1
|
Tamietti C, Stephen T, Rouvinski A, Tenebray B, Leparc‐Goffard I, de Laval F, Fernandes‐Pellerin S, Manuguerra J, Rey F, Hasan M, Badaut C, Flamand M, Matheus S, Briolant S. Prolonged Zika Virus NS1 Protein Circulation in Patient Sera Impacts Clinical Outcome Before the Rise of a Specific IgM Response. J Med Virol 2025; 97:e70368. [PMID: 40263920 PMCID: PMC12015152 DOI: 10.1002/jmv.70368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/19/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Zika virus (ZIKV) is a neurotropic virus that can be transmitted congenitally. In ZIKV-infected pregnant women, placental dysfunction is associated with the secretion of nonstructural protein 1 (NS1). In this study, the kinetics of NS1 secretion and antibody response were assessed and characterized in the serum of ZIKV-positive adult patients recruited in French Guiana. NS1 concentrations were quantified by a single molecule array (SiMoA) in 164 sequential serum samples collected from thirty patients during the first month after onset of symptoms. Serum NS1 concentrations in this cohort were unexpectedly low and ranged from 0.1 pg/mL to 380 pg/mL. The median persistence of NS1 in patients with a clinical score of 2 (6 days) was significantly lower than in patients with a clinical score of 3 (8 days). In both groups of patients, anti-NS1 IgM and IgG kinetics were similar but patients with a milder clinical score of 2 had statistically higher levels of specific IgM than those with a clinical score of 3. Herein, it was shown that NS1 circulating in patient sera is associated with clinical outcome, emphasizing the role of NS1 in ZIKV pathogenesis.
Collapse
Affiliation(s)
- Carole Tamietti
- Institut PasteurUniversité Paris Cité, Unité de Virologie StructuraleParisFrance
| | - Tharshana Stephen
- Institut PasteurUniversité Paris Cité, Single Cell Biomarkers UTechSParisFrance
| | - Alexander Rouvinski
- Institut PasteurUniversité Paris Cité, Unité de Virologie StructuraleParisFrance
| | - Bernard Tenebray
- Unité de virologieInstitut de Recherche Biomédicale des ArméesMarseilleFrance
- Centre National de Référence des ArbovirusInstitut de Recherche Biomédicale des ArméesMarseilleFrance
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, Université di Corsica, IRD 190, Inserm 1207, IRBA)France
| | - Isabelle Leparc‐Goffard
- Unité de virologieInstitut de Recherche Biomédicale des ArméesMarseilleFrance
- Centre National de Référence des ArbovirusInstitut de Recherche Biomédicale des ArméesMarseilleFrance
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, Université di Corsica, IRD 190, Inserm 1207, IRBA)France
| | - Franck de Laval
- Service de Santé des Armées, CESPACentre d'épidémiologie et de santé publique des arméesMarseilleFrance
| | | | - Jean‐Claude Manuguerra
- Institut PasteurUniversité Paris Cité, Laboratory for Urgent Response to Biological Threats (CIBU), Environment and Infectious Risks (ERI) UnitParisFrance
| | - Félix Rey
- Institut PasteurUniversité Paris Cité, Unité de Virologie StructuraleParisFrance
| | - Milena Hasan
- Institut PasteurUniversité Paris Cité, Single Cell Biomarkers UTechSParisFrance
| | - Cyril Badaut
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, Université di Corsica, IRD 190, Inserm 1207, IRBA)France
- Unité de virologieInstitut de Recherche Biomédicale des Armées, Brétigny‐sur‐OrgeFrance
| | - Marie Flamand
- Institut PasteurUniversité Paris Cité, Unité de Virologie StructuraleParisFrance
| | - Séverine Matheus
- Institut PasteurUniversité Paris Cité, Laboratory for Urgent Response to Biological Threats (CIBU), Environment and Infectious Risks (ERI) UnitParisFrance
| | - Sébastien Briolant
- Département Microbiologie et maladies infectieuses, Institut de Recherche Biomédicale des Armées (IRBA)Unité Parasitologie et EntomologieMarseilleFrance
- Aix Marseille Université, SSA, AP‐HMMarseilleFrance
- IHU Méditerranée InfectionMarseilleFrance
| |
Collapse
|
2
|
Santos D, Carrijo Oliveira N, Costa ECA, Ramalho Paes MV, Beltrão-Braga B, Castanha AG, Beltrão-Braga PCB. Modeling potential drugs for Zika virus in animal and in vitro platforms: what is the current state of the art? Expert Opin Drug Discov 2025:1-13. [PMID: 40251755 DOI: 10.1080/17460441.2025.2496461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
INTRODUCTION The Zika virus (ZIKV) poses a significant public health threat due to its association with congenital Zika syndrome (CZS) and severe neurological disorders. Since its discovery, ZIKV has transitioned from sporadic outbreaks to a major epidemic in Brazil in 2015, which highlighted the urgent need for effective therapies, especially for vulnerable groups like pregnant women and newborns. AREAS COVERED This review provides a comprehensive overview of recent advancements in ZIKV drug discovery and their current stage of development, with a particular focus on those tested in animal models from 2018 to date, excluding vaccine candidates. Repurposed drugs, such as molnupiravir and sofosbuvir, have shown the potential to inhibit viral replication and mitigate disease. Novel compounds targeting viral proteins and host-directed therapies are also discussed. Furthermore, advanced in vitro models, including brain organoids, have offered critical insights into therapeutic efficacy. EXPERT OPINION Although some preclinical models have identified potential drugs ready for human translation, no protocol has yet been established for treating ZIKV infection. Currently, the treatment involves supportive care, managing symptoms, and preventing complications, especially for pregnant women. Ongoing research aims to develop specific antiviral therapies and vaccines; however, no such options are currently available.
Collapse
Affiliation(s)
- Debora Santos
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nathalia Carrijo Oliveira
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Institut Pasteur de São Paulo, São Paulo, Brazil
| | | | - Maria Vitória Ramalho Paes
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruna Beltrão-Braga
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andrelissa Gorete Castanha
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Institut Pasteur de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
3
|
Zheng P, Li G, Chen Y, Li S, Yang S, Guo D, Zhou Q, Zhang X. Mini review: SHEN26, a novel oral antiviral drug for COVID-19 treatment. Bioorg Med Chem Lett 2025; 124:130243. [PMID: 40250816 DOI: 10.1016/j.bmcl.2025.130243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Over two years into the pandemic, global collaboration led to effective antiviral drugs targeting SARS-CoV-2's RdRp and 3CL protease. However, the virus continues to evolve, and certain low-virulence variants still circulate. Despite reduced virulence, ongoing transmission raises the risk of new mutations, underscoring the need for continued vigilance, research, and expansion of our antiviral and vaccine strategies. Our research team has developed SHEN26, a promising small-molecule antiviral drug for the treatment of COVID-19. This mini-review explores its development, including history, synthesis, preclinical evaluations, and findings from Phase I and II clinical trials. Data from each research phase further underscores SHEN26's potential as a safe and effective oral antiviral treatment for COVID-19, while also emphasizing its broader relevance in combating emerging RNA viral infections.
Collapse
Affiliation(s)
- Peisen Zheng
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China
| | - Guanguan Li
- Shenzhen AntiV Pharma Co., Ltd., Shenzhen, Guangdong 518081, China
| | - Yuanguang Chen
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China
| | - Shuo Li
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China
| | - Sidi Yang
- Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Deyin Guo
- Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Qifan Zhou
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China.
| | - Xumu Zhang
- Shenzhen Key Laboratory of Small Molecule Drug Discovery and Synthesis, Department of Chemistry, Shenzhen Grubbs Institute and Medi-X Pingshan, Southern University of Science and Technology, Shenzhen 518000, China.
| |
Collapse
|
4
|
Bowers KH, De Angelis D, Birrell PJ. Modelling with SPEED: a Stochastic Predictor of Early Epidemic Detection. J Theor Biol 2025; 607:112120. [PMID: 40189138 DOI: 10.1016/j.jtbi.2025.112120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 04/20/2025]
Abstract
The frequency of emerging infectious disease outbreaks continues to rise, necessitating predictive frameworks for public health decision-making. This study introduces the Stochastic Predictor of Early Epidemic Detection (SPEED) model, an adaptation of the classic Susceptible-Infected-Recovered model, employing a Gillespie-like algorithm to simulate early-stage stochastic disease transmission. SPEED incorporates individual-level detection probabilities based on the infection time and the lag from GP consultation to lab confirmation. The model dynamically adjusts to public health responses by enhancing testing and reducing detection times once a single case has been identified. SPEED serves two key functionalities. First, as a statistical inference tool refining reproduction number estimates following the detection of a small number of cases. SPEED inference uses specified prior distributions for the reproduction number to provide reliable posterior estimates. Second, to simulate epidemic scenarios under specified values of the reproduction number in order to construct a distribution of the time to subsequent detections. The model is used to evaluate how second case timings can rule out higher values of the reproduction number. Comparisons with simulations under heightened surveillance scenarios demonstrate the model's utility in assessing response efficacy on the initial outbreak spread. Our results demonstrate SPEED applied to a single case of influenza A(H1N2)v, detected through routine flu surveillance on the 23rd November 2023.
Collapse
Affiliation(s)
| | - Daniela De Angelis
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Analysis & Intelligence Assessment Directorate, Chief Data Officer Group, UKHSA, UK
| | - Paul J Birrell
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Analysis & Intelligence Assessment Directorate, Chief Data Officer Group, UKHSA, UK.
| |
Collapse
|
5
|
Sheela Rani M, Dhanasekar S. Application of type-2 heptagonal fuzzy sets with multiple operators in multi-criteria decision-making for identifying risk factors of Zika virus. BMC Infect Dis 2025; 25:450. [PMID: 40169983 PMCID: PMC11963685 DOI: 10.1186/s12879-025-10741-9] [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: 08/17/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
PURPOSE This study aims to identify and rank the key risk factors associated with the Zika virus by leveraging a novel multi-criteria decision-making (MCDM) framework based on type-2 heptagonal fuzzy sets. By integrating advanced aggregation operators, the framework effectively addresses uncertainties in expert assessments and enhances decision-making reliability. METHODS A robust MCDM approach was developed using type-2 heptagonal fuzzy sets, which provide a more nuanced representation of uncertainty compared to traditional fuzzy models. These sets were selected due to their superior ability to handle vague, imprecise, and subjective expert judgments, common challenges in epidemiological risk assessments. Arithmetic and geometric aggregation operators were employed to process fuzzy data effectively. To ensure comprehensive and reliable rankings, the framework incorporated both outranking methods and distance-based approaches, specifically TOPSIS and WASPAS. A sensitivity analysis was conducted to validate the stability of the rankings under varying conditions. RESULTS The proposed framework identified Z 3 (unprotected sexual activity) as the most critical risk factor with a score of 0.6717, followed by Z 8 (blood transfusions) at 0.5783, Z 10 (pregnancy) at 0.5753, Z 9 (mosquito bites) at 0.4917, and Z 7 (travel to endemic areas) at 0.4726. The rankings remained consistent across different MCDM methods (TOPSIS and WASPAS), demonstrating the robustness of the proposed approach. Pearson correlation analysis confirmed a strong agreement between methods, with correlation coefficients, reinforcing the reliability of the model. CONCLUSION This study introduces an advanced decision-support system for healthcare professionals to systematically identify and prioritize Zika virus risk factors. By leveraging type-2 heptagonal fuzzy sets, the framework effectively captures and processes uncertainty stemming from incomplete epidemiological data, imprecise expert assessments, and subjective linguistic evaluations. The consistency of rankings across multiple MCDM methods, along with sensitivity analysis confirming their stability, demonstrates the model's reliability. These findings provide a scientifically grounded tool for improving risk analysis and strategic public health interventions.
Collapse
Affiliation(s)
- M Sheela Rani
- Department of Mathematics, School of Advanced Sciences, Vellore Institute of Technology, Chennai, 600127, Tamilnadu, India
| | - S Dhanasekar
- Department of Mathematics, School of Advanced Sciences, Vellore Institute of Technology, Chennai, 600127, Tamilnadu, India.
| |
Collapse
|
6
|
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. [PMID: 40134841 PMCID: PMC11612872 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
|
7
|
Wu XW, Qiao GR, Zhao XR, Li Q, Zeb MA, Li XL, Zheng CB, Xiao WL. 3,4-seco-Prenyllabdane sesterterpenoids and 3,4-seco-labdane diterpenoids with Zika virus inhibitory potential from Callicarpa nudiflora. PHYTOCHEMISTRY 2025; 236:114488. [PMID: 40122274 DOI: 10.1016/j.phytochem.2025.114488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
Two rearranged prenyllabdane sesterterpenoids nudiflorawus A-B (1-2) with a previously unreported carbon skeleton, and five undescribed 3,4-seco-labdane diterpenoids, nudiflorawus C-G (3-7), along with two known diterpenoids (8-9), were isolated from the leaves of Callicarpa nudiflora. Compounds 1-2 exhibited the first example of 3,4-seco-prenyllabdane sesterterpenoids with a unique six-membered ring in the side chain. Their structures were established via various spectroscopic methods. NMR calculations with DP4+ analysis and ECD were further adopted to confirm their relative and absolute configurations. Compound 6 showed significant Zika virus (ZIKV) inhibitory activity with an EC50 value of 25.35 ± 0.742 μM. Western blot, quantitative real-time PCR, and immunofluorescence results further indicated that compound 6 could block ZIKV infection and replication by inhibiting the expression of ZIKV-envelope protein.
Collapse
Affiliation(s)
- Xue-Wen Wu
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650091, People's Republic of China
| | - Guan-Rong Qiao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, People's Republic of China; Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development (Under Construction), Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Xue-Rong Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650091, People's Republic of China
| | - Qi Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650091, People's Republic of China; Southwest United Graduate School, Kunming, 650592, People's Republic of China; Institute of International Rivers and Eco-Security, Yunnan University, Kunming, 650091, People's Republic of China
| | - Muhammad Aurang Zeb
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650091, People's Republic of China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650091, People's Republic of China.
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, People's Republic of China; Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development (Under Construction), Kunming Medical University, Kunming, 650500, People's Republic of China.
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, 650091, People's Republic of China; Southwest United Graduate School, Kunming, 650592, People's Republic of China; Institute of International Rivers and Eco-Security, Yunnan University, Kunming, 650091, People's Republic of China.
| |
Collapse
|
8
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Toll-like receptor response to Zika virus infection: progress toward infection control. NPJ VIRUSES 2025; 3:20. [PMID: 40295746 DOI: 10.1038/s44298-025-00102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025]
Abstract
Infection with the Zika virus (ZIKV) poses a threat to human health. An improved understanding of the host Toll-like receptor response, disease onset, and viral clearance in vivo and in vitro may lead to the development of therapeutic or prophylactic interventions against viral infections. Currently, no clinically approved ZIKV vaccine is available, highlighting the need for its development. In this study, we discuss the progress in the Zika vaccine, including advances in the use of Toll-like receptor agonists as vaccine adjuvants to enhance vaccine efficacy.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, Bangladesh.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
9
|
Pawan, Devi S. Designing of new trans-stilbene derivative: An entry barrier of Zika virus in host cell. J Mol Graph Model 2025; 135:108935. [PMID: 39731815 DOI: 10.1016/j.jmgm.2024.108935] [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: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
A large population in the world lives in tropical and subtropical regions, showing a high risk of Zika viral infection which leads to a situation of global health emergency and demands extensive research to create effective antiviral medicines. Herein, we introduce the design of a new derivatized trans-stilbene molecule to investigate the inhibition of Zika virus entry into the host cell by molecular docking approach. The synthesized compound has been characterized by different analytical techniques such as FTIR, 1H NMR,13C NMR and UV-visible spectroscopy as well as Mass spectrometry (MS). Moreover, the complete structure elucidation was achieved via X-ray crystallography and DFT analysis. The article describes the life cycle and genome of the Zika virus along with its mechanism of entry inhibition by illustrating the structure and function of the ZIKV envelop (E) protein. The docking studies disclosed that the newly synthesized stilbene compound confers an excellent inhibitory response towards the entry of Zika virus in host cells as supported by calculated docking score and its binding conformation with Zika virus E-protein. Further, the normal mode analysis (NMA) simulation technique is used to predict the conformational states of the target E-protein, which explains the potency of the compound to bind with the Zika virus E-protein. We hope that the present study will help and encourage researchers in the field of medicinal chemistry to develop potential drugs against the Zika virus.
Collapse
Affiliation(s)
- Pawan
- Department of Chemistry, Goswami Ganesh Dutta Sanatan Dharma (GGDSD) College, Chandigarh, 160030, India.
| | - Sonia Devi
- Post Graduate Department of Chemistry, Mehr Chand Mahajan DAV College for Women, Chandigarh, 160036, India
| |
Collapse
|
10
|
dos Santos ALS, Rosolen BB, Ferreira FC, Chiancone IS, Pereira SS, Pontes KFM, Traina E, Werner H, Granese R, Araujo Júnior E. Intrauterine Zika Virus Infection: An Overview of the Current Findings. J Pers Med 2025; 15:98. [PMID: 40137414 PMCID: PMC11943202 DOI: 10.3390/jpm15030098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus of the family Flaviviridae. The association between ZIKV and microcephaly was first described in Brazil in 2015. The risk of vertical transmission occurs in pregnant women with or without symptoms, and the risk of malformation appears to be worse when infection occurs in the first and second trimesters of pregnancy. The rate of vertical transmission varies from 26 to 65%, and not all fetuses develop malformations. The incidence of malformations resulting from transmission is uncertain, ranging from 6-8% in the US to 40% in Brazil. Congenital ZIKV syndrome is a set of clinical manifestations that can affect the fetus of a mother infected with ZIKV. The manifestations are broad and nonspecific, including microcephaly, subcortical calcifications, ocular changes, congenital contractures, early hypertension, and pyramidal and extrapyramidal signs. Other findings such as growth restriction and fetal miscarriage/death may also occur. Our aim in this article is to review the literature on mosquito transmission, clinical presentation, serologic diagnosis, intrauterine transmission, pre- and postnatal imaging diagnostic findings, and short- and long-term follow-up.
Collapse
Affiliation(s)
- Ana Luiza Soares dos Santos
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Beatriz Bussi Rosolen
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Fernanda Curvelo Ferreira
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Isabella Samões Chiancone
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Stefany Silva Pereira
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Karina Felippe Monezi Pontes
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
- Service of Gynecology and Obstetrics, Ipiranga Hospital, São Paulo 04262-000, SP, Brazil
| | - Evelyn Traina
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
| | - Heron Werner
- Department of Fetal Medicine, Biodesign Laboratory DASA/PUC, Rio de Janeiro 22453-900, RJ, Brazil;
| | - Roberta Granese
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, “G. Martino” University Hospital, 98100 Messina, Italy
| | - Edward Araujo Júnior
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
| |
Collapse
|
11
|
Gardinali NR, Marchevsky RS, Vieira YC, Pelajo-Machado M, Kugelmeier T, Melgaço JG, Castro MP, de Oliveira JM, Pinto MA. Congenital Zika virus infection in laboratory animals: a comparative review highlights translational studies on the maternal-foetal interface. Mem Inst Oswaldo Cruz 2025; 120:e240125. [PMID: 40052994 PMCID: PMC11884655 DOI: 10.1590/0074-02760240125] [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: 06/17/2024] [Accepted: 10/23/2024] [Indexed: 03/10/2025] Open
Abstract
The 2015-16 Zika virus (ZIKV) epidemic has posed unprecedented concern for maternal-infant health, mainly due to the substantial risk of microcephaly and other neurological birth abnormalities associated with congenital ZIKV syndrome (CZS). As licenced vaccines and effective antivirals are still unavailable, attention has been focused on post-delivery in vitro or translational in vivo studies to understand the impact of maternal ZIKV infection on placentation and neurodevelopmental consequences for the foetus. Here, we review clinical and translational studies highlighting ZIKV-induced maternal-foetal interface dysfunction, adding to our previous observations of experimental ZIKV vertical transmission to pregnant rhesus monkeys and newly published post-epidemic findings about the theme. This comparative review focuses on the mechanisms by which the virus has a cytopathic effect on trophoblasts and macrophages during placentation in humans, nonhuman primates, and rodent transgenic models, crosses the placental barrier, replicates, and establishes a persistent uteroplacental infection. When considering the mechanism of ZIKV-induced birth defects in humans and other susceptible hosts, it becomes apparent how the various stages of the ZIKV cycle in the host (both the parent and offspring) unfold. This understanding presents specific opportunities for pharmacological intervention and the development of preventative vaccines.
Collapse
Affiliation(s)
- Noemi Rovaris Gardinali
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Desenvolvimento Tecnológico em Virologia, Rio de Janeiro, RJ, Brasil
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Laboratório de Tecnologia Virológica, Rio de Janeiro, RJ, Brasil
| | - Renato Sergio Marchevsky
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Departamento de Experimentos Pré-Clínicos, Laboratório de Ensaios Pré-Clínicos, Rio de Janeiro, RJ, Brasil
| | - Yara Cavalcante Vieira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Desenvolvimento Tecnológico em Virologia, Rio de Janeiro, RJ, Brasil
- The Pennsylvania State University, Department of Food Science, University Park, PA, USA
| | - Marcelo Pelajo-Machado
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Medicina Experimental e Saúde, Rio de Janeiro, RJ, Brasil
| | - Tatiana Kugelmeier
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Ciência e Tecnologia em Biomodelos, Rio de Janeiro, RJ, Brasil
| | - Juliana Gil Melgaço
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Departamento de Experimentos Pré-Clínicos, Laboratório de Tecnologia Imunológica, Rio de Janeiro, RJ, Brasil
| | | | - Jaqueline Mendes de Oliveira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Desenvolvimento Tecnológico em Virologia, Rio de Janeiro, RJ, Brasil
| | - Marcelo Alves Pinto
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Desenvolvimento Tecnológico em Virologia, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
12
|
Rajaiah P, Gupta B, Mayilsamy M. ZIKA Virus, an Emerging Arbovirus in India: A Glimpse of Global Genetic Lineages. Microorganisms 2025; 13:544. [PMID: 40142437 PMCID: PMC11946211 DOI: 10.3390/microorganisms13030544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 03/28/2025] Open
Abstract
ZIKA fever (ZIKAF) is an emerging mosquito-borne flavivirus illness in humans. Regarding the etiological agent, ZIKA virus (ZIKAV), though it is known to be distributed in the tropics, causing sporadic cases, its rapid global expansion with pandemic potential has raised global concern. Due to its abrupt emergence in South American countries, the Caribbean, and the Americas, the WHO declared ZIKA a public health emergency of international concern in 2016. ZIKAV usually causes mild infections; however, its recent unusual presentations of Guillen-Barré syndrome in adults and microcephaly in newborn babies of ZIKAV-infected mothers in Brazil has caused concern among global public health authorities. Certain mutations on virus genomes have been found to be correlated with clinical severity, and its unusual transmission routes through sexual and blood transfusions emphasize the necessity for understanding its virological determinants and impact. Its abrupt re-emergence in India (2018-2019), particularly in Gujarat (2016), Tamil Nadu (2017), Uttar Pradesh (2021), Maharashtra, Kerala (2021), and Karnataka (2023), has indicated the need for urgent measures to strengthen surveillance systems and design effective prevention and control measures in this country. Given the global concern around ZIKAV, here, we reviewed current knowledge about global ZIKAV genetic lineages vis à vis the situation in India and discussed future priorities for ZIKAV research in India for effectively designing control strategies.
Collapse
Affiliation(s)
- Paramasivan Rajaiah
- ICMR-Vector Control Research Centre, 4, Sarojini Street, Chinna Chokkikulam, Madurai 625 002, India; (B.G.); (M.M.)
| | | | | |
Collapse
|
13
|
Roy RR, Tadkalkar N, Deshpande GR, Atre NM, Shil P, Sapkal G. Identification of B-cell epitopes of Indian Zika virus strains using immunoinformatics. Front Immunol 2025; 16:1534737. [PMID: 40083545 PMCID: PMC11903408 DOI: 10.3389/fimmu.2025.1534737] [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: 11/26/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction The Zika virus is an emerging Flavivirus known to cause Zika infection in humans. It is associated with severe health problems such as microcephaly and Guillain-Barré syndrome post the Brazilian epidemic in 2015-16. The spread of the Zika virus to the Asian subcontinent, especially to India is a matter of great concern. Two recent co-circulating Indian Zika virus strains such as Rajasthan and Maharashtra detected in 2018 and 2021 were studied to identify B-cell epitopes in the envelope and non-structural 1 protein as these epitopes are major indicators of robust humoral immune response. The study aimed at identifying novel epitopes, followed by molecular docking with potent Zika virus-specific monoclonal antibodies. The novel epitopes identified in this study shall be essential in designing multi-epitope vaccines capable of inducing antibody response against Zika virus infection. Methods ABCpred, BepiPred 2.0 and Kolaskar-Tongaonkar methods were used for predicting the linear B-cell epitopes, and Discotope 2.0 and ElliPro were used for the prediction of conformational epitopes. Linear epitopes were further checked for protective antigenicity, allergenicity and toxicity. Based on the stringent study design criteria, only the novel epitopes were considered for molecular docking with complementary determining regions of potent Zika virus-specific monoclonal antibodies. Results Nineteen linear and five conformational epitopes were shortlisted based on protective potential, non-allergic and non-toxic properties for Zika virus E protein, from which nine linear and three conformational epitopes were identified as novel. Molecular docking studies revealed that the novel linear epitopes, one each from EDIII, EDII, EDI and EDI/DIII hinge were involved in epitope-CDR interactions with potent neutralizing Zika virus E-specific mouse monoclonal antibody ZV-67. Moreover, the novel EDII epitope was exclusively engaged in epitope-CDR interactions of potent neutralizing Zika virus E-specific human monoclonal antibody Z3L1. None of the linear epitopes of Zika virus NS1 were ascertained as novel based on our study criteria. Conformational epitopes were identified as novel for NS1 protein. Conclusion This study identified Zika virus-specific novel epitopes of envelope and non-structural -1 proteins in the currently co-circulating Indian strains. Furthermore, in-silico validation through molecular docking added insight into antigen-antibody interactions, paving way for future in vitro and in vivo studies.
Collapse
Affiliation(s)
- Rohan Raj Roy
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Nitali Tadkalkar
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Gururaj Rao Deshpande
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Nitin M. Atre
- Bioinformatics and Data Management, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Pratip Shil
- Bioinformatics and Data Management, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Gajanan Sapkal
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| |
Collapse
|
14
|
Li A, Coffey LL, Mohr EL, Raper J, Chahroudi A, Ausderau KK, Aliota MT, Friedrich TC, Mitzey AM, Koenig MR, Golos TG, Jaeger HK, Roberts VHJ, Lo JO, Smith JL, Hirsch AJ, Streblow DN, Newman CM, O'Connor DH, Lackritz EM, Van Rompay KKA, Adams Waldorf KM. Role of non-human primate models in accelerating research and developing countermeasures against Zika virus infection. THE LANCET. MICROBE 2025:101030. [PMID: 40024258 DOI: 10.1016/j.lanmic.2024.101030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/19/2024] [Accepted: 10/21/2024] [Indexed: 03/04/2025]
Abstract
Zika virus, a mosquito-transmitted orthoflavivirus, has become a pathogen of global health concern ever since the virus caused an epidemic in Brazil in 2015 associated with approximately 700 000 laboratory-confirmed cases of congenital microcephaly. The subsequent spread of the epidemic in 2016 resulted in a wide spectrum of congenital neurological, ophthalmological, and developmental abnormalities across the Americas, Africa, and Asia. In this context, non-human primate models have become essential tools for Zika virus research to understand the pathogenesis of congenital brain injury and perinatal complications and for developing and testing medical countermeasures such as vaccines, diagnostics, and therapeutics. Fetal brain injury has been observed across various non-human primate species and is influenced by factors such as the Zika virus strain, gestational age at inoculation, and inoculation dose and route. Miscarriages are also seen as common outcomes of first trimester Zika virus infections. This Series paper reviews the diverse non-human primate models currently used for Zika virus research to mitigate the public health effects of future Zika virus epidemics.
Collapse
Affiliation(s)
- Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Lark L Coffey
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, Davis, CA, USA
| | - Emma L Mohr
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessica Raper
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Emory National Primate Research Center, Atlanta, GA, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Emory National Primate Research Center, Atlanta, GA, USA
| | - Karla K Ausderau
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, USA; Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew T Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota Twin Cities, St Paul, MN, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ann M Mitzey
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle R Koenig
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Hannah K Jaeger
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Victoria H J Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Jamie O Lo
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, USA; Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Jessica L Smith
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, USA
| | - Christina M Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin National Primate Research Center, Madison, WI, USA
| | - Eve M Lackritz
- Center for Infectious Disease Research and Policy (CIDRAP), University of Minnesota, Minneapolis, MN, USA
| | - Koen K A Van Rompay
- Department of Pathology, Microbiology, and Immunology, University of California, Davis, Davis, CA, USA; California National Primate Research Center, Davis, CA, USA
| | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Washington National Primate Research Center, Seattle, WA, USA.
| |
Collapse
|
15
|
Michita RT, Tran LB, Bark SJ, Kumar D, Toner SA, Jose J, Mysorekar IU, Narayanan A. Zika virus NS1 drives tunneling nanotube formation for mitochondrial transfer and stealth transmission in trophoblasts. Nat Commun 2025; 16:1803. [PMID: 39979240 PMCID: PMC11842757 DOI: 10.1038/s41467-025-56927-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Zika virus (ZIKV) is unique among orthoflaviviruses in its vertical transmission capacity in humans, yet the underlying mechanisms remain incompletely understood. Here, we show that ZIKV induces tunneling nanotubes (TNTs) in placental trophoblasts which facilitate transfer of viral particles, proteins, mitochondria, and RNA to neighboring uninfected cells. TNT formation is driven exclusively via ZIKV non-structural protein 1 (NS1). Specifically, the N-terminal 1-50 amino acids of membrane-bound ZIKV NS1 are necessary for triggering TNT formation in host cells. Trophoblasts infected with TNT-deficient ZIKVΔTNT mutant virus elicited a robust antiviral IFN-λ 1/2/3 response relative to WT ZIKV, suggesting TNT-mediated trafficking allows ZIKV cell-to-cell transmission camouflaged from host defenses. Using affinity purification-mass spectrometry of cells expressing wild-type NS1 or non-TNT forming NS1, we found mitochondrial proteins are dominant NS1-interacting partners. We demonstrate that ZIKV infection or NS1 expression induces elevated mitochondria levels in trophoblasts and that mitochondria are siphoned via TNTs from healthy to ZIKV-infected cells. Together our findings identify a stealth mechanism that ZIKV employs for intercellular spread among placental trophoblasts, evasion of antiviral interferon response, and the hijacking of mitochondria to augment its propagation and survival and offers a basis for novel therapeutic developments targeting these interactions to limit ZIKV dissemination.
Collapse
Affiliation(s)
- Rafael T Michita
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Long B Tran
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Steven J Bark
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Deepak Kumar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shay A Toner
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Joyce Jose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Huffington Centre on Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Anoop Narayanan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA.
| |
Collapse
|
16
|
Ni X, Richardson RB, Godoy AS, Ferla MP, Kikawa C, Scheen J, Hannon WW, Capkin E, Lahav N, Balcomb BH, Marples PG, Fairhead M, Wang S, Williams EP, Tomlinson CWE, Aschenbrenner JC, Lithgo RM, Winokan M, Giroud C, Chandran AV, Walsh M, Thompson W, Bloom JD, Barr H, Kirkegaard K, Koekemoer L, Fearon D, Evans MJ, von Delft F. Crystallographic fragment screening and deep mutational scanning of Zika virus NS2B-NS3 protease enable development of resistance-resilient inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.29.591502. [PMID: 38746305 PMCID: PMC11092485 DOI: 10.1101/2024.04.29.591502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The Zika viral protease NS2B-NS3 is essential for the cleavage of viral polyprotein precursor into individual structural and non-structural (NS) proteins and is therefore an attractive drug target. Generation of a robust crystal system of co-expressed NS2B-NS3 protease has enabled us to perform a crystallographic fragment screening campaign with 1076 fragments. 47 fragments with diverse scaffolds were identified to bind in the active site of the protease, with another 6 fragments observed in a potential allosteric site. To identify binding sites that are intolerant to mutation and thus suppress the outgrowth of viruses resistant to inhibitors developed from bound fragments, we performed deep mutational scanning of NS2B-NS3 protease. Merging fragment hits yields an extensive set of 'mergers', defined as synthetically accessible compounds that recapitulate constellations of observed fragment-protein interactions. In addition, the highly sociable fragment hits enable rapid exploration of chemical space via algorithmic calculation and thus yield diverse possible starting points that maximally explore the binding opportunities to NS2B-NS3 protease, facilitating its resistance-resilient antiviral development.
Collapse
Affiliation(s)
- Xiaomin Ni
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - R Blake Richardson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andre Schutzer Godoy
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100 - Jardim Santa Angelina, São Carlos, 13563-120, Brazil
| | - Matteo P Ferla
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Caroline Kikawa
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Division of Basic Sciences, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jenke Scheen
- Open Molecular Software Foundation, Davis, CA 95618, USA
| | - William W Hannon
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Eda Capkin
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
| | - Noa Lahav
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science
| | - Blake H Balcomb
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Peter G Marples
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Michael Fairhead
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Siyi Wang
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Eleanor P Williams
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Charlie W E Tomlinson
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Jasmin Cara Aschenbrenner
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Ryan M Lithgo
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Max Winokan
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Charline Giroud
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Anu V Chandran
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Martin Walsh
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Warren Thompson
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Jesse D Bloom
- Division of Basic Sciences, Fred Hutch Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, Washington, USA
| | - Haim Barr
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science
| | - Karla Kirkegaard
- Department of Genetics, Stanford University, Palo Alto, California, USA
- Department of Microbiology and Immunology, Stanford University, Palo Alto, California, USA
| | - Lizbé Koekemoer
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Daren Fearon
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Matthew J Evans
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frank von Delft
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| |
Collapse
|
17
|
Fomda BA, Murtaza M, Kakru DK, Lone SA, Sheikh I, Ayoub S, Nazir M. Prevalence of Emerging Arboviral Infections: A Tertiary Care Hospital-Based Study from Kashmir, Northern India. Vector Borne Zoonotic Dis 2025; 25:148-154. [PMID: 39422570 DOI: 10.1089/vbz.2024.0085] [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] [Indexed: 10/19/2024] Open
Abstract
Background: Arboviruses are becoming a global public health menace. The common diseases worldwide caused by arboviruses are dengue, chikungunya, and Zika. This study aims to determine the prevalence of these three arboviral infections in patients with acute febrile illness at a tertiary care hospital in Kashmir, North India. Materials and Methods: A total of 812 blood samples were collected and tested for anti-dengue, anti-chikungunya immunoglobulin M (IgM), and dengue nonstructural protein 1 antigen by enzyme linked immunosorbent assay. Molecular testing of these samples was also done to detect dengue, chikungunya, and Zika viruses by using the CDC Trioplex real-time PCR assay. Results: The prevalence of diseases found among the studied patients was dengue 105/812 (12.93%) followed by chikungunya 17/812 (2.09%), and 3 cases (0.37%) were positive for both dengue and chikungunya; however, no case of Zika was detected. Interestingly, we found that only individuals that had a history of travel to different destinations within the country were positive for these viruses. Most affected cases were males 105/812 (12.93%) compared with females 20/812 (2.46%). Dengue serotyping results indicate that Dengue virus-1 was the most commonly found serotype. The most common symptoms in patients positive for dengue and chikungunya were fever, intense fatigue, myalgia, arthralgia, retro-orbital pain, anorexia, conjunctivitis, and skin rash. Conclusion: This study showed that dengue, chikungunya, and Zika viruses are not prevalent in the indigenous population of Kashmir. However, screening for these agents is required in people who have recently traveled outside Kashmir and have symptoms of acute febrile illness.
Collapse
Affiliation(s)
- Bashir Ahmad Fomda
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Masqooba Murtaza
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Dalip K Kakru
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Shabir Ahmad Lone
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Imtiyaz Sheikh
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Shahnawaz Ayoub
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Mubashir Nazir
- Department of Microbiology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| |
Collapse
|
18
|
Pavia G, Branda F, Ciccozzi A, Romano C, Locci C, Azzena I, Pascale N, Marascio N, Quirino A, Gigliotti S, Divenuto F, Matera G, Giovanetti M, Casu M, Sanna D, Ceccarelli G, Ciccozzi M, Scarpa F. The issue of climate change and the spread of tropical diseases in Europe and Italy: vector biology, disease transmission, genome-based monitoring and public health implications. Infect Dis (Lond) 2025; 57:121-136. [PMID: 39663537 DOI: 10.1080/23744235.2024.2437027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Climate change significantly influences the distribution and severity of tropical diseases. Rising temperatures, changing precipitation patterns, and extreme weather events are transforming the habitats of vectors like mosquitoes and ticks, promoting their proliferation and geographic spread. These changes have facilitated the resurgence of diseases such as malaria, dengue, and chikungunya fever in previously unaffected areas, including parts of Europe and Italy. OBJECTIVE AND METHODS This review aims to explore the relationship between climate change and the spread of vector-borne and tropical parasitic diseases across Europe, with a particular focus on Italy. Recent studies are analyzed to identify emerging trends in disease transmission influenced by shifting climates. Genome-based monitoring and predictive models incorporating climatic and ecological data are highlighted as methods to enhance disease surveillance and preparedness. RESULTS The analysis reveals a clear link between climate change and altered disease patterns. The proliferation of vectors into new territories is associated with increased incidence of diseases. Genome-based tools demonstrate their utility in tracking the evolution of pathogens, particularly regarding changes in virulence, drug resistance, and adaptability to new climates. Predictive models have proven effective in anticipating outbreaks and supporting timely public health interventions. CONCLUSIONS To mitigate the risks posed by climate-induced changes in disease dynamics, continuous monitoring and international collaboration are essential. Strengthening health systems' resilience through mitigation and adaptation strategies is crucial for preventing future epidemics. These insights contribute to the development of sustainable long-term policies for managing tropical diseases in the context of climate change, ensuring timely responses to public health emergencies.
Collapse
Affiliation(s)
- Grazia Pavia
- Department of Health Sciences, Unit of Clinical Microbiology, "Magna Græcia" University of Catanzaro - "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | | | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Locci
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Ilenia Azzena
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Noemi Pascale
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Nadia Marascio
- Department of Health Sciences, Unit of Clinical Microbiology, "Magna Græcia" University of Catanzaro - "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| | - Angela Quirino
- Department of Health Sciences, Unit of Clinical Microbiology, "Magna Græcia" University of Catanzaro - "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| | - Simona Gigliotti
- Department of Health Sciences, Unit of Clinical Microbiology, "Magna Græcia" University of Catanzaro - "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| | - Francesca Divenuto
- Department of Health Sciences, Unit of Clinical Microbiology, "Magna Græcia" University of Catanzaro - "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| | - Giovanni Matera
- Department of Health Sciences, Unit of Clinical Microbiology, "Magna Græcia" University of Catanzaro - "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| | - Marta Giovanetti
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Rome, Italy
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
- Climate Amplified Diseases And Epidemics (CLIMADE), Brasilia, Brazil
| | - Marco Casu
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University Hospital Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
19
|
Torii S, Lord JS, Lavina M, Prot M, Lecuyer A, Diagne CT, Faye O, Faye O, Sall AA, Bonsall MB, Simon-Lorière E, Montagutelli X, Lambrechts L. Polygenic viral factors enable efficient mosquito-borne transmission of African Zika virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634482. [PMID: 39896559 PMCID: PMC11785240 DOI: 10.1101/2025.01.23.634482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus primarily transmitted among humans by Aedes aegypti. Over the past two decades, it has caused significant outbreaks associated with birth defects and neurological disorders. Phylogenetically, ZIKV consists of two main genotypes referred to as the African and Asian lineages, each exhibiting distinct biological properties. African lineage strains are transmitted more efficiently by mosquitoes, but pinpointing the genetic basis of this difference has remained challenging. Here, we address this question by comparing recent African and Asian strains using chimeric viruses, in which segments of the parental genomes are swapped. Our results show that the structural genes from the African strain enhance viral internalization, while the non-structural genes improve genome replication and infectious particle production in mosquito cells. In vivo mosquito transmission is most significantly influenced by the structural genes, although no single viral gene alone determines this effect. Additionally, we develop a stochastic model of in vivo viral dynamics in mosquitoes that mirrors the observed patterns, suggesting that the primary difference between the African and Asian strains lies in their ability to traverse the mosquito salivary glands. Overall, our findings suggest that the polygenic nature of ZIKV transmissibility has prevented Asian lineage strains from achieving the same epidemic potential as African lineage strains, underscoring the importance of lineage-specific adaptive landscapes in shaping ZIKV evolution and emergence.
Collapse
Affiliation(s)
- Shiho Torii
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Jennifer S. Lord
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Morgane Lavina
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Matthieu Prot
- Institut Pasteur, Université Paris Cité, Paris, Evolutionary Genomics of RNA Viruses Unit, Paris France
| | - Alicia Lecuyer
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Cheikh T. Diagne
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Oumar Faye
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Ousmane Faye
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Amadou A. Sall
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | | | - Etienne Simon-Lorière
- Institut Pasteur, Université Paris Cité, Paris, Evolutionary Genomics of RNA Viruses Unit, Paris France
| | - Xavier Montagutelli
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Louis Lambrechts
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| |
Collapse
|
20
|
Logiudice J, Alberti M, Ciccarone A, Rossi B, Tiecco G, De Francesco MA, Quiros-Roldan E. Introduction of Vector-Borne Infections in Europe: Emerging and Re-Emerging Viral Pathogens with Potential Impact on One Health. Pathogens 2025; 14:63. [PMID: 39861024 PMCID: PMC11768692 DOI: 10.3390/pathogens14010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The rise and resurgence of vector-borne diseases (VBDs) in Europe pose an expanding public health challenge, exacerbated by climate change, globalization, and ecological disruptions. Both arthropod-borne viruses (arboviruses) transmitted by ticks such as Crimean-Congo hemorrhagic fever and arboviruses transmitted by mosquitoes like dengue, Chikungunya, Zika, and Japanese encephalitis have broadened their distribution due to rising temperatures, changes in rainfall, and increased human mobility. By emphasizing the importance of interconnected human, animal, and environmental health, integrated One Health strategies are crucial in addressing this complex issue. Europe faces increased risk due to the expanding habitats of disease-carrying organisms, the spread of new species like Aedes albopictus since 2013, and increased movement of infected individuals between countries, leading European countries to implement strategies such as enhanced surveillance systems, public awareness campaigns, and prompt outbreak response strategies. However, the lack of both targeted antiviral therapies and vaccines for many arboviruses, together with undetected or asymptomatic cases, hamper containment efforts. Therefore, it is important to have integrated strategies that combine climate modeling, disease surveillance, and public health interventions to address expected changes in disease patterns due to global changes. This review explores the spread of arboviruses in Europe, highlighting their historical context, current transmission dynamics, and their impact on public health.
Collapse
Affiliation(s)
- Jacopo Logiudice
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (J.L.); (M.A.); (A.C.); (G.T.); (E.Q.-R.)
| | - Maria Alberti
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (J.L.); (M.A.); (A.C.); (G.T.); (E.Q.-R.)
| | - Andrea Ciccarone
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (J.L.); (M.A.); (A.C.); (G.T.); (E.Q.-R.)
| | - Benedetta Rossi
- Unit of Infectious and Tropical Diseases, ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
- Department of Experimental Medicine and Public Health, School of Advanced Studies, University of Camerino, 62032 Camerino, Italy
| | - Giorgio Tiecco
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (J.L.); (M.A.); (A.C.); (G.T.); (E.Q.-R.)
| | - Maria Antonia De Francesco
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy
| | - Eugenia Quiros-Roldan
- Department of Clinical and Experimental Sciences, Unit of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (J.L.); (M.A.); (A.C.); (G.T.); (E.Q.-R.)
| |
Collapse
|
21
|
Guardado K, López-Balderas N, Morales-Romero J, Sampieri CL, Zenteno-Cuevas R, Álvarez-Bañuelos MT, Ramos-Ligonio Á, Ortiz-León MC, Varela-Cardoso M, Montero H. Zika Virus Infection in Asymptomatic Pregnant Women. Infect Dis Rep 2025; 17:2. [PMID: 39846705 PMCID: PMC11755599 DOI: 10.3390/idr17010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025] Open
Abstract
Background/Objectives: Zika disease is caused by the Zika virus (ZIKV) and represents a major public health problem because of the complications in newborn babies from mothers who were infected during pregnancy. It is estimated that 80% of infected pregnant women are asymptomatic, which complicates the identification of infected individuals. In this study, we aimed to detect ZIKV in asymptomatic pregnant women and the effects in the newborns were analyzed. Methods: The presence of ZIKV was evaluated through endpoint reverse transcription-polymerase chain reaction (RT-PCR) in 114 blood samples from pregnant women treated at two hospitals in the state of Veracruz, Mexico. There was a follow-up of the participants until the birth of their newborns. Results: ZIKV RNA was detected in 4.4% (n = 5) of cases. In two positive cases, two consecutive samples were obtained, and one case of persistence of ZIKV in serum after 90 days after delivery was identified. A total of 80% of the positive cases were identified after the third trimester of pregnancy and 20% after the second trimester. Although ZIKV was shown to be a risk factor for low weight and low size at birth and prematurity, after adjustment for other variables, it did not show a significant association. In contrast, preeclampsia/eclampsia was identified as a significant risk factor for low birth weight. Conclusions: The prevalence of ZIKV found in this study suggests a latent circulation of this virus and highlights the importance of epidemiological surveillance in endemic zones. The prolonged viremia that was found suggests the need for more research because of the high impact which can mean the possible dissemination of the virus to the vector.
Collapse
Affiliation(s)
- Kathia Guardado
- Virus-Cell Interactions Laboratory, Institut Pasteur of Montevideo, Montevideo 11400, Uruguay;
- Biomedical Research Center, University of Veracruz, Xalapa 91190, Mexico
| | | | - Jaime Morales-Romero
- Institute of Public Health, University of Veracruz, Xalapa 91190, Mexico; (J.M.-R.); (C.L.S.); (R.Z.-C.); (M.T.Á.-B.); (M.C.O.-L.)
| | - Clara Luz Sampieri
- Institute of Public Health, University of Veracruz, Xalapa 91190, Mexico; (J.M.-R.); (C.L.S.); (R.Z.-C.); (M.T.Á.-B.); (M.C.O.-L.)
| | - Roberto Zenteno-Cuevas
- Institute of Public Health, University of Veracruz, Xalapa 91190, Mexico; (J.M.-R.); (C.L.S.); (R.Z.-C.); (M.T.Á.-B.); (M.C.O.-L.)
| | - María Teresa Álvarez-Bañuelos
- Institute of Public Health, University of Veracruz, Xalapa 91190, Mexico; (J.M.-R.); (C.L.S.); (R.Z.-C.); (M.T.Á.-B.); (M.C.O.-L.)
| | - Ángel Ramos-Ligonio
- Faculty of Chemical Sciences, University of Veracruz, Orizaba 94340, Mexico;
| | - María Cristina Ortiz-León
- Institute of Public Health, University of Veracruz, Xalapa 91190, Mexico; (J.M.-R.); (C.L.S.); (R.Z.-C.); (M.T.Á.-B.); (M.C.O.-L.)
| | | | - Hilda Montero
- Virus-Cell Interactions Laboratory, Institut Pasteur of Montevideo, Montevideo 11400, Uruguay;
| |
Collapse
|
22
|
Hasan A, Hossain MM, Zamil MF, Trina AT, Hossain MS, Kumkum A, Afreen S, Ahmed D, Rahman M, Alam MS. Concurrent transmission of Zika virus during the 2023 dengue outbreak in Dhaka, Bangladesh. PLoS Negl Trop Dis 2025; 19:e0012866. [PMID: 39883734 PMCID: PMC11813092 DOI: 10.1371/journal.pntd.0012866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/11/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND During the 2023-dengue outbreak in Bangladesh, a diagnostic evaluation study was conducted to investigate concurrent Zika virus (ZIKV) and dengue virus (DENV) transmission in Dhaka in 2023. AIMS The study explored to simultaneously detect the presence of ZIKV, DENV, and/or CHIKV while considering relevant clinical and epidemiological risk factors, using a real-time multiplex RT-PCR system. Following this, it was planned to sequence the selected samples to identify genetic variations of the ZIKV infections within the population. METHODS This study was designed as a diagnostic evaluation, where participants meeting the inclusion criteria were prospectively recruited with written informed consent. A total of 399 febrile individuals were screened, with 185 meeting the inclusion criteria of having a fever onset within 2-5 days, along with one of the following clinical features, e.g. headache, myalgia, arthralgia or bone pain, rash, nausea, vomiting, or diarrhea and 152 undergoing real-time RT-PCR testing. RESULTS Five ZIKV-positive cases were identified, including one DENV-ZIKV co-infection. Phylogenetic analysis revealed the ZIKV strains were part of the Asian lineage, closely related to Cambodian and Chinese strains from 2019. All ZIKV-positive cases were male, residing within a one-kilometer radius, with no prior travel history, suggesting community-level transmission. CONCLUSION This study marks the first identification of ZIKV in Dhaka city and the first report of ZIKV-DENV co-infection in Bangladesh that highlights the diagnostic challenges posed by the symptomatic similarities between ZIKV and other arboviruses and underscores the need for enhanced surveillance and public health interventions to mitigate the spread and impact of ZIKV in dengue-endemic regions.
Collapse
Affiliation(s)
- Anamul Hasan
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mobarok Hossain
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md Fahad Zamil
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Afrida Tabassum Trina
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mohammad Sharif Hossain
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Asifa Kumkum
- Clinical and Diagnostic Services, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Sajia Afreen
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Dilruba Ahmed
- Clinical and Diagnostic Services, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafizur Rahman
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mohammad Shafiul Alam
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
23
|
Zhou F, Xu Y, Liu X, Xu Y, Wang Y, Jiang D, Du P. Zika and Dengue Virus Autoimmunity: An Overview of Related Disorders and Their Potential Mechanisms. Rev Med Virol 2025; 35:e70014. [PMID: 39779915 DOI: 10.1002/rmv.70014] [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: 11/21/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are two major mosquito-borne flaviviruses that pose a significant threat to the global public health system, particularly in tropical regions. The clinical outcomes related to these viral pathogens can vary from self-limiting asymptomatic infections to various forms of life-threatening pathological conditions such as haemorrhagic disorders. In addition to the direct effects of the viral pathogens, immune processes play also a significant function in the development of diseases mediated by ZIKV and DENV. Studing these processes is important for developing safer vaccines and targeted therapeutic strategies. These viruses have been reported to trigger various autoimmune disorders affecting different parts of human organ systems. It also has been shown that preexisting immunity to ZIKV or DENV can impact the outcome of subsequent infections caused by another virus. ZIKV and DENV infection can promote the development of autoimmune disorders by different mechanisms, such as molecular mimicry and autoantibody formation. The present review provides an overview of various autoimmune disorders associated with ZIKV and DENV infection and their potential underlying mechanisms.
Collapse
Affiliation(s)
- Feifan Zhou
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuanze Xu
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xing Liu
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Xu
- Department of Stomach Enterochirurgia, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Wang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Donghui Jiang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Pengfei Du
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
24
|
Fazecas T, Lopes FPPL, Guedes B, Castro P, Nogueira R, Werner H. Zika virus as a new pathogenic agent within the Toxoplasma gondii, Rubella virus, Cytomegalovirus, and Herpes simplex (TORCH) virus family: where do we stand? Pediatr Radiol 2025; 55:65-74. [PMID: 38822063 DOI: 10.1007/s00247-024-05944-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Viral infections in low-income countries such as Brazil pose a significant challenge for medical authorities, with epidemics such as Zika virus infection having lasting effects. The increase in microcephaly among newborns has prompted investigations into the association between Zika virus and this congenital syndrome. The severity and prevalence of microcephaly led to the declaration of national and international emergencies. Extensive research has been conducted to understand the teratogenic effects of Zika virus, particularly its impact on neural progenitor cells in the fetal brain. Various pre- and postnatal imaging techniques, such as ultrasound, magnetic resonance imaging (MRI), and postnatal computed tomography (CT), have played crucial roles in diagnosing and monitoring malformations linked to congenital Zika virus infection in the central nervous system (CNS). These modalities can detect brain parenchymal abnormalities, calcifications, cerebral atrophy, and callosal anomalies. Additionally, three-dimensional ultrasound and fetal MRI provide detailed anatomical images, while CT can identify calcifications that are not easily detected by other methods. Despite advancements in imaging, there are still unanswered questions and ongoing challenges in comprehending the long-term effects and developmental impairments in children affected by Zika virus. Radiologists continue to play a crucial role in diagnosing and assisting in the management of these cases.
Collapse
Affiliation(s)
- Tatiana Fazecas
- Radiology Department, Dasa, Rio de Janeiro, RJ, Brazil.
- Imaging Department, Hospital Municipal Jesus, Rio de Janeiro, RJ, Brazil.
| | | | - Bianca Guedes
- Radiology Department, Dasa, Rio de Janeiro, RJ, Brazil
| | - Pedro Castro
- Radiology Department, Dasa, Rio de Janeiro, RJ, Brazil
| | | | - Heron Werner
- Radiology Department, Dasa, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
25
|
Song BH, Frank JC, Yun SI, Julander JG, Mason JB, Polejaeva IA, Davies CJ, White KL, Dai X, Lee YM. Comparison of Three Chimeric Zika Vaccine Prototypes Developed on the Genetic Background of the Clinically Proven Live-Attenuated Japanese Encephalitis Vaccine SA 14-14-2. Int J Mol Sci 2024; 26:195. [PMID: 39796052 PMCID: PMC11720029 DOI: 10.3390/ijms26010195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Zika virus (ZIKV) is a medically important mosquito-borne orthoflavivirus, but no vaccines are currently available to prevent ZIKV-associated disease. In this study, we compared three recombinant chimeric viruses developed as candidate vaccine prototypes (rJEV/ZIKVMR-766, rJEV/ZIKVP6-740, and rJEV/ZIKVPRVABC-59), in which the two neutralizing antibody-inducing prM and E genes from each of three genetically distinct ZIKV strains were used to replace the corresponding genes of the clinically proven live-attenuated Japanese encephalitis virus vaccine SA14-14-2 (rJEV). In WHO-certified Vero cells (a cell line suitable for vaccine production), rJEV/ZIKVP6-740 exhibited the slowest viral growth, formed the smallest plaques, and displayed a unique protein expression profile with the highest ratio of prM to cleaved M when compared to the other two chimeric viruses, rJEV/ZIKVMR-766 and rJEV/ZIKVPRVABC-59, as well as their vector, rJEV. In IFNAR-/- mice, an animal model of ZIKV infection, subcutaneous inoculation of rJEV/ZIKVP6-740 caused a low-level localized infection limited to the spleen, with no clinical signs of infection, weight loss, or mortality; in contrast, the other two chimeric viruses and their vector caused high-level systemic infections involving multiple organs, consistently leading to clear clinical signs of infection, rapid weight loss, and 100% mortality. Subsequently, subcutaneous immunization with rJEV/ZIKVP6-740 proved highly effective, offering complete protection against a lethal intramuscular ZIKV challenge 28 days after a single-dose immunization. This protection was specific to ZIKV prM/E and likely mediated by neutralizing antibodies targeting ZIKV prM/E. Therefore, our data indicate that the chimeric virus rJEV/ZIKVP6-740 is a highly promising vaccine prototype for developing a safe and effective vaccine for inducing neutralizing antibody-mediated protective immunity against ZIKV.
Collapse
Affiliation(s)
- Byung-Hak Song
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Jordan C. Frank
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Sang-Im Yun
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Justin G. Julander
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Jeffrey B. Mason
- Department of Veterinary Clinical and Life Sciences, College of Veterinary Medicine, Center for Integrated BioSystems, Utah State University, Logan, UT 84322, USA;
| | - Irina A. Polejaeva
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Christopher J. Davies
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Kenneth L. White
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Xin Dai
- Utah Agricultural Experiment Station, Utah State University, Logan, UT 84322, USA;
| | - Young-Min Lee
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| |
Collapse
|
26
|
Nkolola JP, Hope D, Guan R, Colarusso A, Aid M, Weiss D, Misamore J, Andersen H, Lewis MG, Williamson L, Carnahan RH, Crowe JE, Barouch DH. Protective threshold of a potent neutralizing Zika virus monoclonal antibody in rhesus macaques. J Virol 2024; 98:e0142924. [PMID: 39545728 DOI: 10.1128/jvi.01429-24] [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: 08/14/2024] [Accepted: 09/28/2024] [Indexed: 11/17/2024] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that caused a global pandemic in 2016-2017 with continued ongoing transmission at low levels in several countries. In the absence of an approved ZIKV vaccine, neutralizing monoclonal antibodies (mAbs) provide an option for the prevention and treatment of ZIKV infection. Previous studies identified a potent neutralizing human mAb ZIKV-117 that reduced fetal infection and death in mice following ZIKV challenge. In this study, we report exquisite potency of ZIKV-117-LALA-YTE, which has been engineered to reduce Fc receptor binding and to extend half-life, in a titration study in rhesus macaques to protect against ZIKV challenge. We show complete protection at a dose of 0.016 mg/kg ZIKV-117-LALA-YTE, which resulted in median serum concentrations of 0.13 µg/mL. The high potency of this mAb supports its potential clinical development as a novel biotherapeutic intervention for ZIKV.IMPORTANCEIn this study, we report the potency of the Zika virus (ZIKV)-specific neutralizing antibody ZIKV-117-LALA-YTE against ZIKV challenge in a titration study rhesus macaques. This high potency supports the further development of this monoclonal antibody for ZIKV.
Collapse
Affiliation(s)
- Joseph P Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - David Hope
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Ruoran Guan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Alessandro Colarusso
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | | | | | | - Lauren Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Turner EA, Clark SD, Peña-García VH, Christofferson RC. Investigating the Effects of Microclimate on Arboviral Kinetics in Aedes aegypti. Pathogens 2024; 13:1105. [PMID: 39770364 PMCID: PMC11728849 DOI: 10.3390/pathogens13121105] [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: 11/11/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Aedes aegypti are indoor-dwelling vectors of many arboviruses, including Zika (ZIKV) and chikungunya (CHIKV). The dynamics of these viruses within the mosquito are known to be temperature-dependent, and models that address risk and predictions of the transmission efficiency and patterns typically use meteorological temperature data. These data do not differentiate the temperatures experienced by mosquitoes in different microclimates, such as indoor vs. outdoor. Using temperature data collected from Neiva Colombia, we investigated the impact of two microclimate temperature profiles on ZIKV and CHIKV infection dynamics in Ae. aegypti. We found that the vector mortality was not significantly impacted by the difference in temperature profiles. Further, we found that the infection and dissemination rates were largely unaffected, with only ZIKV experiencing a significant increase in infection at outdoor temperatures at 21 days post-infection (dpi). Further, there was a significant increase in viral titers in the abdomens of ZIKV-infected mosquitoes at 21 dpi. With CHIKV, there was a significant titer difference in the abdomens of mosquitoes at both 7 and 14 dpi. While there were differences in vector infection kinetics that were not statistically significant, we developed a simple stochastic SEIR-SEI model to determine if the observed differences might translate to notable differences in simulated outbreaks. With ZIKV, while the probability of secondary transmission was high (>90%) under both microenvironmental scenarios, there was often only one secondary case. However, CHIKV differences between microenvironments were more prominent. With over 90% probability of secondary transmission, at indoor conditions, the peak of transmission was higher (over 850 cases) compared to the outdoor conditions (<350 cases). Further, the time-to-peak for indoor was 130 days compared to 217 days for outdoor scenarios. Further investigations into microenvironmental conditions, including temperature, may be key to increasing our understanding of the nuances of CHIKV and ZIKV vectorial capacity, epidemiology, and risk assessment, especially as it affects other aspects of transmission, such as biting rate. Overall, it is critical to understand the variability of how extrinsic factors affect transmission systems, and these data add to the growing catalog of knowledge of how temperature affects arboviral systems.
Collapse
Affiliation(s)
- Erik A Turner
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Samantha D Clark
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Víctor Hugo Peña-García
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
28
|
Nelson AN, Ploss A. Emerging mosquito-borne flaviviruses. mBio 2024; 15:e0294624. [PMID: 39480108 PMCID: PMC11633211 DOI: 10.1128/mbio.02946-24] [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] [Indexed: 11/02/2024] Open
Abstract
Flaviviruses comprise a genus of enveloped, positive-sense, single-stranded RNA viruses typically transmitted between susceptible and permissive hosts by arthropod vectors. Established flavivirus threats include dengue viruses (DENV), yellow fever virus (YFV), Zika virus (ZIKV), and West Nile virus (WNV), which continue to cause over 400 million infections annually and are significant global health and economic burdens. Additionally, numerous closely related but largely understudied viruses circulate in animals and can conceivably emerge in human populations. Previous flaviviruses that were recognized to have this potential include ZIKV and WNV, which only became extensively studied after causing major outbreaks in humans. More than 50 species exist within the flavivirus genus, which can be further classified as mosquito-borne, tick-borne, insect-specific, or with no known vector. Historically, many of these flaviviruses originated in Africa and have mainly affected tropical and subtropical regions due to the ecological niche of mosquitoes. However, climate change, as well as vector and host migration, has contributed to geographical expansion, thereby posing a potential risk to global populations. For the purposes of this minireview, we focus on the mosquito-borne subgroup and highlight viruses that cause significant pathology or lethality in at least one animal species and/or have demonstrated an ability to infect humans. We discuss current knowledge of these viruses, existing animal models to study their pathogenesis, and potential future directions. Emerging viruses discussed include Usutu virus (USUV), Wesselsbron virus (WSLV), Spondweni virus (SPOV), Ilheus virus (ILHV), Rocio virus (ROCV), Murray Valley encephalitis virus (MVEV), and Alfuy virus (ALFV).
Collapse
Affiliation(s)
- Amy N. Nelson
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
29
|
Zadra N, Rizzoli A, Rota-Stabelli O. Comprehensive phylogenomic analysis of Zika virus: Insights into its origin, past evolutionary dynamics, and global spread. Virus Res 2024; 350:199490. [PMID: 39489463 PMCID: PMC11583807 DOI: 10.1016/j.virusres.2024.199490] [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: 09/27/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Zika virus (ZIKV), a Flaviviridae family member, has been linked to severe neurological disorders. Despite detailed studies on recent outbreaks, the early evolutionary history of ZIKV remains partially unclear. This study elucidates ZIKV origin and evolutionary dynamics, focusing on recombination events, early lineage diversification, and virus spread across continents. METHODS We assessed recombination using multiple methods. We conducted Bayesian phylogenetic analyses to understand the evolutionary relationships and timing of key diversification events. Model selection was carried out to determine the most appropriate evolutionary model for our dataset. RESULTS Our phylogenies revealed recent recombination between Singaporean and African lineages, indicating the co-circulation of diverse lineages during outbreaks. Thailand was identified as a crucial hub in the spread across Asia. The phylogenetic analysis suggests that the ZIKV lineage dates back to the eleventh century, with the first significant diversification occurring in the nineteenth century. The timing of the re-introduction of the Asian lineage into Africa and the delay between probable introduction and outbreak onset were also determined. CONCLUSIONS This study provides novel insights into ZIKV's origin and early evolutionary dynamics, highlighting Thailand's role in the spread of the virus in Asia and recent recombination events between distant lineages. These findings emphasize the need for continuous surveillance and a better understanding of ZIKV biology to forecast and mitigate future outbreaks.
Collapse
Affiliation(s)
- Nicola Zadra
- Conservation Genomics Research Unit, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Trento, Italy; NBFC, National Biodiversity Future Center, Palermo 90133, Italy.
| | - Annapaola Rizzoli
- Applied Ecology Research Unit, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Trento, Italy; NBFC, National Biodiversity Future Center, Palermo 90133, Italy
| | - Omar Rota-Stabelli
- Center Agriculture Food Environment (C3A), University of Trento, 38010, San Michele all'Adige, TN, Italy
| |
Collapse
|
30
|
Cagigi A, Tinnirello R, Iannolo G, Douradinha B. Orthoflavivirus zikaense (Zika) vaccines: What are we waiting for? Int J Antimicrob Agents 2024; 64:107367. [PMID: 39490448 DOI: 10.1016/j.ijantimicag.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Alberto Cagigi
- International Vaccine Institute (IVI) Europe Regional Office, Solna, Sweden
| | | | | | - Bruno Douradinha
- Vaccine Technology Subgroup, Emerging Pathogens Group, Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
31
|
Hassaan NA, Xing L. The mechanisms of Zika virus-induced neuropathogenesis. Braz J Microbiol 2024; 55:3933-3943. [PMID: 39422868 PMCID: PMC11711583 DOI: 10.1007/s42770-024-01543-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024] Open
Abstract
Zika virus (ZIKV), a flavivirus, is one of the most serious re-emerging pathogens. Growing outbreaks in the Americas have linked ZIKV to significant clinical symptoms including Guillain-Barré syndrome in adults and congenital anomalies in newborns. ZIKV affects brain cells in a variety of ways, mostly apoptosis and cell cycle delays. Modulation of the host's immune reaction and the inflammatory process has also been shown to be involved in ZIKV-induced neurological disorders. This review summarized and discussed the latest advances in ZIKV research to shed fresh light on the multiple mechanisms incolved in ZIKV-induced neuropathogenesis.
Collapse
Affiliation(s)
- Nahla Ahmed Hassaan
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, Shanxi province, 030006, China
| | - Li Xing
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, Shanxi province, 030006, China.
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, China.
| |
Collapse
|
32
|
Wu Z, He Y, Wang T, Wang M, Cheng A, Chen S. DENV and ZIKV infection: Species specificity and broad cell tropism. Virology 2024; 600:110276. [PMID: 39467358 DOI: 10.1016/j.virol.2024.110276] [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: 09/06/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Nearly one-third of countries worldwide have reported cases of Dengue virus (DENV) and Zika virus (ZIKV) infections, highlighting the significant threat these viruses pose to global public health. As members of the Flavivirus genus within the Flaviviridae family, DENV and ZIKV have demonstrated the ability to infect a wide range of cell lines from multiple species in vitro. However, the range of susceptible animal models is notably limited, and field studies indicate that their capacity to infect host organisms is highly restricted, with a very narrow range of target cells in vivo. The virus's ability to hijack host cellular machinery plays a crucial role in determining its cellular and species specificity. In this review, we examine how DENV and ZIKV exploit host cells to facilitate their replication, offering new insights that could inform the development of antiviral drugs and therapeutic targets.
Collapse
Affiliation(s)
- Zhen Wu
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Agricultural Bioinformatics of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yu He
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Agricultural Bioinformatics of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Tao Wang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Mingshu Wang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Anchun Cheng
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Shun Chen
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Agricultural Bioinformatics of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
33
|
Andrade P, Sosa-Moreno A, Vivero S, Nipaz V, Lee GO, Cevallos W, Eisenberg JNS, Coloma J. The Impact of Zika Emergence in Remote Communities in Northwestern Ecuador. J Infect Dis 2024; 230:e1058-e1066. [PMID: 39082780 PMCID: PMC11566036 DOI: 10.1093/infdis/jiae384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/29/2024] [Indexed: 11/16/2024] Open
Abstract
The Zika virus (ZIKV) epidemic in Latin America (2015-2016) has primarily been studied in urban centers, with less understanding of its impact on smaller rural communities. To address this gap, we analyzed ZIKV seroepidemiology in 6 rural Ecuadorian communities (2018-2019) with varying access to a commercial hub. Seroprevalence ranged from 19% to 54%, measured by nonstructural protein 1 blockade of binding enzyme-linked immunosorbent assay. We observed a decline in ZIKV seroprevalence between 2018 and 2019 that was greater among younger populations, suggesting that the attack rates in the 2015-2016 epidemic were significantly higher than our 2018 observations. These data indicate that the 2015-2016 epidemic included significant transmission in rural and more remote settings. Our observations of high seroprevalence in our area of study highlights the importance of surveillance and research in rural areas lacking robust health systems to manage future Zika outbreaks and vaccine initiatives.
Collapse
Affiliation(s)
- Paulina Andrade
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, USA
- Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito, Quito, Ecuador
| | - Andrea Sosa-Moreno
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Sandra Vivero
- Centro de Biomedicina, Universidad Central, Quito, Ecuador
| | - Victoria Nipaz
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| | - Gwenyth O Lee
- Global Health Institute, Rutgers University, New Brunswick, New Jersey, USA
| | | | - Joseph N S Eisenberg
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Josefina Coloma
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, USA
| |
Collapse
|
34
|
Al Beloushi M, Saleh H, Ahmed B, Konje JC. Congenital and Perinatal Viral Infections: Consequences for the Mother and Fetus. Viruses 2024; 16:1698. [PMID: 39599813 PMCID: PMC11599085 DOI: 10.3390/v16111698] [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: 06/05/2024] [Revised: 10/13/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Viruses are the most common congenital infections in humans and an important cause of foetal malformations, neonatal morbidity, and mortality. The effects of these infections, which are transmitted in utero (transplacentally), during childbirth or in the puerperium depend on the timing of the infections. These vary from miscarriages (usually with infections in very early pregnancy), congenital malformations (when the infections occur during organogenesis) and morbidity (with infections occurring late in pregnancy, during childbirth or after delivery). The most common of these viruses are cytomegalovirus, hepatitis, herpes simplex type-2, parvovirus B19, rubella, varicella zoster and zika viruses. There are currently very few efficacious antiviral agents licensed for use in pregnancy. For most of these infections, therefore, prevention is mainly by vaccination (where there is a vaccine). The administration of immunoglobulins to those exposed to the virus to offer passive immunity or appropriate measures to avoid being infected would be options to minimise the infections and their consequences. In this review, we discuss some of the congenital and perinatal infections and their consequences on both the mother and fetus and their management focusing mainly on prevention.
Collapse
Affiliation(s)
- Mariam Al Beloushi
- Women’s Wellness and Research Centre Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (M.A.B.); (H.S.)
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Huda Saleh
- Women’s Wellness and Research Centre Hamad Medical Corporation, Doha P.O. Box 3050, Qatar; (M.A.B.); (H.S.)
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Badreldeen Ahmed
- Department of Obstetrics and Gynaecology, Qatar University, Doha P.O. Box 2713, Qatar;
- Feto Maternal Centre, Al Markhiya Doha, Doha P.O. Box 34181, Qatar
- Department of Obstetrics and Gynaecology Weill Cornell Medicine, Doha P.O. Box 24144, Qatar
| | - Justin C. Konje
- Feto Maternal Centre, Al Markhiya Doha, Doha P.O. Box 34181, Qatar
- Department of Obstetrics and Gynaecology Weill Cornell Medicine, Doha P.O. Box 24144, Qatar
- Department of Health Sciences, University of Leicester, P.O. Box 7717, Leicester LE2 7LX, UK
| |
Collapse
|
35
|
Tajik S, Farahani AV, Ardekani OS, Seyedi S, Tayebi Z, Kami M, Aghaei F, Hosseini TM, Nia MMK, Soheili R, Letafati A. Zika virus tropism and pathogenesis: understanding clinical impacts and transmission dynamics. Virol J 2024; 21:271. [PMID: 39472938 PMCID: PMC11523830 DOI: 10.1186/s12985-024-02547-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
The Zika virus (ZIKV) is classified within the Flavivirus genus of the Flaviviridae family and is categorized as an arbovirus. The virus was initially identified in a rhesus monkey in Uganda in 1947 and later in a human in Nigeria in 1952. Since 2007, the prevalence of the virus has been on the rise, culminating in a major outbreak in the United States (US) in 2015. During this outbreak, the adult population was severely impacted, experiencing a range of symptoms, including organ failure, microcephaly, fetal death, and Guillain-Barré syndrome (GBS). Additionally, skin rash, limb swelling, fever, headache, and heightened sensitivity are found in most adults with Zika syndrome. Although the virus can be transmitted through blood, vertical transmission from mother to child, and sexual contact, the primary way of transmission of the virus is through the Aedes mosquito. Cells such as neurons, macrophages, peripheral dendritic cells, and placental cells are among the target cells that the virus can infect. The TAM AXL receptor plays a crucial role in infection. After the virus enters the body through the bloodstream, it spreads in the body with a latent period of 3 to 12 days. Currently, there is no specific treatment or publicly available vaccine for the ZIKV. Limited laboratory testing has been conducted, and existing drugs originally designed for other pathogens have been repurposed for treatment. Given the Aedes mosquito's role as a vector and the wide geographical impact of the virus, this study aims to comprehensively investigate Zika's pathogenesis and clinical symptoms based on existing knowledge and research. By doing so, we seek to enhance our understanding of the virus and inform future prevention and treatment strategies.
Collapse
Affiliation(s)
- Saeed Tajik
- Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Vasheghani Farahani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Saba Seyedi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Zahra Tayebi
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Department of Microbiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mostafa Kami
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Department of Pathology, Faculty of Veterinary Medicine, Babol Branch, Islamic Azad University, Babol, Iran
| | - Faezeh Aghaei
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | | | - Mohammad Mahdi Khosravi Nia
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Student Research Committee, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Roben Soheili
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Arash Letafati
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran.
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Badu P, Baniulyte G, Sammons MA, Pager CT. Activation of ATF3 via the integrated stress response pathway regulates innate immune response to restrict Zika virus. J Virol 2024; 98:e0105524. [PMID: 39212382 PMCID: PMC11494902 DOI: 10.1128/jvi.01055-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Zika virus (ZIKV) is a re-emerging mosquito-borne flavivirus that can have devastating health consequences. The developmental and neurological effects of a ZIKV infection arise in part from the virus triggering cellular stress pathways and perturbing transcriptional programs. To date, the underlying mechanisms of transcriptional control directing viral restriction and virus-host interaction are understudied. Activating Transcription Factor 3 (ATF3) is a stress-induced transcriptional effector that modulates the expression of genes involved in a myriad of cellular processes, including inflammation and antiviral responses, to restore cellular homeostasis. While ATF3 is known to be upregulated during ZIKV infection, the mode by which ATF3 is activated, and the specific role of ATF3 during ZIKV infection is unknown. In this study, we show via inhibitor and RNA interference approaches that ZIKV infection initiates the integrated stress response pathway to activate ATF4 which in turn induces ATF3 expression. Additionally, by using CRISPR-Cas9 system to delete ATF3, we found that ATF3 acts to limit ZIKV gene expression in A549 cells. We also determined that ATF3 enhances the expression of antiviral genes such as STAT1 and other components in the innate immunity pathway to induce an ATF3-dependent anti-ZIKV response. Our study reveals crosstalk between the integrated stress response and innate immune response pathways and highlights an important role for ATF3 in establishing an antiviral effect during ZIKV infection. IMPORTANCE Zika virus (ZIKV) is a re-emerging mosquito-borne flavivirus that co-opts cellular mechanisms to support viral processes that can reprogram the host transcriptional profile. Such viral-directed transcriptional changes and the pro- or anti-viral outcomes remain understudied. We previously showed that ATF3, a stress-induced transcription factor, is significantly upregulated in ZIKV-infected mammalian cells, along with other cellular and immune response genes. We now define the intracellular pathway responsible for ATF3 activation and elucidate the impact of ATF3 expression on ZIKV infection. We show that during ZIKV infection, the integrated stress response pathway stimulates ATF3 which enhances the innate immune response to antagonize ZIKV infection. This study establishes a link between viral-induced stress response and transcriptional regulation of host defense pathways and thus expands our knowledge of virus-mediated transcriptional mechanisms and transcriptional control of interferon-stimulated genes during ZIKV infection.
Collapse
Affiliation(s)
- Pheonah Badu
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
| | - Gabriele Baniulyte
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
| | - Morgan A. Sammons
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
| | - Cara T. Pager
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, New York, USA
| |
Collapse
|
37
|
Taufer NP, Santos-Souza C, Larentis LT, Santos CND, Creuzet SE, Garcez RC. Integrative analysis of molecular pathways and morphological anomalies associated with congenital Zika syndrome. J Neurol Sci 2024; 465:123190. [PMID: 39182423 DOI: 10.1016/j.jns.2024.123190] [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: 06/05/2024] [Revised: 08/09/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Congenital Zika syndrome (CZS) comprises a set of clinical manifestations that can be presented by neonates born to mothers infected by the Zika virus (ZIKV). CZS-associated phenotypes include neurological, skeletal, and systemic alterations and long-term developmental sequelae. One of the most frequently reported clinical conditions is microcephaly characterized by a reduction in head circumference and cognitive complications. Nevertheless, the associations among the diverse signaling pathways underlying CZS phenotypes remain to be elucidated. To shed light on CZS, we have extensively reviewed the morphological anomalies resulting from ZIKV infection, as well as genes and proteins of interest obtained from the published literature. With this list of genes or proteins, we performed computational analyses to explore the cellular processes, molecular mechanisms, and molecular pathways related to ZIKV infection. Therefore, in this review, we comprehensively describe the morphological abnormalities caused by congenital ZIKV infection and, through the analysis noted above, propose common molecular pathways altered by ZIKV that could explain both central nervous system and craniofacial skeletal alterations.
Collapse
Affiliation(s)
- Nathali Parise Taufer
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Camila Santos-Souza
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Lucas Trentin Larentis
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Sophie Emmanuelle Creuzet
- Institut des Neurosciences Paris-Saclay (NeuroPSI), Paris-Saclay University, Centre National de la Recherche Scientifique UMR 9197, Saclay, France.
| | - Ricardo Castilho Garcez
- Graduate Program in Cell and Developmental Biology, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Laboratório de Células-Tronco e Regeneração Tecidual (LACERT), Department of Cell Biology, Embryology, and Genetics, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
38
|
Roth C, Pitard B, Levillayer L, Lay S, Vo HTM, Cantaert T, Sakuntabhai A. Zika virus T-cell based 704/DNA vaccine promotes protection from Zika virus infection in the absence of neutralizing antibodies. PLoS Negl Trop Dis 2024; 18:e0012601. [PMID: 39418312 PMCID: PMC11521268 DOI: 10.1371/journal.pntd.0012601] [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: 03/26/2024] [Revised: 10/29/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are closely related flaviviruses co-circulating in the same endemic areas. Infection can raise cross-reactive antibodies that can be either protective or increase risk of severe disease, depending on the infection sequence, DENV serotype and elapsed time between infection. On the contrast, T cell-mediated immunity against DENV and ZIKV is considered protective. Therefore, we have developed a T cell vaccine enriched in immunodominant T cell epitopes derived from ZIKV and evaluated its immunogenicity and efficacy against ZIKV and DENV infection. Mice were vaccinated using DNA vaccine platform using the tetrafunctional amphiphilic block copolymer 704. We show that vaccination of 2 different HLA class I transgenic mice with the ZIKV non-structural (NS) poly-epitope elicits T cell response against numerous ZIKV epitopes. Moreover, vaccination induces a significant protection against ZIKV infection, in the absence of neutralizing or enhancing antibodies against ZIKV. However, vaccination does not induce a significant protection against DENV2. In contrast, immunization with a DENV1-NS poly-epitope induces a significant protection against both DENV1 and DENV2, in the absence of humoral immunity. Taken together, we have shown that T-cell based vaccination could protect against multiple flavivirus infections and could overcome the complexity of antibody-mediated enhancement.
Collapse
Affiliation(s)
- Claude Roth
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| | - Bruno Pitard
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in Immunotherapy, INCIT UMR1232/EMR6001, F-44000 Nantes, France
| | - Laurine Levillayer
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- Oxford University Clinical Research Unit, Ho Chi Minh, Vietnam
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Anavaj Sakuntabhai
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| |
Collapse
|
39
|
Porier DL, Adam A, Kang L, Michalak P, Tupik J, Santos MA, Tanelus M, López K, Auguste DI, Lee C, Allen IC, Wang T, Auguste AJ. Humoral and T-cell-mediated responses to an insect-specific flavivirus-based Zika virus vaccine candidate. PLoS Pathog 2024; 20:e1012566. [PMID: 39388457 PMCID: PMC11495591 DOI: 10.1371/journal.ppat.1012566] [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: 12/13/2023] [Revised: 10/22/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
Flaviviruses represent a significant global health threat and relatively few licensed vaccines exist to protect against them. Insect-specific flaviviruses (ISFVs) are incapable of replication in humans and have emerged as a novel and promising tool for flavivirus vaccine development. ISFV-based flavivirus vaccines have shown exceptional safety, immunogenicity, and efficacy, however, a detailed assessment of the correlates of protection and immune responses induced by these vaccines are still needed for vaccine optimization. Here, we explore the mechanisms of protective immunity induced by a previously created pre-clinical Zika virus (ZIKV) vaccine candidate, called Aripo/Zika (ARPV/ZIKV). In brief, immunocompromised IFN-αβR-/- mice passively immunized with ARPV/ZIKV immune sera experienced protection after lethal ZIKV challenge, although this protection was incomplete. ARPV/ZIKV-vaccinated IFN-αβR-/- mice depleted of CD4+ or CD8+ T-cells at the time of ZIKV challenge showed no morbidity or mortality. However, the adoptive transfer of ARPV/ZIKV-primed T-cells into recipient IFN-αβR-/- mice resulted in a two-day median increase in survival time compared to controls. Altogether, these results suggest that ARPV/ZIKV-induced protection is primarily mediated by neutralizing antibodies at the time of challenge and that T-cells may play a comparatively minor but cumulative role in the protection observed. Lastly, ARPV/ZIKV-vaccinated Tcra KO mice, which are deficient in T-cell responses, experienced significant mortality post-challenge. These results suggest that ARPV/ZIKV-induced cell-mediated responses are critical for development of protective immune responses at vaccination. Despite the strong focus on neutralizing antibody responses to novel flavivirus vaccine candidates, these results suggest that cell-mediated responses induced by ISFV-based vaccines remain important to overall protective responses.
Collapse
Affiliation(s)
- Danielle L. Porier
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lin Kang
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, United States of America
| | - Pawel Michalak
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Juselyn Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
| | - Matthew A. Santos
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Manette Tanelus
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Krisangel López
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Dawn I. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Christy Lee
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Albert J. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| |
Collapse
|
40
|
Blyden K, Thomas J, Emami-Naeini P, Fashina T, Conrady CD, Albini TA, Carag J, Yeh S. Emerging Infectious Diseases and the Eye: Ophthalmic Manifestations, Pathogenesis, and One Health Perspectives. Int Ophthalmol Clin 2024; 64:39-54. [PMID: 39480207 PMCID: PMC11512616 DOI: 10.1097/iio.0000000000000539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infectious diseases may lead to ocular complications including uveitis, an ocular inflammatory condition with potentially sight-threatening sequelae, and conjunctivitis, inflammation of the conjunctiva. Emerging infectious pathogens with known ocular findings include Ebola virus, Zika virus, Avian influenza virus, Nipah virus, severe acute respiratory syndrome coronaviruses, and Dengue virus. Re-emerging pathogens with ocular findings include Toxoplasma gondii and Plasmodium species that lead to malaria. The concept of One Health involves a collaborative and interdisciplinary approach to achieve optimal health outcomes by combining human, animal, and environmental health factors. This approach examines the interconnected and often complex human-pathogen-intermediate host interactions in infectious diseases that may also result in ocular disease, including uveitis and conjunctivitis. Through a comprehensive review of the literature, we review the ophthalmic findings of emerging infectious diseases, pathogenesis, and One Health perspectives that provide further insight into the disease state. While eye care providers and vision researchers may often focus on key local aspects of disease process and management, additional perspective on host-pathogen-reservoir life cycles and transmission considerations, including environmental factors, may offer greater insight to improve outcomes for affected individuals and stakeholders.
Collapse
Affiliation(s)
- K’Mani Blyden
- Medical College of Georgia, Augusta University, Augusta, GA
| | - Joanne Thomas
- Emory Eye Center, Emory University School of Medicine, Atlanta, GA
- Emory University School of Medicine, Atlanta, GA
| | - Parisa Emami-Naeini
- Department of Ophthalmology, University of California, Davis, Sacramento, CA
| | - Tolulope Fashina
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE
| | - Christopher D. Conrady
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Thomas A. Albini
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | | | - Steven Yeh
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
41
|
Haddad H, Tangy F, Ouahchi I, Sahtout W, Ouni B, Zaïri A. Evaluation of the antiviral activity of new dermaseptin analogs against Zika virus. Biochem Biophys Rep 2024; 39:101747. [PMID: 38939125 PMCID: PMC11208914 DOI: 10.1016/j.bbrep.2024.101747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/29/2024] Open
Abstract
Zika virus represents the primary cause of infection during pregnancy and can lead to various neurological disorders such as microcephaly and Guillain-Barré syndrome affecting both children and adults. This infection is also associated with urological and nephrological problems. So far, evidence of mosquito-borne Zika virus infection has been reported in a total of 89 countries and territories. However, surveillance efforts primarily concentrate on outbreaks that this virus can cause, yet the measures implemented are typically limited. Currently, there are no specific treatments or vaccines designed for the prevention or treatment of Zika virus infection or its associated disease. The development of effective therapeutic agents presents an urgent need. Importantly, an alternative for advancing the discovery of new molecules could be dermaseptins, a family of antimicrobial peptides known for their potential antiviral properties. In this study, we carried out the synthesis of dermaseptins and their analogs and subsequently assessed the bioactivity tests against Zika virus (ZIKV PF13) of dermaseptins B2 and S4 and their derivatives. The cytotoxicity of these peptides was investigated on HMC3 cell line and HeLa cells by CellTiter-Glo® Luminescent Cell Viability Assay. Thereafter, we evaluated the antiviral activity caused by the action of our dermaseptins on the viral envelope using the Fluorescence Activated Cell Sorting (FACS). The cytotoxicity of our molecules was concentration-dependent at microgram concentrations Expect for dermaseptin B2 and its derivative which present no toxicity against HeLa and HMC3 cell lines. It was observed that all tested analogs from S4 family exhibited antiviral activity with low concentrations ranging from 3 to 12.5 μg/ml , unlike the native B2 and its derivative which increased the infectivity. Pre-incubating of dermaseptins with ZIKV PF13 before infection revealed that these derivatives inhibit the initial stages of virus infection. In summary, these results suggest that dermaseptins could serve as novel lead structures for the development of potent antiviral agents against Zika virus infections.
Collapse
Affiliation(s)
- Houda Haddad
- BIOLIVAL Laboratory, LR14ES06, The Higher Institute of Biotechnology of Monastir ISBM, University of Monastir, Monastir, 5000, Tunisia
- Biochemistry Department, Faculty of Medicine, University of Sousse, Sousse, 4002, Tunisia
| | - Frédéric Tangy
- Institut Pasteur, Université Paris Cité, Vaccines-innovation Laboratory, 75015, Paris, France
| | - Ines Ouahchi
- Cytogenetics and Reproductive Biology department, Farhat Hached University Teaching Hospital, University of Sousse, 4000, Sousse, Tunisia
| | - Wissal Sahtout
- Nephrology Department, Sahloul University Hospital, University of Sousse, 4054, Sousse, Tunisia
- Research Laboratory LR12SP11, Biochemistry Department, Sahloul University Hospital, University of Sousse, 4054, Sousse, Tunisia
| | - Bouraoui Ouni
- Pharmacology Department, Faculty of Medicine, University of Sousse, 4002, ousse, Tunisia
| | - Amira Zaïri
- Biochemistry Department, Faculty of Medicine, University of Sousse, Sousse, 4002, Tunisia
| |
Collapse
|
42
|
Roy A, Liu Q, Yang Y, Debnath AK, Du L. Envelope Protein-Targeting Zika Virus Entry Inhibitors. Int J Mol Sci 2024; 25:9424. [PMID: 39273370 PMCID: PMC11394925 DOI: 10.3390/ijms25179424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Zika virus (ZIKV; family, Flaviviridae), which causes congenital Zika syndrome, Guillain-Barré Syndrome, and other severe diseases, is transmitted mainly by mosquitoes; however, the virus can be transmitted through other routes. Among the three structural and seven nonstructural proteins, the surface envelope (E) protein of ZIKV plays a critical role in viral entry and pathogenesis, making it a key target for the development of effective entry inhibitors. This review article describes the life cycle, genome, and encoded proteins of ZIKV, illustrates the structure and function of the ZIKV E protein, summarizes E protein-targeting entry inhibitors (with a focus on those based on natural products and small molecules), and highlights challenges that may potentially hinder the development of effective inhibitors of ZIKV infection. Overall, the article will provide useful guidance for further development of safe and potent ZIKV entry inhibitors targeting the viral E protein.
Collapse
Affiliation(s)
- Abhijeet Roy
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Qian Liu
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Asim K. Debnath
- Lindsey F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
43
|
Girmay BS, Ayele SA, Abbas SA, Jang SS, Jung E, Shin JS, Han SB, Kim H. Discovery and structure-activity relationship study of novel isoxazole-based small molecules targeting Zika virus infections. RSC Med Chem 2024; 15:2792-2805. [PMID: 39157190 PMCID: PMC11325340 DOI: 10.1039/d4md00240g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/24/2024] [Indexed: 08/20/2024] Open
Abstract
The Zika virus (ZIKV), a significant public health threat, is transmitted by Aedes aegypti mosquitoes and is associated with severe neurological disorders, particularly in newborns. Currently, there are no approved vaccines or specific therapeutics for ZIKV. Our study focuses on the identification and optimization of isoxazole-based small molecules, specifically through the structural modification of KR-26827, to combat ZIKV infections. Among the synthesized derivatives, 7l emerged as the most promising candidate, showing potent antiviral activity against ZIKV strains and an improved safety profile in vitro. This research underlines the potential of 7l for further development as a ZIKV therapeutic agent.
Collapse
Affiliation(s)
- Berehe Solomon Girmay
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology Daejeon 34113 Republic of Korea
| | - Sileshi Abera Ayele
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology Daejeon 34113 Republic of Korea
| | - Syed Azeem Abbas
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology Daejeon 34113 Republic of Korea
| | - Su San Jang
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
| | - Eunhye Jung
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
| | - Soo Bong Han
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology Daejeon 34113 Republic of Korea
| | - Hyejin Kim
- Infectious Diseases Therapeutic Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology Daejeon 34114 Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology Daejeon 34113 Republic of Korea
- School of Pharmacy, Sungkyunkwan University Suwon 16419 Republic of Korea
| |
Collapse
|
44
|
Omme S, Wang J, Sifuna M, Rodriguez J, Owusu NR, Goli M, Jiang P, Waziha P, Nwaiwu J, Brelsfoard CL, Vigneron A, Ciota AT, Kramer LD, Mechref Y, Onyangos MG. Multi-omics analysis of antiviral interactions of Elizabethkingia anophelis and Zika virus. Sci Rep 2024; 14:18470. [PMID: 39122799 PMCID: PMC11315927 DOI: 10.1038/s41598-024-68898-3] [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: 02/13/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
The microbial communities residing in the mosquito midgut play a key role in determining the outcome of mosquito pathogen infection. Elizabethkingia anophelis, originally isolated from the midgut of Anopheles gambiae possess a broad-spectrum antiviral phenotype, yet a gap in knowledge regarding the mechanistic basis of its interaction with viruses exists. The current study aims to identify pathways and genetic factors linked to E. anophelis antiviral activity. The understanding of E. anophelis antiviral mechanism could lead to novel transmission barrier tools to prevent arboviral outbreaks. We utilized a non-targeted multi-omics approach, analyzing extracellular lipids, proteins, metabolites of culture supernatants coinfected with ZIKV and E. anophelis. We observed a significant decrease in arginine and phenylalanine levels, metabolites that are essential for viral replication and progression of viral infection. This study provides insights into the molecular basis of E. anophelis antiviral phenotype. The findings lay a foundation for in-depth mechanistic studies.
Collapse
Affiliation(s)
- S Omme
- Department of Biological Sciences, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - J Wang
- Department of Biochemistry and Chemistry, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - M Sifuna
- Department of Biological Sciences, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - J Rodriguez
- Department of Biological Sciences, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - N R Owusu
- Department of Biological Sciences, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - M Goli
- Department of Biochemistry and Chemistry, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - P Jiang
- Department of Biochemistry and Chemistry, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - P Waziha
- Department of Biochemistry and Chemistry, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - J Nwaiwu
- Department of Biochemistry and Chemistry, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - C L Brelsfoard
- Department of Biological Sciences, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - A Vigneron
- Laboratoire d'Ecologie Microbienne, Claude Bernard University Lyon, University of Lyon, Lyon, France
| | - A T Ciota
- Wadsworth Centre, New York State Department of Health, Griffin Laboratory, 5668 State Farm Road, Slingerlands, NY, 12159, USA
- School of Public Health, State University of New York Albany, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - L D Kramer
- School of Public Health, State University of New York Albany, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Y Mechref
- Department of Biochemistry and Chemistry, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA
| | - M G Onyangos
- Department of Biological Sciences, Texas Tech University, 2901 Main St, Lubbock, TX, 79409, USA.
| |
Collapse
|
45
|
Zhao C, Chen J, Liu Z, Liang H, Chen X, Cheng L, Xie S, Lin Z, Wu R, Zhao Q, Xue Y, Lai X, Jin X, Xu JF, Su X. Activation of nicotinic acetylcholine receptor α7 subunit limits Zika viral infection via promoting autophagy and ferroptosis. Mol Ther 2024; 32:2641-2661. [PMID: 38822526 PMCID: PMC11405161 DOI: 10.1016/j.ymthe.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/13/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024] Open
Abstract
Vagus nerve regulates viral infection and inflammation via the alpha 7 nicotinic acetylcholine receptor (α7 nAChR); however, the role of α7 nAChR in ZIKA virus (ZIKV) infection, which can cause severe neurological diseases such as microcephaly and Guillain-Barré syndrome, remains unknown. Here, we first examined the role of α7 nAChR in ZIKV infection in vitro. A broad effect of α7 nAChR activation was identified in limiting ZIKV infection in multiple cell lines. Combined with transcriptomics analysis, we further demonstrated that α7 nAChR activation promoted autophagy and ferroptosis pathways to limit cellular ZIKV viral loads. Additionally, activation of α7 nAChR prevented ZIKV-induced p62 nucleus accumulation, which mediated an enhanced autophagy pathway. By regulating proteasome complex and an E3 ligase NEDD4, activation of α7 nAChR resulted in increased amount of cellular p62, which further enhanced the ferroptosis pathway to reduce ZIKV infection. Moreover, utilizing in vivo neonatal mouse models, we showed that α7 nAChR is essential in controlling the disease severity of ZIKV infection. Taken together, our findings identify an α7 nAChR-mediated effect that critically contributes to limiting ZIKV infection, and α7 nAChR activation offers a novel strategy for combating ZIKV infection and its complications.
Collapse
Affiliation(s)
- Caiqi Zhao
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Jie Chen
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; University of Chinese Academy of Sciences, Beijing 100190, China; Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhihua Liu
- University of Chinese Academy of Sciences, Beijing 100190, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Huabin Liang
- University of Chinese Academy of Sciences, Beijing 100190, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Xiaoyan Chen
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lianping Cheng
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Shitao Xie
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Zhekai Lin
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Renlan Wu
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Qi Zhao
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yue Xue
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Xiaoyun Lai
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Xia Jin
- University of Chinese Academy of Sciences, Beijing 100190, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China.
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China.
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
46
|
Yan Y, Yang H, Yang Y, Wang J, Zhou Y, Tang C, Li B, Huang Q, An R, Liang X, Lin D, Yu W, Fan C, Lu S. The inoculum dose of Zika virus can affect the viral replication dynamics, cytokine responses and survival rate in immunocompromised AG129 mice. MOLECULAR BIOMEDICINE 2024; 5:30. [PMID: 39095588 PMCID: PMC11297010 DOI: 10.1186/s43556-024-00195-x] [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: 02/19/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
Zika virus, a mosquito-borne arbovirus, has repeatedly caused large pandemics with symptoms worsening from mild and self-limiting diseases to Guillain-Barré syndrome in adults and fetal microcephaly in newborns. In recent years, Zika virus diseases have posed a serious threat to human health. The shortage of susceptible small animal models makes it difficult to study pathogenic mechanisms and evaluate potential therapies for Zika virus infection. Therefore, we chose immunocompromised mice (AG129 mice) deficient in IFN-α/β and IFN-γ receptors, which can abolish the innate immune system that prevents Zika virus infection early. AG129 mice were infected with the Zika virus, and this mouse model exhibited replication dynamics, tissue tropism, pathological lesion and immune activation of the Zika virus. Our results suggest that the inoculum dose of Zika virus can affect the viral replication dynamics, cytokine responses and survival rate in AG129 mice. By testing the potential antiviral drug favipiravir, several critical indicators, including replication dynamics and survival rates, were identified in AG129 mice after Zika virus infection. It is suggested that the model is reliable for drug evaluation. In brief, this model provides a potential platform for studies of the infectivity, virulence, and pathogenesis of the Zika virus. Moreover, the development of an accessible mouse model of Zika virus infection will expedite the research and deployment of therapeutics and vaccines.
Collapse
Affiliation(s)
- Yuhuan Yan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Hao Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Junbin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Yanan Zhou
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Cong Tang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Bai Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Qing Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Ran An
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Xiaoming Liang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Dongdong Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China.
| | - Changfa Fan
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China.
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical School, Kunming, 650118, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Kunming, China.
| |
Collapse
|
47
|
Lai ZZ, Yen IC, Hung HY, Hong CY, Lai CW, Lee YM. In Vitro Antiviral Activity of Rhodiola crenulata Extract against Zika Virus and Japanese Encephalitis Virus: Viral Binding and Stability. Pharmaceuticals (Basel) 2024; 17:988. [PMID: 39204093 PMCID: PMC11357342 DOI: 10.3390/ph17080988] [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: 06/17/2024] [Revised: 07/13/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Zika virus (ZIKV) and Japanese encephalitis virus (JEV) can cause permanent neurological damage and death, yet no approved drugs exist for these infections. Rhodiola crenulate, an herb used in traditional Chinese medicine for its antioxidation and antifatigue properties, was studied for its antiviral activity against ZIKV and JEV in vitro. The cytotoxicity of Rhodiola crenulata extract (RCE) was evaluated using the CCK-8 reagent. Antiviral effects of RCE were assessed in ZIKV-infected or JEV-infected Vero cells via quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blotting, fluorescent focus assay (FFA), and immunofluorescence assay (IFA). The cell-free antiviral effects of RCE were evaluated using an inactivation assay. To determine the stage of the viral life cycle affected by RCE, time-of-addition, binding, and entry assays were conducted. Three bioactive constituents of RCE (salidroside, tyrosol, and gallic acid) were tested for antiviral activity. RCE exhibited dose-dependent anti-ZIKV and anti-JEV activities at non-cytotoxic concentrations, which were likely achieved by disrupting viral binding and stability. Gallic acid exhibited antiviral activity against ZIKV and JEV. Our findings indicate that RCE disrupts viral binding and stability, presenting a potential strategy to treat ZIKV and JEV infections.
Collapse
Affiliation(s)
- Zheng-Zong Lai
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei 114, Taiwan;
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114, Taiwan; (H.-Y.H.); (C.-Y.H.)
| | - I-Chuan Yen
- School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan; (I.-C.Y.); (C.-W.L.)
| | - Hao-Yuan Hung
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114, Taiwan; (H.-Y.H.); (C.-Y.H.)
| | - Chen-Yang Hong
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114, Taiwan; (H.-Y.H.); (C.-Y.H.)
| | - Chih-Wei Lai
- School of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan; (I.-C.Y.); (C.-W.L.)
| | - Yen-Mei Lee
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114, Taiwan; (H.-Y.H.); (C.-Y.H.)
| |
Collapse
|
48
|
Berglund G, Lennon CD, Badu P, Berglund JA, Pager CT. Transcriptomic Signatures of Zika Virus Infection in Patients and a Cell Culture Model. Microorganisms 2024; 12:1499. [PMID: 39065267 PMCID: PMC11278784 DOI: 10.3390/microorganisms12071499] [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: 05/27/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Zika virus (ZIKV), a re-emerging flavivirus, is associated with devasting developmental and neurological disease outcomes particularly in infants infected in utero. Towards understanding the molecular underpinnings of the unique ZIKV disease pathologies, numerous transcriptome-wide studies have been undertaken. Notably, these studies have overlooked the assimilation of RNA-seq analysis from ZIKV-infected patients with cell culture model systems. In this study we find that ZIKV-infection of human lung adenocarcinoma A549 cells, mirrored both the transcriptional and alternative splicing profiles from previously published RNA-seq data of peripheral blood mononuclear cells collected from pediatric patients during early acute, late acute, and convalescent phases of ZIKV infection. Our analyses show that ZIKV infection in cultured cells correlates with transcriptional changes in patients, while the overlap in alternative splicing profiles was not as extensive. Overall, our data indicate that cell culture model systems support dissection of select molecular changes detected in patients and establishes the groundwork for future studies elucidating the biological implications of alternative splicing during ZIKV infection.
Collapse
Affiliation(s)
- Gillian Berglund
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
| | - Claudia D. Lennon
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
| | - Pheonah Badu
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
| | - John Andrew Berglund
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
| | - Cara T. Pager
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222, USA
| |
Collapse
|
49
|
Mendonça RZ, Nascimento RM, Fernandes ACO, Silva PI. Antiviral action of aqueous extracts of propolis from Scaptotrigona aff. postica (Hymenoptera; Apidae) against Zica, Chikungunya, and Mayaro virus. Sci Rep 2024; 14:15289. [PMID: 38961137 PMCID: PMC11222429 DOI: 10.1038/s41598-024-65636-7] [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: 02/06/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
The limited availability of antivirals for new highly pathogenic strains of virus has become a serious public health. Therefore, news products against these pathogens has become an urgent necessity. Among the multiple sources for news antibiotics and antivirals, insect exudates or their products has become an increasingly frequent option. Insects emerged 350 million years ago and have showed a high adaptability and resistance to the most varied biomes. Their survival for so long, in such different environments, is an indication that they have a very efficient protection against environmental infections, despite not having a developed immune system like mammals. Since the ancient civilizations, the products obtained from the bee have been of great pharmacological importance, being used as antimicrobial, anti-inflammatory, antitumor and several other functions. Investigations of biological activity of propolis have been carried out, mainly in the species Apis mellifera, and its product have showed activity against some important viruses. However, for the Meliponini species, known as stingless bees, there are few studies, either on their chemical composition or on their biological activities. The importance of studying these bees is because they come from regions with native forests, and therefore with many species of plants not yet studied, in addition to which they are regions still free of pesticides, which guarantees a greater fidelity of the obtained data. Previous studies by our group with crude hydroalcoholic extract of propolis demonstrated an intense antiviral activity against Herpes, influenza, and rubella viruses. In this work, we chose to use aqueous extracts, which eliminates the presence of other compounds besides those originally present in propolis, in addition to extracting substances different from those obtained in alcoholic extracts. Therefore, this study aimed to identify, isolate and characterize compounds with antiviral effects from aqueous propolis extracts from Scaptotrigona aff postica, in emerging viruses such as zicavirus, chikungunya, and mayaro virus. The evaluation of the antiviral activity of the crude and purified material was performed by reducing infectious foci in VERO cell cultures. The results obtained with crude propolis, indicate a high reduction of zica virus (64×) and mayaro (128×) when was used 10% v/v of propolis. The reduction of chikungunya virus was of 256 fold, even when was used 5% v/v of propolis. The chemical characterization of the compounds present in the extracts was performed by high-pressure liquid chromatography. Through the purification of propolis by HPLC and mass spectrometry, it was possible to identify and isolate a peak with antiviral activity. This substance showed activity against all viruses tested. When purified fraction was used, the reduction observed was of 16 fold for zicavirus, 32 fold for mayaro virus and 512 fold for chikungunya virus. Likewise, it was observed that the antiviral response was concentration dependent, being more intense when propolis was added 2 h after the viral infection. Now we are carrying out the chemical characterization of the purified compounds that showed antiviral action.
Collapse
Affiliation(s)
- R Z Mendonça
- Laboratory of Parasitology, Butantan Institute, São Paulo, Brazil
| | - R M Nascimento
- Laboratory of Parasitology, Butantan Institute, São Paulo, Brazil
| | - A C O Fernandes
- Laboratory of Parasitology, Butantan Institute, São Paulo, Brazil
| | - P I Silva
- Laboratory for Applied Toxinology (LETA), Center of Toxins, Immune-Response and Cell Signaling (CeTICS/CEPID), Butantan Institute, São Paulo, Brazil.
| |
Collapse
|
50
|
Badu P, Baniulyte G, Sammons MA, Pager CT. Activation of ATF3 via the Integrated Stress Response Pathway Regulates Innate Immune Response to Restrict Zika Virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.26.550716. [PMID: 37546954 PMCID: PMC10402074 DOI: 10.1101/2023.07.26.550716] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Zika virus (ZIKV) is a re-emerging mosquito-borne flavivirus that can have devastating health consequences. The developmental and neurological effects from a ZIKV infection arise in part from the virus triggering cellular stress pathways and perturbing transcriptional programs. To date, the underlying mechanisms of transcriptional control directing viral restriction and virus-host interaction are understudied. Activating Transcription Factor 3 (ATF3) is a stress-induced transcriptional effector that modulates the expression of genes involved in a myriad of cellular processes, including inflammation and antiviral responses, to restore cellular homeostasis. While ATF3 is known to be upregulated during ZIKV infection, the mode by which ATF3 is activated and the specific role of ATF3 during ZIKV infection is unknown. In this study, we show via inhibitor and RNA interference approaches that ZIKV infection initiates the integrated stress response pathway to activate ATF4 which in turn induces ATF3 expression. Additionally, by using CRISPR-Cas9 system to delete ATF3, we found that ATF3 acts to limit ZIKV gene expression in A549 cells. We also determined that ATF3 enhances the expression of antiviral genes such as STAT1 and other components in the innate immunity pathway to induce an ATF3-dependent anti-ZIKV response. Our study reveals crosstalk between the integrated stress response and innate immune response pathways and highlights an important role for ATF3 in establishing an antiviral effect during ZIKV infection.
Collapse
Affiliation(s)
- Pheonah Badu
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
| | - Gabriele Baniulyte
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
| | - Morgan A. Sammons
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
| | - Cara T. Pager
- Department of Biological Sciences, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
- The RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, NY 12222
| |
Collapse
|