without microcephaly, without any other congenital infection, and 14 neonates (2017-2018) eligible to be controls and to match cases. 29 inflammation mediators were measured using Luminex immunoassay and multidimensional analyses were employed. Neonates with ZIKV-associated microcephaly presented substantially higher degree of inflammatory perturbation, associated with uncoupled inflammatory response and decreased correlations between concentrations of inflammatory biomarkers. The groups of microcephalic and non-microcephalic ZIKV-exposed neonates were distinguished from the control group (area under curve [AUC] = 1; P < 0.0001). Between controls and those non-microcephalic exposed to ZIKV, IL-1β, IL-3, IL-4, IL-7 and EOTAXIN were the top CSF markers. By comparing the microcephalic cases with controls, the top discriminant scores were for IL-1β, IL-3, EOTAXIN and IL-12p70. The degree of inflammatory imbalance may be associated with microcephaly in CZI and it may aid additional investigations in experimental pre-clinical models testing immune modulators in preventing extensive damage of the Central Nervous System.
Collapse
Affiliation(s)
| | | | - Clara L Ramos
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
| | - Ana-Luisa Vilas-Boas
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
| | | | - Caian L Vinhaes
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
| | - Beatriz Barreto-Duarte
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
- University Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, 41820-021, Brazil
| | - Artur T L Queiroz
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
| | - Bruno B Andrade
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
- University Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, 41820-021, Brazil
- School of Medicine, Faculdade de Tecnologia e Ciências (Uni-FTC), Salvador, Bahia, 41741-590, Brazil
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7700, South Africa
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | | |
Collapse
|
31
|
Mittal S, Federman HG, Sievert D, Gleeson JG. The Neurobiology of Modern Viral Scourges: ZIKV and COVID-19. Neuroscientist 2021; 28:438-452. [PMID: 33874789 DOI: 10.1177/10738584211009149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The interactions of viruses with the nervous system were thought to be well understood until the recent outbreaks of Zika and SARS-CoV-2. In this review, we consider these emerging pathogens, the range and mechanisms of the neurological disease in humans, and how the biomedical research enterprise has pivoted to answer questions about viral pathogenesis, immune response, and the special vulnerability of the nervous system. ZIKV stands out as the only new virus in a generation, associating with congenital brain defects, neurological manifestations of microcephaly in newborns, and radiculopathy in adults. COVID-19, the disease caused by SARS-CoV-2, has swept the planet in an unprecedented manner and is feared worldwide for its effect on the respiratory system, but recent evidence points to important neurological sequelae. These can include anosmia, vasculopathy, paresthesias, and stroke. Evidence of ZIKV and SARS-CoV-2 genetic material from neural tissue, and evidence of infection of neural cells, raises questions about how these emerging viruses produce disease, and where new therapies might emerge.
Collapse
Affiliation(s)
- Swapnil Mittal
- Department of Neurosciences and Pediatrics, University of California San Diego, La Jolla, CA, USA.,Rady Children's Institute of Genomic Medicine, San Diego, CA, USA
| | - Hannah G Federman
- Department of Neurosciences and Pediatrics, University of California San Diego, La Jolla, CA, USA.,Rady Children's Institute of Genomic Medicine, San Diego, CA, USA.,Department of Medicine, New Jersey Medical School, Center for Immunity and Inflammation, Rutgers University, Newark, NJ, USA
| | - David Sievert
- Department of Neurosciences and Pediatrics, University of California San Diego, La Jolla, CA, USA.,Rady Children's Institute of Genomic Medicine, San Diego, CA, USA
| | - Joseph G Gleeson
- Department of Neurosciences and Pediatrics, University of California San Diego, La Jolla, CA, USA.,Rady Children's Institute of Genomic Medicine, San Diego, CA, USA
| |
Collapse
|
32
|
Sonon P, Collares CVA, Ferreira MLB, Almeida RS, Sadissou I, Cordeiro MT, de Fátima Militão de Albuquerque M, Castelli EC, Lucena-Silva N, Donadi EA. Peripheral spectrum neurological disorder after arbovirus infection is associated with HLA-F variants among Northeastern Brazilians. INFECTION GENETICS AND EVOLUTION 2021; 92:104855. [PMID: 33839310 DOI: 10.1016/j.meegid.2021.104855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Non-classical class I human leukocyte antigens (HLA) molecules are known to modulate the function of cytotoxic cells (NK and T CD8+) during viral infection by interacting with inhibitory/activating receptors. However, little is known about the HLA-E/-F genetic variability on arbovirus infections. METHODS We evaluated by massive parallel sequencing the full HLA-E/-F genetic diversity among patients infected during the arbovirus (ZIKV, DENV, and CHIKV) outbreak leading to a broad range of neurological complications in the Brazilian State of Pernambuco. In parallel, healthy blood donors from the same area were also studied. Plink and R software were used for genetic association study. To limit the false-positive results and enhance the reliability of the results, we adopted P-values <0.01 as significant levels. RESULTS Compared to controls, the HLA-F alleles: -1610 C (rs17875375), +1383 G (rs17178385), and +3537 A (rs17875384), all in complete linkage disequilibrium with each other (r2 = 1), were overrepresented in patients presenting peripheral spectrum disorders (PSD). The HLA-F*Distal-D haplotype that harbored the -1610 C allele exhibited a trend increase in PSD group. No associations were found for HLA-E. CONCLUSIONS Our findings showed that the HLA-F genetic background seems to be more important than HLA-E on the susceptibility to PSD complications.
Collapse
Affiliation(s)
- Paulin Sonon
- Immunogenetic Laboratory, Immunology Department, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Av. Moraes rego, s/n, Campus da UFPE, Cidade Universitária, 50670420 Recife, PE, Brazil; Ribeirão Preto Medical School, University of São Paulo, AV Bandeirantes, 3900, HC, Vila Monte Alegre, 14049900 Ribeirão Preto, SP, Brazil
| | - Cristhianna V A Collares
- Ribeirão Preto Medical School, University of São Paulo, AV Bandeirantes, 3900, HC, Vila Monte Alegre, 14049900 Ribeirão Preto, SP, Brazil
| | - Maria Lúcia Brito Ferreira
- Hospital da Restauração Gov. Paulo Guerra, Av. Gov. Agamenon Magalhães, s/n, Derby, 52171011 Recife, PE, Brazil
| | - Renata Santos Almeida
- Immunogenetic Laboratory, Immunology Department, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Av. Moraes rego, s/n, Campus da UFPE, Cidade Universitária, 50670420 Recife, PE, Brazil
| | - Ibrahim Sadissou
- Ribeirão Preto Medical School, University of São Paulo, AV Bandeirantes, 3900, HC, Vila Monte Alegre, 14049900 Ribeirão Preto, SP, Brazil
| | - Marli Tenório Cordeiro
- Virology Department, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Av. Moraes rego, s/n, Campus da UFPE, Cidade Universitária, 50670420 Recife, PE, Brazil
| | - Maria de Fátima Militão de Albuquerque
- Public Health Department, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Av. Moraes rego, s/n, Campus da UFPE, Cidade Universitária, 50670420 Recife, PE, Brazil
| | - Erick C Castelli
- São Paulo State University (UNESP), School of Medicine, Molecular Genetics and Bioinformatics Laboratory, Prof. Dr. Walter Maurício Correa, s/n Unesp, Campus de Botucatu, Botucatu CEP 18618681, SP, Brazil; São Paulo State University (UNESP), Department of Pathology, School of Medicine, Botucatu, SP, Brazil
| | - Norma Lucena-Silva
- Immunogenetic Laboratory, Immunology Department, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Av. Moraes rego, s/n, Campus da UFPE, Cidade Universitária, 50670420 Recife, PE, Brazil
| | - Eduardo A Donadi
- Ribeirão Preto Medical School, University of São Paulo, AV Bandeirantes, 3900, HC, Vila Monte Alegre, 14049900 Ribeirão Preto, SP, Brazil.
| |
Collapse
|
33
|
Silva AJD, Jesus ALS, Leal LRS, Silva GAS, Melo CML, Freitas AC. Pichia pastoris displaying ZIKV protein epitopes from the Envelope and NS1 induce in vitro immune activation. Vaccine 2021; 39:2545-2554. [PMID: 33814233 DOI: 10.1016/j.vaccine.2021.03.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/26/2021] [Accepted: 03/19/2021] [Indexed: 02/08/2023]
Abstract
The severe consequences of ZIKV infection and its emergence and re-emergence in several countries have boosted vaccines' development. Yeasts such as Pichia pastoris has been widely employed as antigen carriers for immunization against infectious agents. Components of the yeast cell wall have immunostimulatory properties, and recombinant antigens can be anchored to the cell surface to enhance the presentation to the immune system. Here we aimed at producing and anchoring ZIKV proteins in the P. pastoris surface as a vaccine approach. Expression cassettes were designed with epitopes of the Envelope and NS1 proteins. Immunofluorescence microscopy confirmed the anchoring of recombinant proteins. Yeasts' ability to stimulate immune cells was evaluated in vitro by incubation with lymphocytes and monocytes isolated from mouse spleen. P. pastoris expressing EnvNS1 epitopes promoted increased levels of IL-6, IL-10, and TNF-α cytokines and an increase in the number of CD4+, CD8+, and CD16+ lymphocytes, similarly to ZIKV. This profile is indicative of the activation of immunological cells and suggests an immunogenic potential of the proposed yeast vaccines against ZIKV, reinforcing the possibility of P. pastoris as adjuvant and carrier of antigens.
Collapse
Affiliation(s)
- Anna Jéssica D Silva
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - André Luiz S Jesus
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Lígia Rosa S Leal
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Guilherme Antonio S Silva
- Laboratório de Análises Imunológicas e Antitumorais, Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, Brazil
| | - Cristiane Moutinho L Melo
- Laboratório de Análises Imunológicas e Antitumorais, Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, Brazil
| | - Antonio C Freitas
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
34
|
Almeida RS, Ferreira MLB, Sonon P, Cordeiro MT, Sadissou I, Diniz GTN, Militão-Albuquerque MDFP, Franca RFDO, Donadi EA, Lucena-Silva N. Cytokines and Soluble HLA-G Levels in the Acute and Recovery Phases of Arbovirus-Infected Brazilian Patients Exhibiting Neurological Complications. Front Immunol 2021; 12:582935. [PMID: 33776990 PMCID: PMC7994272 DOI: 10.3389/fimmu.2021.582935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/08/2021] [Indexed: 12/19/2022] Open
Abstract
Severe neurological complications following arbovirus infections have been a major concern in seasonal outbreaks, as reported in the Northeast region of Brazil, where the same mosquito transmitted Zika (ZIKV), Dengue (DENV), and Chikungunya (CHIKV) viruses. In this study, we evaluated the levels of 36 soluble markers, including cytokines, chemokines, growth factors, and soluble HLA-G (Luminex and ELISA) in: i) serum and cerebrospinal fluid (CSF), during the acute phase and two years after the infection (recovery phase, only serum), ii) the relationship among all soluble molecules in serum and CSF, and iii) serum of infected patients without neurological complications, during the acute infection. Ten markers (sHLA-G, IL-10, IL-22, IL-8, MIP-1α, MIP-1β, MCP-1, HGF, VEGF, and IL-1RA) exhibited differential levels between the acute and recovery phases, with pronounced increases in MIP-1α (P<0.0001), MCP-1 (P<0.0001), HGF (P= 0.0001), and VEGF (P<0.0001) in the acute phase. Fourteen molecules (IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-9, IL-13, IL-15, IL-17A, IFN-α, TNF, and G-CSF) exhibited distinct levels between arbovirus patients presenting or not neurological complications. IL-8, EGF, IL-6, and MCP-1 levels were increased in CSF, while RANTES and Eotaxin levels were higher in serum. Soluble serum (IL-22, RANTES, Eotaxin) and CSF (IL-8, EGF, IL-3) mediators may discriminate putative risks for neurological complications following arbovirus infections. Neurological complications were associated with the presence of a predominant inflammatory profile, whereas in non-complicated patients an anti-inflammatory profile may predominate. Mediators associated with neuroregeneration (EGF and IL-3) may be induced in response to neurological damage. Broad spectrum immune checkpoint molecules (sHLA-G) interact with cytokines, chemokines, and growth factors. The identification of soluble markers may be useful to monitor neurological complications and may aid in the development of novel therapies against neuroinflammation.
Collapse
Affiliation(s)
- Renata Santos Almeida
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | - Paulin Sonon
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil.,Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marli Tenório Cordeiro
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Ibrahim Sadissou
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - George Tadeu Nunes Diniz
- Department of Collective Health, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | | | | | - Norma Lucena-Silva
- Laboratory of Immunogenetics, Department of Immunology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| |
Collapse
|
35
|
Hansen M, Nolan MS, Gorchakov R, Hasbun R, Murray KO, Ronca SE. Unique Cytokine Response in West Nile Virus Patients Who Developed Chronic Kidney Disease: A Prospective Cohort Study. Viruses 2021; 13:v13020311. [PMID: 33671257 PMCID: PMC7922428 DOI: 10.3390/v13020311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/21/2022] Open
Abstract
West Nile virus (WNV) is a widespread and devastating disease, especially in those who develop neuroinvasive disease. A growing body of evidence describes sequelae years after infection, including neurological complications and chronic kidney disease (CKD). Eighty-nine out of 373 WNV-positive cases were followed for approximately two years and compared to 127 WNV-negative controls with and without CKD. Adjusted risk ratios (aRRs) were calculated via a log binomial regression to determine the impact of WNV exposure and other possible confounders on the likelihood of developing CKD. Cytokine profiles of WNV patients and controls were evaluated to characterize differences and describe potential underlying pathophysiological mechanisms. The associated risk for developing CKD was significantly associated with history of WNV infection (aRR = 1.91, 95% CI 1.13–3.25). Additionally, five distinct cytokines were found to be significantly associated with WNV infection (eotaxin, IL-8, IL-12p70, IP-10, and TNFα) after the p-value was adjusted to <0.0019 due to the Bonferroni correction. These data support that WNV infection is an independent risk factor for CKD, even after accounting for confounding comorbidities. WNV participants who developed CKD had high activity of proinflammatory markers, indicating underlying inflammatory disease. This study provides new insights into CKD resultant of WNV infection.
Collapse
Affiliation(s)
- Michael Hansen
- Department of Family and Community Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Melissa S. Nolan
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, Columbia, SC 29208, USA;
| | - Rodion Gorchakov
- Department of Pediatrics, Section Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (R.G.); (K.O.M.)
| | - Rodrigo Hasbun
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Kristy O. Murray
- Department of Pediatrics, Section Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (R.G.); (K.O.M.)
| | - Shannon E. Ronca
- Department of Pediatrics, Section Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (R.G.); (K.O.M.)
- Correspondence:
| |
Collapse
|
36
|
Vinhaes CL, Arriaga MB, de Almeida BL, Oliveira JV, Santos CS, Calcagno JI, Carvalho TX, Giovanetti M, Alcantara LCJ, de Siqueira IC, Andrade BB. Newborns With Zika Virus-Associated Microcephaly Exhibit Marked Systemic Inflammatory Imbalance. J Infect Dis 2021; 222:670-680. [PMID: 32311029 DOI: 10.1093/infdis/jiaa197] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/17/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Zika virus (ZIKV) is an emergent flavivirus initially considered a benign and self-limited exanthematic illness. In 2015, a new epidemic emerged in northeastern of Brazil with increased incidence of a previously rare clinical outcome, microcephaly, in newborns from mothers who were infected during pregnancy. Little is known about the immunopathogenesis of ZIKV-associated microcephaly. Understanding the inflammatory profile and degree of inflammation of persons affected with such condition is an important step towards development of innovative therapeutic strategies. METHODS A case-control study compared plasma levels of several inflammatory biomarkers from newborns with ZIKV microcephaly, asymptomatic ZKV infection, or uninfected controls. Plasma biomarkers were assessed using Luminex. A series of multidimensional analysis was performed to characterize the systemic immune activation profile of the clinical groups. RESULTS We identified an inflammatory signature associated with ZIKV microcephaly that suggested an increased inflammation. Network analysis suggested that ZIKV microcephaly is associated with imbalanced immune activation and inflammation. The cephalic perimeter was inversely proportional with the degree of inflammatory perturbation. Furthermore, a combination of plasma inflammatory biomarkers could discriminate ZIKV with microcephaly from those with ZIKV without microcephaly or uninfected neonates. CONCLUSIONS An intense inflammatory imbalance that is proportional to the disease severity hallmarks ZIKV microcephaly.
Collapse
Affiliation(s)
- Caian L Vinhaes
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Curso de Medicina, Faculdade de Tecnologia e Ciências (UniFTC), Salvador, Brazil
| | - María B Arriaga
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil
| | | | - João V Oliveira
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Cleiton S Santos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Juan I Calcagno
- Maternidade de Referência Prof. José Maria de Magalhães Netto-Secretaria de Saúde do Estado da Bahia (SESAB), Salvador, Brazil
| | - Tereza X Carvalho
- Maternidade de Referência Prof. José Maria de Magalhães Netto-Secretaria de Saúde do Estado da Bahia (SESAB), Salvador, Brazil
| | - Marta Giovanetti
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Carlos J Alcantara
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Bruno B Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Curso de Medicina, Faculdade de Tecnologia e Ciências (UniFTC), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil.,Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil.,Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| |
Collapse
|
37
|
Zhang X, Li G, Chen G, Zhu N, Wu D, Wu Y, James TD. Recent progresses and remaining challenges for the detection of Zika virus. Med Res Rev 2021; 41:2039-2108. [PMID: 33559917 DOI: 10.1002/med.21786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/26/2022]
Abstract
Zika virus (ZIKV) has emerged as a particularly notorious mosquito-borne flavivirus, which can lead to a devastating congenital syndrome in the fetuses of pregnant mothers (e.g., microcephaly, spasticity, craniofacial disproportion, miscarriage, and ocular abnormalities) and cause the autoimmune disorder Guillain-Barre' syndrome of adults. Due to its severity and rapid dispersal over several continents, ZIKV has been acknowledged to be a global health concern by the World Health Organization. Unfortunately, the ZIKV has recently resurged in India with the potential for devastating effects. Researchers from all around the world have worked tirelessly to develop effective detection strategies and vaccines for the prevention and control of ZIKV infection. In this review, we comprehensively summarize the most recent research into ZIKV, including the structural biology and evolution, historical overview, pathogenesis, symptoms, and transmission. We then focus on the detection strategies for ZIKV, including viral isolation, serological assays, molecular assays, sensing methods, reverse transcription loop mediated isothermal amplification, transcription-mediated amplification technology, reverse transcription strand invasion based amplification, bioplasmonic paper-based device, and reverse transcription isothermal recombinase polymerase amplification. To conclude, we examine the limitations of currently available strategies for the detection of ZIKV, and outline future opportunities and research challenges.
Collapse
Affiliation(s)
- Xianlong Zhang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guoliang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guang Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Niu Zhu
- Department of Public Health, Xi'an Medical University, Xi'an, China
| | - Di Wu
- Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment, Food Safety Research Unit (2019RU014) of Chinese Academy of Medical Science, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, UK.,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| |
Collapse
|
38
|
Chang Y, Jiang Y, Li C, Wang Q, Zhang F, Qin CF, Wu QF, Li J, Xu Z. Different Gene Networks Are Disturbed by Zika Virus Infection in A Mouse Microcephaly Model. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 18:737-748. [PMID: 33418086 PMCID: PMC8377042 DOI: 10.1016/j.gpb.2019.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/17/2019] [Accepted: 08/02/2019] [Indexed: 12/27/2022]
Abstract
The association of Zika virus (ZIKV) infection with microcephaly has raised alarm worldwide. Their causal link has been confirmed in different animal models infected by ZIKV. However, the molecular mechanisms underlying ZIKV pathogenesis are far from clear. Hence, we performed global gene expression analysis of ZIKV-infected mouse brains to unveil the biological and molecular networks underpinning microcephaly. We found significant dysregulation of the sub-networks associated with brain development, immune response, cell death, microglial cell activation, and autophagy amongst others. We provided detailed analysis of the related complicated gene networks and the links between them. Additionally, we analyzed the signaling pathways that were likely to be involved. This report provides systemic insights into not only the pathogenesis, but also a path to the development of prophylactic and therapeutic strategies against ZIKV infection.
Collapse
Affiliation(s)
- Yafei Chang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yisheng Jiang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Cui Li
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qin Wang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Zhang
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
39
|
Spencer Clinton JL, Tran LL, Vogt MB, Rowley DR, Kimata JT, Rico-Hesse R. IP-10 and CXCR3 signaling inhibit Zika virus replication in human prostate cells. PLoS One 2020; 15:e0244587. [PMID: 33378361 PMCID: PMC7773246 DOI: 10.1371/journal.pone.0244587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/12/2020] [Indexed: 11/18/2022] Open
Abstract
Our previous studies have shown that Zika virus (ZIKV) replicates in human prostate cells, suggesting that the prostate may serve as a long-term reservoir for virus transmission. Here, we demonstrated that the innate immune responses generated to three distinct ZIKV strains (all isolated from human serum) were significantly different and dependent on their passage history (in mosquito, monkey, or human cells). In addition, some of these phenotypic differences were reduced by a single additional cell culture passage, suggesting that viruses that have been passaged more than 3 times from the patient sample will no longer reflect natural phenotypes. Two of the ZIKV strains analyzed induced high levels of the IP-10 chemokine and IFNγ in human prostate epithelial and stromal mesenchymal stem cells. To further understand the importance of these innate responses on ZIKV replication, we measured the effects of IP-10 and its downstream receptor, CXCR3, on RNA and virus production in prostate cells. Treatment with IP-10, CXCR3 agonist, or CXCR3 antagonist significantly altered ZIKV viral gene expression, depending on their passage in cells of relevant hosts (mosquito or human). We detected differences in gene expression of two primary CXCR3 isoforms (CXCR3-A and CXCR3-B) on the two cell types, possibly explaining differences in viral output. Lastly, we examined the effects of IP-10, agonist, or antagonist on cell death and proliferation under physiologically relevant infection rates, and detected no significant differences. Although we did not measure protein expression directly, our results indicate that CXCR3 signaling may be a target for therapeutics, to ultimately stop sexual transmission of this virus.
Collapse
Affiliation(s)
- Jennifer L. Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Linda L. Tran
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Megan B. Vogt
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - David R. Rowley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rebecca Rico-Hesse
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
40
|
Guzeloglu-Kayisli O, Guo X, Tang Z, Semerci N, Ozmen A, Larsen K, Mutluay D, Guller S, Schatz F, Kayisli UA, Lockwood CJ. Zika Virus-Infected Decidual Cells Elicit a Gestational Age-Dependent Innate Immune Response and Exaggerate Trophoblast Zika Permissiveness: Implication for Vertical Transmission. THE JOURNAL OF IMMUNOLOGY 2020; 205:3083-3094. [PMID: 33139490 DOI: 10.4049/jimmunol.2000713] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Vertical transmission of the Zika virus (ZIKV) causes severe fetal defects, but the exact pathogenic mechanism is unclear. We identified up to a 10,480-fold higher expression of viral attachment factors AXL, GAS6, and PROS1 and a 3880-fold increase in ZIKV infectiousness/propagation in human term decidual stromal cells versus trophoblasts. Moreover, levels of viral attachment factors and ZIKV are significantly increased, whereas expression of innate immune response genes are significantly decreased, in human first trimester versus term decidual cells. ZIKV-infected decidual cell supernatants increased cytotrophoblasts infection up to 252-fold compared with directly infected cytotrophoblasts. Tizoxanide treatment efficiently inhibited Zika infection in both maternal and fetal cells. We conclude that ZIKV permissiveness, as well as innate immune responsiveness of human decidual cells, are gestational age dependent, and decidual cells augment ZIKV infection of primary human cytotrophoblast cultures, which are otherwise ZIKV resistant. Human decidual cells may act as reservoirs for trimester-dependent placental transmission of ZIKV, accounting for the higher Zika infection susceptibility and more severe fetal sequelae observed in early versus late pregnancy. Moreover, tizoxanide is a promising agent in preventing perinatal Zika transmission as well as other RNA viruses such as coronavirus.
Collapse
Affiliation(s)
- Ozlem Guzeloglu-Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Xiaofang Guo
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Zhonghua Tang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Asli Ozmen
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Kellie Larsen
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Duygu Mutluay
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06510
| | - Frederick Schatz
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Umit Ali Kayisli
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| | - Charles Joseph Lockwood
- Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612; and
| |
Collapse
|
41
|
Rabelo K, de Souza LJ, Salomão NG, Machado LN, Pereira PG, Portari EA, Basílio-de-Oliveira R, Dos Santos FB, Neves LD, Morgade LF, Provance DW, Higa LM, Tanuri A, de Carvalho JJ, Paes MV. Zika Induces Human Placental Damage and Inflammation. Front Immunol 2020; 11:2146. [PMID: 32983175 PMCID: PMC7490298 DOI: 10.3389/fimmu.2020.02146] [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] [Received: 05/21/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022] Open
Abstract
In Brazil, an epidemic of Zika virus (ZIKV) infections was declared in 2015 that coincided with alarming reports of microcephaly in newborns associated with mother infection. Although the virus has placental tropism, changes in the tissue morphology and immunity of infected patients have not yet been elucidated. Here, we investigated the histopathological and ultrastructural changes along with the immunological profile and the BDNF expression in rare placental material. Tissues were obtained in the 2015–2016 Brazilian epidemic, of ten ZIKV-infected patients during pregnancy, five resulting in cases of fetal microcephaly and five non-microcephaly, compared to five non-infected control placentae. Viral antigens were only detected in samples from the ZIKV infected patients. Infected placentae presented histopathological severe damage, while the ultrastructural evaluation showed abnormal organelles, such as clusters of virus-like particles consistent with the ZIKV dimensions. Increased infiltration of CD68+ and TCD8+ cells, expression of MMPs, cytokines (IFN-γ and TNF-α) and other immunological mediators (RANTES/CCL5 and VEGFR-2) confirmed excessive inflammation and vascular permeability dysfunction. An evaluation of BDNF showed a decrease that could modulate neuronal damage in the developing fetus. The placental changes caused by ZIKV are not pathognomonic, however, the data provide evidence that this infection leads to severe placental injury.
Collapse
Affiliation(s)
- Kíssila Rabelo
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Natália Gedeão Salomão
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | | | - Priscila Gomes Pereira
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Laura Dias Neves
- Hospital Geral Dr. Beda, CEPLIN - Uti Neonatal Nicola Albano, Rio de Janeiro, Brazil
| | - Luciana Faes Morgade
- Hospital Geral Dr. Beda, CEPLIN - Uti Neonatal Nicola Albano, Rio de Janeiro, Brazil
| | - David William Provance
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.,Centro de Desenvolvimento Tecnológico em Saúde, Fiocruz, Rio de Janeiro, Brazil
| | - Luiza Mendonça Higa
- Laboratório de Virologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amilcar Tanuri
- Laboratório de Virologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge José de Carvalho
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marciano Viana Paes
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Alkaff AH, Yohan B, Tambunan USF, Sasmono RT. Zika, chikungunya, and dengue viral infections in human peripheral blood mononuclear cells: cell susceptibility and gene expression. MEDICAL JOURNAL OF INDONESIA 2020. [DOI: 10.13181/mji.oa.193548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Infections of Zika (ZIKV), dengue (DENV), and chikungunya viruses (CHIKV) are presented with similar clinical symptoms; these often lead to misdiagnosis. Viremia levels and host immune responses may contribute to disease severity. This study was aimed to characterize the ability of ZIKV, CHIKV, and DENV to infect human peripheral blood mononuclear cells (PBMCs) and assess the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-10, and interferon gamma-induced protein (IP)- 10 genes in response to the viral infections.
METHODS PBMCs were isolated from healthy donors using gradient centrifugation. Cells were infected with Indonesian isolates of ZIKV, CHIKV, and DENV for 48 hours. Plaque assays were performed to measure viable virus titers, while viral genomic RNA and the gene expression of TNF-α, IL-10, and IP-10 were determined using real-time quantitative reverse transcription-polymerase chain reaction.
RESULTS The susceptibility of PBMCs to ZIKV, CHIKV, and DENV infection was observed, and the viable virus titer and viral genome quantity were found to be significantly higher in ZIKV and CHIKV. All viruses induced the expression of immune-related proteins. The TNF-α gene was upregulated by all viruses to relatively similar levels. IL-10 expression was highest in response to ZIKV, followed by CHIKV. In contrast, IP-10 expression was highly upregulated in DENV-infected cells and only moderately expressed in ZIKV- and CHIKV-infected cells.
CONCLUSIONS ZIKV, CHIKV, and DENV clinical isolates infected PBMCs with different levels of virus infectivity. The gene expression of IL-10 was highly upregulated in ZIKV infection and IP-10 in DENV infection.
Collapse
|
43
|
Cárdenas DM, Jaimes MA, Vega LD, Oliveros NL, Soto JA, Chía CR, Osorio JE, Ciuoderis KA. Immunological Memory to Zika Virus in a University Community in Colombia, South America. AN ACAD BRAS CIENC 2020; 92:e20190883. [PMID: 32491129 DOI: 10.1590/0001-3765202020190883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/18/2019] [Indexed: 11/22/2022] Open
Abstract
Zika virus appeared in South America in 2015, generating alarm worldwide as it causes microcephaly and autoimmunity. This study aims to determine the serological footprint of the incoming epidemic in a student community and to characterize the memory functional cell response during post convalescence. In a cross-sectional study, Zika-specific IgG using LIA immunoassay was found in 328 university students (CI=95%), while in the second phase, the functional cellular memory response for IFN-γ and IL-2 was quantified using post-stimulus ELISpot with inactivated virus, starting with individuals seropositive for Zika and control individuals (seropositive only for Dengue and seronegative for Zika-Dengue). Depending on the antigen used, memory humoral response (IgG) against Zika Virus was observed in >60% of the population; seropositivity for NS1 was 21.1% higher than E antigen with high intensity. The analysis of cell functionality in 22 individuals seropositive for Zika virus revealed either IFN-γ+ or IL-2+ cells in 86.3% of cases (Th1 profile), presenting multifunctionality in 50% (11 individuals), 64% of which presented> 6 SFC/104 PBMCs (>600 SFC/106 PBMC), reflecting memory circulating cells. A good agreement (Kappa= 0.754) was observed between the coexistence of both cellular and humoral responses but not in their intensity.
Collapse
Affiliation(s)
- Denny M Cárdenas
- Faculty of Health Sciences, Universidad de Santander, Cucuta, Colombia
| | - Miguel A Jaimes
- Faculty of Health Sciences, Universidad de Santander, Cucuta, Colombia
| | - Leidy D Vega
- Faculty of Health Sciences, Universidad de Santander, Cucuta, Colombia
| | | | - Javier A Soto
- Faculty of Health Sciences, Universidad de Santander, Cucuta, Colombia
| | - Claudia R Chía
- Faculty of Health Sciences, Universidad de Santander, Cucuta, Colombia
| | - Jorge E Osorio
- Department of Pathobiological Sciences, University of Wisconsin, Madison, USA
| | - Karl A Ciuoderis
- Center for Research and Surveillance of Tropical and Infectious Diseases, Universidad Nacional de Colombia, Medellin, Colombia
| |
Collapse
|
44
|
Limonta D, Jovel J, Kumar A, Lu J, Hou S, Airo AM, Lopez-Orozco J, Wong CP, Saito L, Branton W, Wong GKS, Mason A, Power C, Hobman TC. Fibroblast Growth Factor 2 Enhances Zika Virus Infection in Human Fetal Brain. J Infect Dis 2020; 220:1377-1387. [PMID: 30799482 DOI: 10.1093/infdis/jiz073] [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: 12/14/2018] [Accepted: 02/12/2019] [Indexed: 12/19/2022] Open
Abstract
Zika virus (ZIKV) is an emerging pathogen that can cause microcephaly and other neurological defects in developing fetuses. The cellular response to ZIKV in the fetal brain is not well understood. Here, we show that ZIKV infection of human fetal astrocytes (HFAs), the most abundant cell type in the brain, results in elevated expression and secretion of fibroblast growth factor 2 (FGF2). This cytokine was shown to enhance replication and spread of ZIKV in HFAs and human fetal brain explants. The proviral effect of FGF2 is likely mediated in part by suppression of the interferon response, which would represent a novel mechanism by which viruses antagonize host antiviral defenses. We posit that FGF2-enhanced virus replication in the fetal brain contributes to the neurodevelopmental disorders associated with in utero ZIKV infection. As such, targeting FGF2-dependent signaling should be explored further as a strategy to limit replication of ZIKV.
Collapse
Affiliation(s)
- Daniel Limonta
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Juan Jovel
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Anil Kumar
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Julia Lu
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Shangmei Hou
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Adriana M Airo
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | | | - Cheung Pang Wong
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Leina Saito
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - William Branton
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Gane Ka-Shu Wong
- Department of Medicine, University of Alberta, Edmonton, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, Canada.,BGI Group, Shenzhen, China
| | - Andrew Mason
- Department of Medicine, University of Alberta, Edmonton, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| | - Christopher Power
- Department of Medicine, University of Alberta, Edmonton, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Tom C Hobman
- Department of Cell Biology, University of Alberta, Edmonton, Canada.,Department of Medicine, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada
| |
Collapse
|
45
|
Zuñiga J, Choreño-Parra JA, Jiménez-Alvarez L, Cruz-Lagunas A, Márquez-García JE, Ramírez-Martínez G, Goodina A, Hernández-Montiel E, Fernández-López LA, Cabrera-Cornejo MF, Cabello C, Castillejos M, Hernández A, Regino-Zamarripa NE, Mendoza-Milla C, Vivanco-Cid H, Escobar-Gutierrez A, Fonseca-Coronado S, Belaunzarán-Zamudio PF, Pérez-Patrigeon S, Guerrero L, Regalado J, Nájera-Cancino G, Caballero-Sosa S, Rincón-León H, Smolskis M, Mateja A, Hunsberger S, Beigel JH, Ruiz-Palacios G. A unique immune signature of serum cytokine and chemokine dynamics in patients with Zika virus infection from a tropical region in Southern Mexico. Int J Infect Dis 2020; 94:4-11. [PMID: 32081772 PMCID: PMC7362833 DOI: 10.1016/j.ijid.2020.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/24/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To describe the kinetics of circulating cytokines and chemokines in humans with ZIKAV infection. METHODS Serum levels of different immune mediators in patients with ZIKAV infection were measured at distinct stages of the disease, as well as in culture supernatants from human monocytes infected with a clinical ZIKAV isolate. We also looked for clinical features associated with specific immune signatures among symptomatic patients. RESULTS We evaluated 23 ZIKAV-infected patients. Their mean age was 32 ± 8.3 years and 65% were female. ZIKAV patients showed elevated IL-9, IL-17A, and CXCL10 levels at acute stages of the disease. At day 28, levels of CCL4 and CCL5 were increased, whereas IL-1RA, CXCL8 and CCL2 were decreased. At baseline, IL-7 was increased among patients with headache, whereas CCL2, and CCL3 were decreased in patients with bleeding and rash, respectively. Our clinical ZIKAV isolate induced a broad immune response in monocytes that did not resemble the signature observed in ZIKAV patients. CONCLUSIONS We showed a unique immune signature in our cohort of ZIKAV-infected patients. Our study may provide valuable evidence helpful to identify immune correlates of protection against ZIKAV.
Collapse
Affiliation(s)
- Joaquín Zuñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico.
| | - José Alberto Choreño-Parra
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Luis Jiménez-Alvarez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - José Eduardo Márquez-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Gustavo Ramírez-Martínez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Aminadab Goodina
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Erika Hernández-Montiel
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Luis Alejandro Fernández-López
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - María Fernanda Cabrera-Cornejo
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Carlos Cabello
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Manuel Castillejos
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Andrés Hernández
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Nora E Regino-Zamarripa
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Criselda Mendoza-Milla
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Héctor Vivanco-Cid
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - Alejandro Escobar-Gutierrez
- Department for Immunological Investigations, Instituto de Diagnóstico y Referencia Epidemiológica, Mexico City, Mexico
| | | | - Pablo F Belaunzarán-Zamudio
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Maryland, USA
| | - Santiago Pérez-Patrigeon
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Lourdes Guerrero
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Justino Regalado
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | | | - Sandra Caballero-Sosa
- Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Tapachula, Chiapas, Mexico
| | | | - Mary Smolskis
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Maryland, USA
| | | | - Sally Hunsberger
- Biostatistics Research Branch (BRB), National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - John H Beigel
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Maryland, USA
| | - Guillermo Ruiz-Palacios
- Departamento de Infectología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
46
|
Yoshikawa FSY, Pietrobon AJ, Branco ACCC, Pereira NZ, Oliveira LMDS, Machado CM, Duarte AJDS, Sato MN. Zika Virus Infects Newborn Monocytes Without Triggering a Substantial Cytokine Response. J Infect Dis 2020; 220:32-40. [PMID: 30785182 DOI: 10.1093/infdis/jiz075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/18/2022] Open
Abstract
Zika virus (ZIKV) is a clinically important flavivirus that can cause neurological disturbances in newborns. Here, we investigated comparatively the outcome of in vitro infection of newborn monocytes by ZIKV. We observed that neonatal cells show defective production of interleukin 1β, interleukin 10, and monocyte chemoattractant protein 1 in response to ZIKV, although they were as efficient as adult cells in supporting viral infection. Although CLEC5A is a classical flavivirus immune receptor, it is not essential to the cytokine response, but it regulates the viral load only in adult cells. Greater expression of viral entry receptors may create a favorable environment for viral invasion in neonatal monocytes. We are the first to suggest a role for CLEC5A in human monocyte infectivity and to show that newborn monocytes are interesting targets in ZIKV pathogenesis, owing to their ability to carry the virus with only a partial triggering of the immune response, creating a potentially favorable environment for virus-related pathologies in young individuals.
Collapse
Affiliation(s)
- Fabio Seiti Yamada Yoshikawa
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina
| | - Anna Julia Pietrobon
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| | - Anna Cláudia Calvielli Castelo Branco
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| | - Nátalli Zanete Pereira
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| | - Luanda Mara da Silva Oliveira
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina
| | | | - Alberto José da Silva Duarte
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina
| | - Maria Notomi Sato
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| |
Collapse
|
47
|
Mocelin HJ, Catão RC, Freitas PS, Prado TN, Bertolde AI, Castro MC, Maciel EL. Analysis of the spatial distribution of cases of Zika virus infection and congenital Zika virus syndrome in a state in the southeastern region of Brazil: Sociodemographic factors and implications for public health. Int J Gynaecol Obstet 2020; 148 Suppl 2:61-69. [PMID: 31975398 PMCID: PMC7065179 DOI: 10.1002/ijgo.13049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To perform spatial distribution analysis of reported cases of Zika virus and congenital Zika syndrome (CZS) in the state of Espírito Santo, Brazil, by neighborhood, and relate the results to sociodemographic indicators and implications for the health process. METHODS An ecological study using data from the 2016 National Notifiable Diseases Surveillance System, epidemiological records, and information on neighborhoods of families confirmed with CZS from qualitative field research. RESULTS Sociodemographic indicators were analyzed in three distinct groups: general population with Zika virus, pregnant women with Zika virus, and cases of CZS visited. For the three groups, average literacy rates were 71.1%, 71.0%, and 68.3%; the average income per minimum wage was 1.4, 1.1, and 1.4; sanitary sewage coverage was 75.6%, 76.1%, and 71.4%; garbage coverage was 90.8%, 91.2%, and 89.2%; and water supply was 93.8%, 94.1% and 93.8%, respectively. Socioeconomic indicators showed no significant differences between groups, although they were above the national average. A nonsignificant variation of 68.3%-71.1% was seen in the average literacy level above 15 years of age. CONCLUSION Socioeconomic and demographic indicators of cases of Zika virus infection and CZS may indicate that the outbreak had different impacts according to class, social group, or gender, reflecting the persistence and social geography of inequality in Brazil.
Collapse
Affiliation(s)
- Helaine J.S. Mocelin
- Laboratory of EpidemiologyFederal University of Espírito SantoVitóriaESBrazil
- Graduate Program in Collective HealthFederal University of Espírito SantoVitóriaESBrazil
| | - Rafael C. Catão
- Departament of GeographyFederal University of Espírito SantoVitóriaESBrazil
| | - Paula S.S. Freitas
- Laboratory of EpidemiologyFederal University of Espírito SantoVitóriaESBrazil
- Graduate Program in Collective HealthFederal University of Espírito SantoVitóriaESBrazil
| | - Thiago N. Prado
- Laboratory of EpidemiologyFederal University of Espírito SantoVitóriaESBrazil
- Graduate Program in Collective HealthFederal University of Espírito SantoVitóriaESBrazil
| | - Adelmo I. Bertolde
- Laboratory of EpidemiologyFederal University of Espírito SantoVitóriaESBrazil
- Department of StatisticsFederal University of Espírito SantoVitóriaESBrazil
| | - Marcia C. Castro
- Department of Global Health and PopulationHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - Ethel L.N. Maciel
- Laboratory of EpidemiologyFederal University of Espírito SantoVitóriaESBrazil
- Graduate Program in Collective HealthFederal University of Espírito SantoVitóriaESBrazil
| |
Collapse
|
48
|
Avelino-Silva VI, Mayaud P, Tami A, Miranda MC, Rosenberger KD, Alexander N, Nacul L, Segurado A, Pohl M, Bethencourt S, Villar LA, Viana IFT, Rabello R, Soria C, Salgado SP, Gotuzzo E, Guzmán MG, Martínez PA, López-Gatell H, Hegewisch-Taylor J, Borja-Aburto VH, Gonzalez C, Netto EM, Saba Villarroel PM, Hoen B, Brasil P, Marques ETA, Rockx B, Koopmans M, de Lamballerie X, Jaenisch T. Study protocol for the multicentre cohorts of Zika virus infection in pregnant women, infants, and acute clinical cases in Latin America and the Caribbean: the ZIKAlliance consortium. BMC Infect Dis 2019; 19:1081. [PMID: 31878895 PMCID: PMC6933915 DOI: 10.1186/s12879-019-4685-9] [Citation(s) in RCA: 7] [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: 04/27/2018] [Accepted: 12/02/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The European Commission (EC) Horizon 2020 (H2020)-funded ZIKAlliance Consortium designed a multicentre study including pregnant women (PW), children (CH) and natural history (NH) cohorts. Clinical sites were selected over a wide geographic range within Latin America and the Caribbean, taking into account the dynamic course of the ZIKV epidemic. METHODS Recruitment to the PW cohort will take place in antenatal care clinics. PW will be enrolled regardless of symptoms and followed over the course of pregnancy, approximately every 4 weeks. PW will be revisited at delivery (or after miscarriage/abortion) to assess birth outcomes, including microcephaly and other congenital abnormalities according to the evolving definition of congenital Zika syndrome (CZS). After birth, children will be followed for 2 years in the CH cohort. Follow-up visits are scheduled at ages 1-3, 4-6, 12, and 24 months to assess neurocognitive and developmental milestones. In addition, a NH cohort for the characterization of symptomatic rash/fever illness was designed, including follow-up to capture persisting health problems. Blood, urine, and other biological materials will be collected, and tested for ZIKV and other relevant arboviral diseases (dengue, chikungunya, yellow fever) using RT-PCR or serological methods. A virtual, decentralized biobank will be created. Reciprocal clinical monitoring has been established between partner sites. Substudies of ZIKV seroprevalence, transmission clustering, disabilities and health economics, viral kinetics, the potential role of antibody enhancement, and co-infections will be linked to the cohort studies. DISCUSSION Results of these large cohort studies will provide better risk estimates for birth defects and other developmental abnormalities associated with ZIKV infection including possible co-factors for the variability of risk estimates between other countries and regions. Additional outcomes include incidence and transmission estimates of ZIKV during and after pregnancy, characterization of short and long-term clinical course following infection and viral kinetics of ZIKV. STUDY REGISTRATIONS: clinicaltrials.gov NCT03188731 (PW cohort), June 15, 2017; clinicaltrials.gov NCT03393286 (CH cohort), January 8, 2018; clinicaltrials.gov NCT03204409 (NH cohort), July 2, 2017.
Collapse
Affiliation(s)
- Vivian I Avelino-Silva
- Department of Infectious and Parasitic Diseases and Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
- Hospital Sirio-Libanes, Sao Paulo, Brazil
| | | | - Adriana Tami
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Facultad de Ciencias de la Salud, Universidad de Carabobo, Valencia, Venezuela
| | | | - Kerstin D Rosenberger
- Department of Infectious Diseases, Section Clinical Tropical Medicine, Heidelberg University Hospital, INF 324, 69120, Heidelberg, Germany
- German Centre for Infection Research (DZIF), Heidelberg Site, Cologne, Germany
| | - Neal Alexander
- London School of Hygiene and Tropical Medicine, London, UK
| | - Luis Nacul
- London School of Hygiene and Tropical Medicine, London, UK
| | - Aluisio Segurado
- Department of Infectious and Parasitic Diseases and Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Moritz Pohl
- Institute of Medical Biometry and Informatic, Heidelberg University Hospital, Heidelberg, Germany
| | - Sarah Bethencourt
- Facultad de Ciencias de la Salud, Universidad de Carabobo, Valencia, Venezuela
| | - Luis A Villar
- Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Isabelle F T Viana
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation (FIOCRUZ), Recife, Brazil
| | - Renata Rabello
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Carmen Soria
- Universidad Católica Santiago de Guayaquil, Guayaquil, Ecuador
- SOSECALI C., Ltda, Guayaquil, Ecuador
| | - Silvia P Salgado
- Instituto Nacional de Investigación en Salud Pública "Dr. Leopoldo Izquieta Pérez", (INSPI), Guayaquil, Ecuador
| | - Eduardo Gotuzzo
- Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - María G Guzmán
- Pedro Kouri Tropical Medicine Institute (IPK), Havana, Cuba
| | | | - Hugo López-Gatell
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Jennifer Hegewisch-Taylor
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | | | - Cesar Gonzalez
- Mexican Institute of Social Security, Mexico City, Mexico
| | | | | | - Bruno Hoen
- INSERM Centre d'Investigation Clinique 1424, Centre Hospitalier Universitaire de Pointe-à-Pitre, Pointe-à-Pitre, France
- Service de Maladies Infectieuses et Tropicales, Dermatologie, Médecine Interne, Centre Hospitalier Universitaire de Pointe-à-Pitre, Pointe-à-Pitre, France
- Université des Antilles et de la Guyane, Faculté de Médecine Hyacinthe Bastaraud, 4537, Pointe-à-Pitre, EA, France
| | - Patrícia Brasil
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Barry Rockx
- Department of Viroscience, WHO CC Arbovirus and hemorrhagic fever viruses reference and research, Erasmus MC, Rotterdam, The Netherlands
| | - Marion Koopmans
- Department of Viroscience, WHO CC Arbovirus and hemorrhagic fever viruses reference and research, Erasmus MC, Rotterdam, The Netherlands
| | | | - Thomas Jaenisch
- Department of Infectious Diseases, Section Clinical Tropical Medicine, Heidelberg University Hospital, INF 324, 69120, Heidelberg, Germany.
- German Centre for Infection Research (DZIF), Heidelberg Site, Cologne, Germany.
| |
Collapse
|
49
|
Differential Pattern of Soluble Immune Markers in Asymptomatic Dengue, West Nile and Zika Virus Infections. Sci Rep 2019; 9:17172. [PMID: 31748599 PMCID: PMC6868147 DOI: 10.1038/s41598-019-53645-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/04/2019] [Indexed: 01/14/2023] Open
Abstract
Infections with dengue virus (DENV), West Nile virus (WNV) and Zika virus (ZIKV) usually present similar mild symptoms at early stages, and most infections (~80%) are asymptomatic. However, these infections may progress to severe disease with different clinical manifestations. In this study we attempted to identify unique characteristics for each infection at the presymptomatic/asymptomatic stage of infection and compared levels of soluble immune markers that have been shown to be altered during clinical course of these viral infections. Levels of soluble markers were determined by Luminex-based assays or by ELISA in plasma samples from asymptomatic blood donors who were reactive for RNA from DENV (n = 71), WNV (n = 52) or ZIKV (n = 44), and a control or non-infected (NI) group (n = 22). Results showed that even in the absence of symptoms, increased interleukin (IL) levels of IL-12, IL-17, IL-10, IL-5, CXCL9, E-Selectin and ST2/IL-1R4; and decreased levels of IL-13 and CD40 were found in all flavivirus group samples, compared to those from NI donors. DENV-infected donors demonstrated variation in expression of IL-1ra and IL-2; WNV-infected donors demonstrated variation in expression of IL-1ra, P-Selectin, IL-4 and IL-5; ZIKV-infected donors demonstrated variation in expression of IL-1ra, P-Selectin, IL-4, RANK-L, CD40L and C3a. The findings suggest that, even in the presymptomatic/asymptomatic phase of the infection, different immunomodulation profiles were associated with DENV, WNV and ZIKV infections.
Collapse
|
50
|
Lineage-dependent differences of Zika virus infection in a susceptible mouse model are associated with different profiles of cytokines, chemokines, growth factors and acute phase proteins. Cytokine 2019; 125:154864. [PMID: 31577989 DOI: 10.1016/j.cyto.2019.154864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/28/2022]
Abstract
Zika virus (ZIKV) is phylogenetically divided into two lineages comprising African (ZIKVAF) and Asian (ZIKVAS) genotypes. In the type-I interferon receptor deficient mouse model, ZIKVAF causes severe disease with all mice meeting humane endpoints with doses as low as 10 plaque-forming units (pfu) whereas a much milder infection is seen after challenge with ZIKVAS, including with doses as high as 106 pfu. Using this mouse model, the elucidation of cytokine, chemokine, growth factor and acute phase protein responses over the course of infection were studied to determine whether these analytes contributed to the stark difference in clinical outcome. Results demonstrated some significant differences, with the ZIKVAF infection being associated with increases in a higher number of biomarkers than ZIKVAS. When low (10 pfu) and high (106 pfu) challenge doses were compared, animals given the lower virus inoculum showed a wider range of responses, indicating a different disease progression compared to those challenged with high doses. These results aid with elucidating the different outcomes with the two lineages of ZIKV and with future work to assess pathogenicity of virus infection.
Collapse
|