1
|
Romero-Leiton JP, Laison EK, Alfaro R, Parmley EJ, Arino J, Acharya KR, Nasri B. Exploring Zika's dynamics: A scoping review journey from epidemic to equations through mathematical modelling. Infect Dis Model 2025; 10:536-558. [PMID: 39897087 PMCID: PMC11786632 DOI: 10.1016/j.idm.2024.12.016] [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: 08/01/2024] [Revised: 11/24/2024] [Accepted: 12/29/2024] [Indexed: 02/04/2025] Open
Abstract
Zika virus (ZIKV) infection, along with the concurrent circulation of other arboviruses, presents a great public health challenge, reminding the utilization of mathematical modelling as a crucial tool for explaining its intricate dynamics and interactions with co-circulating pathogens. Through a scoping review, we aimed to discern current mathematical models investigating ZIKV dynamics, focusing on its interplay with other pathogens, and to identify underlying assumptions and deficiencies supporting attention, particularly regarding the epidemiological attributes characterizing Zika outbreaks. Following the PRISMA-Sc guidelines, a systematic search across PubMed, Web of Science, and MathSciNet provided 137 pertinent studies from an initial pool of 2446 papers, showing a diversity of modelling approaches, predominantly centered on vector-host compartmental models, with a notable concentration on the epidemiological landscapes of Colombia and Brazil during the 2015-2016 Zika epidemic. While modelling studies have been important in explaining Zika transmission dynamics and their intersections with diseases such as Dengue, Chikungunya, and COVID-19 so far, future Zika models should prioritize robust data integration and rigorous validation against diverse datasets to improve the accuracy and reliability of epidemic prediction. In addition, models could benefit from adaptable frameworks incorporating human behavior, environmental factors, and stochastic parameters, with an emphasis on open-access tools to foster transparency and research collaboration.
Collapse
Affiliation(s)
- Jhoana P. Romero-Leiton
- Department of Mathematical Sciences, University of Puerto Rico at Mayagüez, Puerto Rico, PR 00681-9000, USA
| | - Elda K.E. Laison
- Département de Médecine Sociale et Préventive, École de Santé Publique de L’Université de Montréal, Montréal, QC Québec, H3N 1X9, Canada
| | - Rowin Alfaro
- Département de Médecine Sociale et Préventive, École de Santé Publique de L’Université de Montréal, Montréal, QC Québec, H3N 1X9, Canada
| | - E. Jane Parmley
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Julien Arino
- Department of Mathematics, University of Manitoba, Winnipeg, MB, R3T 1E9, Canada
| | - Kamal R. Acharya
- Asia-Pacific Center for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010 Australia
| | - Bouchra Nasri
- Département de Médecine Sociale et Préventive, École de Santé Publique de L’Université de Montréal, Montréal, QC Québec, H3N 1X9, Canada
- Centre de Recherches Mathématiques, Montréal, Canada
- Centre de Recherche en Santé Publique, Montréal, Canada
- Data Informatics Center of Epidemiology, PathCheck, Cambridge, USA
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Bahojb Mahdavi SZ, Jebelli A, Aghbash PS, Baradaran B, Amini M, Oroojalian F, Pouladi N, Baghi HB, de la Guardia M, Mokhtarzadeh AA. A comprehensive overview on the crosstalk between microRNAs and viral pathogenesis and infection. Med Res Rev 2025; 45:349-425. [PMID: 39185567 PMCID: PMC11796338 DOI: 10.1002/med.22073] [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: 11/06/2021] [Revised: 04/11/2023] [Accepted: 08/04/2024] [Indexed: 08/27/2024]
Abstract
Infections caused by viruses as the smallest infectious agents, pose a major threat to global public health. Viral infections utilize different host mechanisms to facilitate their own propagation and pathogenesis. MicroRNAs (miRNAs), as small noncoding RNA molecules, play important regulatory roles in different diseases, including viral infections. They can promote or inhibit viral infection and have a pro-viral or antiviral role. Also, viral infections can modulate the expression of host miRNAs. Furthermore, viruses from different families evade the host immune response by producing their own miRNAs called viral miRNAs (v-miRNAs). Understanding the replication cycle of viruses and their relation with host miRNAs and v-miRNAs can help to find new treatments against viral infections. In this review, we aim to outline the structure, genome, and replication cycle of various viruses including hepatitis B, hepatitis C, influenza A virus, coronavirus, human immunodeficiency virus, human papillomavirus, herpes simplex virus, Epstein-Barr virus, Dengue virus, Zika virus, and Ebola virus. We also discuss the role of different host miRNAs and v-miRNAs and their role in the pathogenesis of these viral infections.
Collapse
Affiliation(s)
- Seyedeh Zahra Bahojb Mahdavi
- Department of Biology, Faculty of Basic SciencesAzarbaijan Shahid Madani UniversityTabrizIran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Asiyeh Jebelli
- Department of Biological Science, Faculty of Basic ScienceHigher Education Institute of Rab‐RashidTabrizIran
- Tuberculosis and Lung Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | | | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohammad Amini
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic SciencesAzarbaijan Shahid Madani UniversityTabrizIran
| | | | | | | |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
Chung NPY, Cheng CY. Testis Is a Sanctuary Site for HIV-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:433-440. [PMID: 40301268 DOI: 10.1007/978-3-031-82990-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
In this review, we summarize some recent findings regarding the likely mechanisms by which HIV-1 uses the testis as a sanctuary site for evading the effects of antiviral drugs from reaching the testis, due to the presence of the blood-testis barrier. This discussion also sheds insights into the possibility of eradicating the small viral pool in the testis behind the blood-testis barrier in future functional studies. These findings also bridge the knowledge gap of eradicating viral particles hiding behind the blood-brain barrier in the brain since it is anticipated that the blood-tissue barriers, namely the blood-testis and the blood-brain barriers, are utilizing similar mechanisms to regulate the dynamic nature of their tight junctions.
Collapse
Affiliation(s)
- Nancy P Y Chung
- Department of Biological Sciences, New York City College of Technology, City University of New York, New York, NY, USA
| | - C Yan Cheng
- Department of Biology, St Francis College, New York, NY, USA
| |
Collapse
|
9
|
Wang J, Yu Z, Chen Z, Ye F, Sun Z. The Potential Role of Zika and Dengue Virus Infection in the Urogenital System Disorders: An Overview. Rev Med Virol 2025; 35:e70010. [PMID: 39804234 DOI: 10.1002/rmv.70010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 05/02/2025]
Abstract
Arboviruses currently are regarded as a major worldwide public health concern. The clinical outcomes associated with this group of viruses may vary from asymptomatic infections to severe forms of haemorrhagic fever characterised by bleeding disorders. Similar to other systemic viral infections, arboviruses can either directly or indirectly affect different parts of the body, such as the urogenital system. The human urogenital system anatomically consists of two major subdivisions: (i) the urinary system, including the kidneys, ureters, bladder, and urethra, which plays a significant role in osmoregulation, control of blood volume, pressure, and PH, absorption/excretion of different ions, and toxin metabolism, and (ii) the genital system, composed of the prostate, uterus, testes, ovaries, penis, and vagina, which are responsible for reproductive functions. Arboviruses can impair normal urogenital system functions by direct viral pathogen activity, systemic forms of inflammation, haemorrhagic events and related dysfunctions, and the nephrotoxic side effects of specific medications employed for treatment leading to various urogenital disorders. The present review provides an overview of the potential capacity of two main arboviruses, known as Zika and dengue viruses, to affect the urogenital system. Moreover, it addresses Zika virus as a potential therapeutic oncolytic virus for urogenital cancers.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zongze Yu
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
| | - Zhigui Chen
- Department of Urology, The Second People's Hospital of Meishan City, Sichuan, China
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Abdelbasset M, Saron WAA, Ma D, Rathore APS, Kozaki T, Zhong C, Mantri CK, Tan Y, Tung CC, Tey HL, Chu JJH, Chen J, Ng LG, Wang H, Ginhoux F, St John AL. Differential contributions of fetal mononuclear phagocytes to Zika virus neuroinvasion versus neuroprotection during congenital infection. Cell 2024; 187:7511-7532.e20. [PMID: 39532096 DOI: 10.1016/j.cell.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/08/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Fetal immune cell functions during congenital infections are poorly understood. Zika virus (ZIKV) can vertically transmit from mother to fetus, causing nervous system infection and congenital ZIKV syndrome (CZS). We identified differential functional roles for fetal monocyte/macrophage cell types and microglia in ZIKV dissemination versus clearance using mouse models. Trafficking of ZIKV-infected primitive macrophages from the yolk sac allowed initial fetal virus inoculation, while recruited monocytes promoted non-productive neuroinflammation. Conversely, brain-resident differentiated microglia were protective, limiting infection and neuronal death. Single-cell RNA sequencing identified transcriptional profiles linked to the protective versus detrimental contributions of mononuclear phagocyte subsets. In human brain organoids, microglia also promoted neuroprotective transcriptional changes and infection clearance. Thus, microglia are protective before birth, contrasting with the disease-enhancing roles of primitive macrophages and monocytes. Differential modulation of myeloid cell phenotypes by genetically divergent ZIKVs underscores the potential of immune cells to regulate diverse outcomes during fetal infections.
Collapse
Affiliation(s)
- Muhammad Abdelbasset
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wilfried A A Saron
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Dongliang Ma
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Abhay P S Rathore
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA
| | - Tatsuya Kozaki
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chengwei Zhong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chinmay Kumar Mantri
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Yingrou Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Chi-Ching Tung
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Hong Liang Tey
- National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine affiliated Renji Hospital, Shanghai, China
| | - Hongyan Wang
- Neuroscience & Behavioral Disorders Programme, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France; Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Ashley L St John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pathology, Duke University Medical Center, Durham, NC 27705, USA; SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| |
Collapse
|
11
|
Wahaab A, Mustafa BE, Hameed M, Batool H, Tran Nguyen Minh H, Tawaab A, Shoaib A, Wei J, Rasgon JL. An Overview of Zika Virus and Zika Virus Induced Neuropathies. Int J Mol Sci 2024; 26:47. [PMID: 39795906 PMCID: PMC11719530 DOI: 10.3390/ijms26010047] [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: 11/09/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Flaviviruses pose a major public health concern across the globe. Among them, Zika virus (ZIKV) is an emerging and reemerging arthropod-borne flavivirus that has become a major international public health problem following multiple large outbreaks over the past two decades. The majority of infections caused by ZIKV exhibit mild symptoms. However, the virus has been found to be associated with a variety of congenital neural abnormalities, including microcephaly in children and Guillain-Barre syndrome in adults. The exact prediction of the potential of ZIKV transmission is still enigmatic and underlines the significance of routine detection of the virus in suspected areas. ZIKV transmission from mother to fetus (including fetal abnormalities), viral presence in immune-privileged areas, and sexual transmission demonstrate the challenges in understanding the factors governing viral persistence and pathogenesis. This review illustrates the transmission patterns, epidemiology, control strategies (through vaccines, antivirals, and vectors), oncolytic aspects, molecular insights into neuro-immunopathogenesis, and other neuropathies caused by ZIKV. Additionally, we summarize in vivo and in vitro models that could provide an important platform to study ZIKV pathogenesis and the underlying governing cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Abdul Wahaab
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; (A.W.); (H.T.N.M.)
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Bahar E Mustafa
- School of Veterinary Science, Faculty of Science, The University of Melbourne, Melbourne, VIC 3030, Australia;
- Sub Campus Toba Tek Singh, University of Agriculture, Faisalabad 36050, Pakistan;
| | - Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
- Center for Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
- Department of Otolaryngology-Head and Neck Surgery, Department of Pathology and Immunology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Hira Batool
- Chughtai Lab, Head Office, 7-Jail Road, Main Gulberg, Lahore 54000, Pakistan;
| | - Hieu Tran Nguyen Minh
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; (A.W.); (H.T.N.M.)
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Abdul Tawaab
- Sub Campus Toba Tek Singh, University of Agriculture, Faisalabad 36050, Pakistan;
| | - Anam Shoaib
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA;
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China;
| | - Jason L. Rasgon
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; (A.W.); (H.T.N.M.)
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Graham JB, Swarts JL, Koehne AL, Watson CE, Lund JM. Regulatory T cells restrict immunity and pathology in distal tissue sites following a localized infection. Mucosal Immunol 2024; 17:923-938. [PMID: 38908483 PMCID: PMC11484473 DOI: 10.1016/j.mucimm.2024.06.007] [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: 02/15/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
Regulatory T cells (Tregs) are well-known to mediate peripheral tolerance at homeostasis, and there is a growing appreciation for their role in modulating infectious disease immunity. Following acute and chronic infections, Tregs can restrict pathogen-specific T cell responses to limit immunopathology. However, it is unclear if Tregs mediate control of pathology and immunity in distal tissue sites during localized infections. We investigated the role of Tregs in immunity and disease in various tissue compartments in the context of "mild" vaginal Zika virus infection. We found that Tregs are critical to generating robust virus-specific CD8 T cell responses in the initial infection site. Further, Tregs limit inflammatory cytokines and immunopathology during localized infection; a dysregulated immune response in Treg-depleted mice leads to increased T cell infiltrates and immunopathology in both the vagina and the central nervous system (CNS). Importantly, these CNS infiltrates are not present at the same magnitude during infection of Treg-sufficient mice, in which there is no CNS immunopathology. Our data suggest that Tregs are necessary to generate a robust virus-specific response at the mucosal site of infection, while Treg-mediated restriction of bystander inflammation limits immunopathology both at the site of infection as well as distal tissue sites.
Collapse
Affiliation(s)
- Jessica B Graham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jessica L Swarts
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Amanda L Koehne
- Experimental Histopathology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Christine E Watson
- Experimental Histopathology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jennifer M Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Motta A, Musella G, Dai Prà T, Ballini A, Dioguardi M, Bizzoca ME, Lo Muzio L, Cantore S. The literature discusses oral manifestations caused by sexually transmitted viruses: a narrative review. Minerva Dent Oral Sci 2024; 73:238-247. [PMID: 38869836 DOI: 10.23736/s2724-6329.24.04996-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
This review provides practical recommendations for dental practitioners in dealing with oral lesions associated with sexually transmitted diseases (STDs), offering clinically relevant insights to increase the awareness of these diseases in the mainstream of everyday practice. STDs are acquired through unprotected vaginal, anal or oral sex and are caused by more than 30 different types of bacteria, viruses and protozoa. Saliva, precum, semen, vaginal secretions and menstrual blood could be likely vehicles of infections and defensive barriers to infection by pathogenic microbes could be represented via intact mucosal membrane, the diluent function of saliva, and the antimicrobial action of salivary enzymes that collectively contribute to oral health and protection. STD, can directly and indirectly affect mucous membranes, manifesting with characteristic diagnostic signs and lesions. Given their potential oral manifestations, dental professionals need a comprehensive understanding of STD. The findings of this review lay a foundation for comprehending several STDs, emphasizing the importance of physicians as well dental practitioners being open to discussing sexuality issues with patients and providing appropriate therapeutic interventions.
Collapse
Affiliation(s)
- Alessandro Motta
- Unit of Oral Surgery for Special Needs and Dentistry, Borgo Valsugana, Trento, Italy
| | - Gennaro Musella
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Tommaso Dai Prà
- Unit of Oral Surgery for Special Needs and Dentistry, Borgo Valsugana, Trento, Italy
| | - Andrea Ballini
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Mario Dioguardi
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria E Bizzoca
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy -
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Stefania Cantore
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| |
Collapse
|
17
|
CONSIGNY PH. [Zika virus infection: sexual transmission and implications for prevention]. MEDECINE TROPICALE ET SANTE INTERNATIONALE 2024; 4:mtsi.v4i2.2024.502. [PMID: 39099710 PMCID: PMC11292433 DOI: 10.48327/mtsi.v4i2.2024.502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/19/2024] [Indexed: 08/06/2024]
Abstract
Zika virus infection, most oft n responsible for a benign arboviral disease or an asymptomatic infection, rarely Guillain-Barré syndrome, can become problematic in pregnant women, due to a risk of fetal malformations, in particular microcephaly linked to its neurotropism. The most recent large-scale epidemic was observed throughout Latin America between 2015 and 2017, causing several hundred thousand cases. Transmission is predominantly vector-borne, but sexual transmission has been described, mainly among travelers, although it undoubtedly accounts for a significant proportion of transmission in epidemic areas. The aim of this review is to describe this sexual transmission, mainly through examples linked to this large-scale epidemic in Latin America, to describe the link with prolonged excretion of infectious viral particles in genital secretions, especially semen but also vaginal secretions, and to highlight possible preventive measures apart from vector transmission, in particular the need for pregnant women or women wishing to become pregnant to avoid visiting countries where circulation of Zika virus is described.
Collapse
Affiliation(s)
- Paul Henri CONSIGNY
- Centre médical de l’Institut Pasteur, 211 rue de Vaugirard, 75015 Paris, France
| |
Collapse
|
18
|
Campos RK, Liang Y, Azar SR, Ly J, Camargos VN, Hager-Soto EE, Eyzaguirre E, Sun J, Rossi SL. CD8 + T cells promote ZIKV clearance and mitigate testicular damage in mice. NPJ VIRUSES 2024; 2:20. [PMID: 40295722 PMCID: PMC11721072 DOI: 10.1038/s44298-024-00033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/06/2024] [Indexed: 04/30/2025]
Abstract
Zika virus (ZIKV) causes human testicular inflammation and alterations in sperm parameters and causes testicular damage in mouse models. The involvement of individual immune cells in testicular damage is not fully understood. We detected virus in the testes of the interferon (IFN) α/β receptor-/- A129 mice three weeks post-infection and found elevated chemokines in the testes, suggesting chronic inflammation and long-term infection play a role in testicular damage. In the testes, myeloid cells and CD4+ T cells were absent at 7 dpi but were present at 23 days post-infection (dpi), and CD8+ T cell infiltration started at 7 dpi. CD8-/- mice with an antibody-depleted IFN response had a significant reduction in spermatogenesis, indicating that CD8+ T cells are essential to prevent testicular damage during long-term ZIKV infections. Our findings on the dynamics of testicular immune cells and the importance of CD8+ T cells function as a framework to understand mechanisms underlying observed inflammation and sperm alterations in humans.
Collapse
Affiliation(s)
- Rafael K Campos
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sasha R Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
| | - Judy Ly
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | - Eduardo Eyzaguirre
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Shannan L Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
- Center of Tropical Disease, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
19
|
Leite-Aguiar R, Cristina-Rodrigues F, Ciarlini-Magalhães R, Dantas DP, Alves VS, Gavino-Leopoldino D, Neris RLS, Schmitz F, Silveira JS, Kurtenbach E, Wyse ATS, Clarke JR, Figueiredo CP, Assunção-Miranda I, Pimentel-Coelho PM, Coutinho-Silva R, Savio LEB. ATP-P2X7 signaling mediates brain pathology while contributing to viral control in perinatal Zika virus infection. Brain Behav Immun 2024; 118:318-333. [PMID: 38460804 DOI: 10.1016/j.bbi.2024.02.035] [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: 10/10/2023] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/11/2024] Open
Abstract
Zika virus (ZIKV), the causative agent of Zika fever, is a flavivirus transmitted by mosquitoes of the Aedes genus. Zika virus infection has become an international concern due to its association with severe neurological complications such as fetal microcephaly. Viral infection can induce the release of ATP in the extracellular environment, activating receptors sensitized by extracellular nucleotides, such as the P2X7 receptor. This receptor is the primary purinergic receptor involved in neuroinflammation, neurodegeneration, and immunity. In this work, we investigated the role of ATP-P2X7 receptor signaling in Zika-related brain abnormalities. Wild-type mice (WT) and P2X7 receptor-deficient (P2X7-/-) C57BL/6 newborn mice were subcutaneously inoculated with 5 × 106plaque-forming units of ZIKV or mock solution. P2X7 receptor expression increased in the brain of Zika virus-infected mice compared to the mock group. Comparative analyses of the hippocampi from WT and P2X7-/-mice revealed that the P2X7 receptor increased hippocampal damage in CA1/CA2 and CA3 regions. Doublecortin expression decreased significantly in the brains of ZIKV-infected mice. WT ZIKV-infected mice showed impaired motor performance compared to P2X7-/- infected mice. WT ZIKV-infected animals showed increased expression of glial markers GFAP (astrocytes) and IBA-1 (microglia) compared to P2X7-/- infected mice. Although the P2X7 receptor contributes to neuronal loss and neuroinflammation, WT mice were more efficient in controlling the viral load in the brain than P2X7 receptor-deficient mice. This result was associated with higher induction of TNF-α, IFN-β, and increased interferon-stimulated gene expression in WT mice than P2X7-/-ZIKV-infected. Finally, we found that the P2X7 receptor contributes to inhibiting the neuroprotective signaling pathway AKT/mTOR while stimulating the caspase-3 activation, possibly two distinct pathways contributing to neurodegeneration. These findings suggest that ATP-P2X7 receptor signaling contributes to the antiviral response in the brain of ZIKV-infected mice while increasing neuronal loss, neuroinflammation, and related brain abnormalities.
Collapse
Affiliation(s)
- Raíssa Leite-Aguiar
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiana Cristina-Rodrigues
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Roberta Ciarlini-Magalhães
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danillo Pereira Dantas
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinícius Santos Alves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel Gavino-Leopoldino
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rômulo Leão Silva Neris
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe Schmitz
- Departamento de Bioquímica, Instituto de Ciências Básicas de Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Josiane Silva Silveira
- Departamento de Bioquímica, Instituto de Ciências Básicas de Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Eleonora Kurtenbach
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Angela T S Wyse
- Departamento de Bioquímica, Instituto de Ciências Básicas de Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Julia Rosauro Clarke
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | | | - Iranaia Assunção-Miranda
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
20
|
Wong JCC, Tay M, Hapuarachchi HC, Lee B, Yeo G, Maliki D, Lee W, Mohamed Suhaimi NA, Chio K, Tan WCH, Ng LC. Case report: Zika surveillance complemented with wastewater and mosquito testing. EBioMedicine 2024; 101:105020. [PMID: 38387403 PMCID: PMC10897811 DOI: 10.1016/j.ebiom.2024.105020] [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: 07/18/2023] [Revised: 02/04/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND In June 2023, a local cluster of 15 Zika cases was reported in a neighbourhood in Northeastern Singapore. The last significant local transmission of Zika virus (ZIKV) with more than 450 cases was in 2016-2017. To monitor the situation and mitigate further transmission, case, entomological and wastewater-based surveillance were carried out. METHODS Primary healthcare practitioners and the community were alerted to encourage timely case identification. Surveillance was enhanced through testing of Aedes mosquitoes collected from the National Gravitrap surveillance system, and wastewater samples were collected from a network of autosamplers deployed at manholes across the country. FINDINGS ZIKV RNA was detected in mosquito pools (3/43; 7%) and individual mosquitoes (3/82; 3.7%) captured, and in wastewater samples (13/503) collected from the vicinity of the cluster of cases. Respective samples collected from other sites across the country were negative. The peak detection of ZIKV RNA in mosquitoes and wastewater coincided temporally with the peak in the number of cases in the area (15-25 May 2023). INTERPRETATION The restriction of ZIKV signals from wastewater and mosquitoes within the neighbourhood suggested limited ZIKV transmission. The subsequent waning of signals suggested effectiveness of control measures. We demonstrate the utility of wastewater-based surveillance of ZIKV, which complements existing case- and entomological-based surveillance. The non-intrusive approach is particularly useful to monitor diseases such as Zika, which generally causes silent or mild infections, but may cause severe outcomes such as congenital Zika syndrome. FUNDING This study was funded by Singapore's Ministry of Finance and the National Environment Agency, Singapore.
Collapse
Affiliation(s)
| | - Martin Tay
- Environmental Health Institute, National Environment Agency, Singapore
| | | | - Benjamin Lee
- Environmental Health Institute, National Environment Agency, Singapore
| | - Gladys Yeo
- Environmental Health Institute, National Environment Agency, Singapore
| | | | - Winston Lee
- Environmental Health Institute, National Environment Agency, Singapore
| | | | - Kaiyun Chio
- Environmental Public Health Operations Group, National Environment Agency, Singapore
| | | | - Lee Ching Ng
- Environmental Health Institute, National Environment Agency, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
21
|
Rabaan AA, AlShehail BM, Halwani MA, Alshengeti A, Najim MA, Garout M, Bajunaid HA, Alshamrani SA, Al Fares MA, Alissa M, Alwashmi ASS. Investigation of Zika virus methyl transferase inhibitors using steered molecular dynamics. J Biomol Struct Dyn 2024; 42:1711-1724. [PMID: 37325855 DOI: 10.1080/07391102.2023.2224882] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/05/2023] [Indexed: 06/17/2023]
Abstract
Zika virus (ZIKV) spread is considered a major public health threat by the World Health Organization (WHO). There are no vaccines or drugs available to control the infection of the Zika virus, therefore a highly effective medicinal molecule is urgently required. In this study, a computationally intensive investigation was performed to identify a potent natural compound that could inhibit the ZIKV NS5 methyltransferase. This research approach is based on target-based drug identification principles where the native inhibitor SAH (S-adenosylhomocysteine) of ZIKV NS5 methyltransferase was selected as a reference. High-throughput virtual screening and tanimoto similarity coefficient were applied to the natural compound library for ranking the potential candidates. The top five compounds were selected for interaction analysis, MD simulation, total binding free energy through MM/GBSA, and steered MD simulation. Among these compounds, Adenosine 5'-monophosphate monohydrate, Tubercidin, and 5-Iodotubercidin showed stable binding to the protein compared to the native compound, SAH. These three compounds also showed less fluctuations in RMSF in contrast to native compound. Additionally, the same interacting residues observed in SAH also made strong interactions with these three compounds. Adenosine 5'-monophosphate monohydrate and 5-Iodotubercidin had greater total binding free energies than the reference ligand. Moreover, the dissociation resistance of all three compounds was equivalent to that of the reference ligand. This study suggested binding properties of three-hit compounds that could be used to develop drugs against Zika virus infections.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Bashayer M AlShehail
- Pharmacy Practice Department, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Muhammad A Halwani
- Department of Medical Microbiology, Faculty of Medicine, Al Baha University, Saudi Arabia
| | - Amer Alshengeti
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah, Saudi Arabia
- Department of Infection Prevention and Control, Prince Mohammad Bin Abdulaziz Hospital, National Guard Health Affairs, Al-Madinah, Saudi Arabia
| | - Mustafa A Najim
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Mohammed Garout
- Department of Community Medicine and Health Care for Pilgrims, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Huda A Bajunaid
- Makkah Specialized Laboratory, Fakeeh Care group, Hadda, Saudi Arabia
| | - Saleh A Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Mona A Al Fares
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ameen S S Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
22
|
Campos RK, Liang Y, Azar SR, Ly J, Camargos VN, Hager-Soto EE, Eyzaguirre E, Sun J, Rossi SL. CD8 + T cell response promotes viral clearance and reduces chances of severe testicular damage in mouse models of long-term Zika virus infection of the testes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.575592. [PMID: 38328060 PMCID: PMC10849515 DOI: 10.1101/2024.01.22.575592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Zika virus (ZIKV) causes human testicular inflammation and alterations in sperm parameters and causes testicular damage in mouse models. The involvement of individual immune cells in testicular damage is not fully understood. We detected virus in the testes of the interferon (IFN) α/β receptor -/- A129 mice three weeks post-infection and found elevated chemokines in the testes, suggesting chronic inflammation and long-term infection play a role in testicular damage. In the testes, myeloid cells and CD4 + T cells were absent at 7 dpi but were present at 23 days post-infection (dpi), and CD8 + T cell infiltration started at 7 dpi. CD8 -/- mice with an antibody-depleted IFN response had a significant reduction in spermatogenesis, indicating that CD8 + T cells are essential to prevent testicular damage during long-term ZIKV infections. Our findings on the dynamics of testicular immune cells and importance of CD8 + T cells functions as a framework to understand mechanisms underlying observed inflammation and sperm alterations in humans.
Collapse
|
23
|
Abstract
Flaviviruses such as dengue, Zika, and West Nile viruses are highly concerning pathogens that pose significant risks to public health. The NS1 protein is conserved among flaviviruses and is synthesized as a part of the flavivirus polyprotein. It plays a critical role in viral replication, disease progression, and immune evasion. Post-translational modifications influence NS1's stability, secretion, antigenicity, and interactions with host factors. NS1 protein forms extensive interactions with host cellular proteins allowing it to affect vital processes such as RNA processing, gene expression regulation, and cellular homeostasis, which in turn influence viral replication, disease pathogenesis, and immune responses. NS1 acts as an immune evasion factor by delaying complement-dependent lysis of infected cells and contributes to disease pathogenesis by inducing endothelial cell damage and vascular leakage and triggering autoimmune responses. Anti-NS1 antibodies have been shown to cross-react with host endothelial cells and platelets, causing autoimmune destruction that is hypothesized to contribute to disease pathogenesis. However, in contrast, immunization of animal models with the NS1 protein confers protection against lethal challenges from flaviviruses such as dengue and Zika viruses. Understanding the multifaceted roles of NS1 in flavivirus pathogenesis is crucial for effective disease management and control. Therefore, further research into NS1 biology, including its host protein interactions and additional roles in disease pathology, is imperative for the development of strategies and therapeutics to combat flavivirus infections successfully. This Review provides an in-depth exploration of the current available knowledge on the multifaceted roles of the NS1 protein in the pathogenesis of flaviviruses.
Collapse
Affiliation(s)
- Dayangi R Perera
- Department of Chemistry, Faculty of Science, University of Colombo, Sri Lanka 00300
| | - Nadeeka D Ranadeva
- Department of Biomedical Science, Faculty of Health Sciences, KIU Campus Sri Lanka 10120
| | - Kavish Sirisena
- Department of Chemistry, Faculty of Science, University of Colombo, Sri Lanka 00300
- Section of Genetics, Institute for Research and Development in Health and Social Care, Sri Lanka 10120
| | | |
Collapse
|
24
|
Liu ZY, Li XF, Qin CF. A Stable Reverse Genetics System of Zika Virus Based on a Self-Splicing Group II Intron. Methods Mol Biol 2024; 2733:207-229. [PMID: 38064035 DOI: 10.1007/978-1-0716-3533-9_13] [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] [Indexed: 12/18/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus of the Flaviviridae family first isolated from a sentinel monkey in the Zika Forest, Uganda, in 1947. Since 2007, the virus has had a vast geographic expansion that extended to the Americas in 2015, leading to a series of large outbreaks. Although mainly transmitted by the bite of Aedes mosquitoes, human infection of ZIKV can also happen through unconventional routes such as sexual intercourse and, more importantly, vertical transmission. The genome of ZIKV is a single-stranded, positive-sense RNA molecule about 11 kb in length. The genome contains a single opening reading frame (ORF) flanked by highly structured 5' and 3' untranslated regions. To understand the mechanisms about ZIKV replication, transmission, and pathogenesis, reverse genetic tools are of great importance. In this chapter, a novel system is described for the generation and manipulation of a ZIKV infectious clone stabilized by a self-splicing group II intron, a mobile element with ribozyme activity. The intron can be spliced in vitro, and thus full-length vRNA can be prepared allowing virus genome manipulation required for further studies.
Collapse
Affiliation(s)
- Zhong-Yu Liu
- School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, China.
| |
Collapse
|
25
|
Zina SM, Hoarau G, Labetoulle M, Khairallah M, Rousseau A. Ocular Manifestations of Flavivirus Infections. Pathogens 2023; 12:1457. [PMID: 38133340 PMCID: PMC10747099 DOI: 10.3390/pathogens12121457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Flaviviruses are a group of positive-sense, single-stranded RNA viruses predominantly transmitted by arthropods (mainly mosquitoes) that cause severe endemic infections and epidemics on a global scale. They represent a major cause of systemic morbidity and death and are expanding worldwide. Among this group, dengue fever, the West Nile virus, yellow fever, Japanese Encephalitis, and, recently, the Zika virus have been linked to a spectrum of ocular manifestations. These manifestations encompass subconjunctival hemorrhages and conjunctivitis, anterior and posterior uveitis (inclusive of vitritis, chorioretinitis, and retinal vasculitis), maculopathy, retinal hemorrhages, and optic neuritis. Clinical diagnosis of these infectious diseases is primarily based on epidemiological data, history, systemic symptoms and signs, and the pattern of ocular involvement. Diagnosis confirmation relies on laboratory testing, including RT-PCR and serological testing. Ocular involvement typically follows a self-limited course but can result in irreversible visual impairment. Effective treatments of flavivirus infections are currently unavailable. Prevention remains the mainstay for arthropod vector and zoonotic disease control. Effective vaccines are available only for the yellow fever virus, dengue virus, and Japanese Encephalitis virus. This review comprehensively summarizes the current knowledge regarding the ophthalmic manifestations of the foremost flavivirus-associated human diseases.
Collapse
Affiliation(s)
- Sourour Meziou Zina
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
- Department of Ophthalmology, Faculty of Medicine, University of Monastir, Monastir 5019, Tunisia;
| | - Gautier Hoarau
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
| | - Marc Labetoulle
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB), Infectious Diseases Models for Innovative Therapies (IDMIT), French Alternative Energies and Atomic Commission (CEA), 92260 Fontenay-aux-Roses, France
| | - Moncef Khairallah
- Department of Ophthalmology, Faculty of Medicine, University of Monastir, Monastir 5019, Tunisia;
| | - Antoine Rousseau
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB), Infectious Diseases Models for Innovative Therapies (IDMIT), French Alternative Energies and Atomic Commission (CEA), 92260 Fontenay-aux-Roses, France
| |
Collapse
|
26
|
Mysorekar I, Michita R, Tran L, Bark S, Kumar D, Toner S, Jose J, Narayanan A. Zika Virus NS1 Drives Tunneling Nanotube Formation for Mitochondrial Transfer, Enhanced Survival, Interferon Evasion, and Stealth Transmission in Trophoblasts. RESEARCH SQUARE 2023:rs.3.rs-3674059. [PMID: 38106210 PMCID: PMC10723532 DOI: 10.21203/rs.3.rs-3674059/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Zika virus (ZIKV) infection continues to pose a significant public health concern due to limited available preventive measures and treatments. ZIKV is unique among flaviviruses in its vertical transmission capacity (i.e., transmission from mother to fetus) yet the underlying mechanisms remain incompletely understood. Here, we show that both African and Asian lineages of ZIKV induce tunneling nanotubes (TNTs) in placental trophoblasts and multiple other mammalian cell types. Amongst investigated flaviviruses, only ZIKV strains trigger TNTs. We show that ZIKV-induced TNTs facilitate transfer of viral particles, proteins, and RNA to neighboring uninfected cells. ZIKV TNT formation is driven exclusively via its non-structural protein 1 (NS1); specifically, the N-terminal region (50 aa) of membrane-bound NS1 is necessary and sufficient for triggering TNT formation in host cells. Using affinity purification-mass spectrometry of cells infected with wild-type NS1 or non-TNT forming NS1 (pNS1ΔTNT) proteins, we found mitochondrial proteins are dominant NS1-interacting partners, consistent with the elevated mitochondrial mass we observed in infected trophoblasts. We demonstrate that mitochondria are siphoned via TNTs from healthy to ZIKV-infected cells, both homotypically and heterotypically, and inhibition of mitochondrial respiration reduced viral replication in trophoblast cells. Finally, ZIKV strains lacking TNT capabilities due to mutant NS1 elicited a robust antiviral IFN-λ 1/2/3 response, indicating ZIKV's TNT-mediated trafficking also allows ZIKV cell-cell transmission that is camouflaged from host defenses. Together, our findings identify a new 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. Discerning the mechanisms of ZIKV intercellular strategies offers a basis for novel therapeutic developments targeting these interactions to limit its dissemination.
Collapse
|
27
|
Sarratea MB, Alberti AS, Redolfi DM, Truant SN, Iannantuono Lopez LV, Bivona AE, Mariuzza RA, Fernández MM, Malchiodi EL. Zika virus NS4B protein targets TANK-binding kinase 1 and inhibits type I interferon production. Biochim Biophys Acta Gen Subj 2023; 1867:130483. [PMID: 37802371 DOI: 10.1016/j.bbagen.2023.130483] [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: 04/23/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND During viral infections, nucleic acid sensing by intracellular receptors can trigger type I interferon (IFN-I) production, key mediators in antiviral innate immunity. However, many flaviviruses use non-structural proteins to evade immune sensing favoring their survival. These mechanisms remain poorly characterized. Here, we studied the role of Zika virus (ZIKV) NS4B protein in the inhibition of IFN-I induction pathway and its biophysical interaction with host proteins. METHODS Using different cell-based assays, we studied the effect of ZIKV NS4B in the activation of interferon regulatory factors (IRFs), NF-κB, cytokines secretion and the expression of interferon-stimulating genes (ISG). We also analyzed the in vitro interaction between recombinant ZIKV NS4B and TANK-binding kinase 1 (TBK1) using surface plasmon resonance (SPR). RESULTS Transfection assays showed that ZIKV NS4B inhibits IRFs activation involved in different nucleic acid sensing cascades. Cells expressing NS4B secreted lower levels of IFN-β and IL-6. Furthermore, early induction of ISGs was also restricted by ZIKV NS4B. For the first time, we demonstrate by SPR assays that TBK1, a critical component in IFN-I production pathway, binds directly to ZIKV NS4B (KD of 3.7 × 10-6 M). In addition, we show that the N-terminal region of NS4B is directly involved in this interaction. CONCLUSIONS Altogether, our results strongly support that ZIKV NS4B affects nucleic acid sensing cascades and disrupts the TBK1/IRF3 axis, leading to an impairment of IFN-β production. SIGNIFICANCE This study provides the first biophysical data of the interaction between ZIKV NS4B and TBK1, and highlights the role of ZIKV NS4B in evading the early innate immune response.
Collapse
Affiliation(s)
- Maria B Sarratea
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina; W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
| | - Andrés Sánchez Alberti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología-IMPAM (UBA-CONICET), Paraguay 2155, C1121ABG Buenos Aires, Argentina
| | - Daniela M Redolfi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina
| | - Sofía Noli Truant
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina
| | - Laura V Iannantuono Lopez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina
| | - Augusto E Bivona
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología-IMPAM (UBA-CONICET), Paraguay 2155, C1121ABG Buenos Aires, Argentina
| | - Roy A Mariuzza
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Marisa M Fernández
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina.
| | - Emilio L Malchiodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU (UBA-CONICET), Junín 956, C1113AAD Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología-IMPAM (UBA-CONICET), Paraguay 2155, C1121ABG Buenos Aires, Argentina.
| |
Collapse
|
28
|
Valega-Mackenzie W, Ríos-Soto K, Lenhart S. Optimal control applied to Zika virus epidemics in Colombia and Puerto Rico. J Theor Biol 2023; 575:111647. [PMID: 39492547 DOI: 10.1016/j.jtbi.2023.111647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 09/22/2023] [Accepted: 10/16/2023] [Indexed: 11/05/2024]
Abstract
Zika virus (ZIKV) is a mostly non-lethal disease in humans transmitted by mosquitoes or humans that can produce severe brain defects such as microcephaly in babies and Guillain-Barré syndrome in elderly adults. The use of optimal control strategies involving information campaigns about insect repellents and condoms alongside an available safe and effective vaccine can prevent the number of infected humans with ZIKV. A system of nonlinear ordinary differential equations is formulated for the transmission dynamics of ZIKV in the presence of three control strategies to evaluate the impact of various scenarios during a ZIKV epidemic. In addition, we estimate parameters using weekly incidence data from previous ZIKV outbreaks in Colombia and Puerto Rico to capture the dynamics of an epidemic in each country when control measures are available. The basic reproduction number, R0, of each country is calculated using estimated parameters (without the controls). The vector-borne transmission threshold (Rv) is dominant in both countries , but the sexual transmission threshold (Rd) in Colombia is considerably higher than in Puerto Rico. Numerical simulations for Colombia show that the most effective strategies are to use three controls since the start of the outbreak. However, for Puerto Rico only information campaigns about mosquito repellents and vaccination are the most effective ways to mitigate the epidemic.
Collapse
Affiliation(s)
| | - Karen Ríos-Soto
- Department of Mathematics, University of Puerto Rico, Mayagüez, United States of America
| | - Suzanne Lenhart
- Department of Mathematics, University of Tennessee, Knoxville, United States of America
| |
Collapse
|
29
|
Phumee A, Chitcharoen S, Sutthanont N, Intayot P, Wacharapluesadee S, Siriyasatien P. Genetic diversity and phylogenetic analyses of Asian lineage Zika virus whole genome sequences derived from Culex quinquefasciatus mosquitoes and urine of patients during the 2020 epidemic in Thailand. Sci Rep 2023; 13:18470. [PMID: 37891235 PMCID: PMC10611781 DOI: 10.1038/s41598-023-45814-9] [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: 09/14/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, has been continually emerging and re-emerging since 2010, with sporadic cases reported annually in Thailand, peaking at over 1000 confirmed positive cases in 2016. Leveraging high-throughput sequencing technologies, specifically whole genome sequencing (WGS), has facilitated rapid pathogen genome sequencing. In this study, we used multiplex amplicon sequencing on the Illumina Miseq instrument to describe ZIKV WGS. Six ZIKV WGS were derived from three samples of field-caught Culex quinquefasciatus mosquitoes (two males and one female) and three urine samples collected from patients in three different provinces of Thailand. Additionally, successful isolation of a ZIKV isolate occurred from a female Cx. quinquefasciatus. The WGS analysis revealed a correlation between the 2020 outbreak and the acquisition of five amino acid changes in the Asian lineage ZIKV strains from Thailand (2006), Cambodia (2010 and 2019), and the Philippines (2012). These changes, including C-T106A, prM-V1A, E-V473M, NS1-A188V, and NS5-M872V, were identified in all seven WGS, previously linked to significantly higher mortality rates. Furthermore, phylogenetic analysis indicated that the seven ZIKV sequences belonged to the Asian lineage. Notably, the genomic region of the E gene showed the highest nucleotide diversity (0.7-1.3%). This data holds significance in informing the development of molecular tools that enhance our understanding of virus patterns and evolution. Moreover, it may identify targets for improved methods to prevent and control future ZIKV outbreaks.
Collapse
Affiliation(s)
- Atchara Phumee
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
- Excellent Center for Dengue and Community Public Health (EC for DACH), Walailak University, Nakhon Si Thammarat, Thailand
| | - Suwalak Chitcharoen
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nataya Sutthanont
- Department of Medical Entomology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Proawpilart Intayot
- Pharmaceutical Ingredient and Medical Device Research Division, Research Development and Innovation Department, The Government Pharmaceutical Organization, Bangkok, Thailand
| | - Supaporn Wacharapluesadee
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Padet Siriyasatien
- Center of Excellence in Vector Biology and Vector Borne Diseases, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
30
|
Vasquez D, Palacio A, Chedraui P, Del Mar Sanchez M, Briones W, Tamariz L, Calle MA. Is Prior Zika Virus Infection Associated With Cardiovascular Disease? Cureus 2023; 15:e47141. [PMID: 38022258 PMCID: PMC10651160 DOI: 10.7759/cureus.47141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Zika virus (ZIKV) infection is associated with severe complications. Recently, reports have raised the possibility of cardiovascular complications. However, the complications that are reported are in case reports and occur immediately after infection. Our aim is to evaluate the cardiovascular complications of ZIKV infection in a younger patient population. METHODS We conducted a prospective cohort and included patients with a one-year history of prior confirmed ZIKV infection. We performed an echocardiogram, a 24-hour automated blood pressure, and a 24-hour Holter. Our primary outcome included a composite of having diastolic dysfunction, left ventricular hypertrophy, arrhythmias, valvular regurgitation, premature beats, or non-dipper status. RESULTS We included 47 patients with ZIKV and 16 patients without ZIKV. Patients with ZIKV had a similar age as controls (p>0.05). Having had a prior ZIKV infection was associated with diastolic dysfunction, left ventricular hypertrophy, valvular regurgitation, arrhythmias or premature beats, and non-dipper status (p<0.05). The adjusted OR of having the primary outcome was 2.3; 95% CI 1.3-2.7. After one year, IL-10 and C-reactive protein (CRP) were higher in ZIKV-infected patients compared to controls (p<0.05). CONCLUSIONS Our study found that young patients with a prior ZIKV infection have more echocardiographic, arrhythmic, and blood pressure changes when compared to similar-aged controls.
Collapse
Affiliation(s)
- Diego Vasquez
- Department of Epidemiology, Universidad Católica de Santiago de Guayaquil, Guayaquil, ECU
| | - Ana Palacio
- Population Health and Computational Medicine, University of Miami Miller School of Medicine, Miami, USA
| | - Peter Chedraui
- Escuela de Posgrado en Salud, Universidad Espíritu Santo, Samborondón, ECU
| | - Maria Del Mar Sanchez
- Department of Public Health and Epidemiology, Universidad Católica de Santiago de Guayaquil, Guayaquil, ECU
| | - Wladimir Briones
- Department of Epidemiology, Ministerio de Salud Publica, Quito, ECU
| | - Leonardo Tamariz
- Population Health and Computational Medicine, University of Miami Miller School of Medicine, Miami, USA
| | - Marco A Calle
- Department of Medicine, Universidad Católica de Santiago de Guayaquil, Guayaquil, ECU
| |
Collapse
|
31
|
Dobrzyńska M, Moniuszko-Malinowska A, Skrzydlewska E. Metabolic response to CNS infection with flaviviruses. J Neuroinflammation 2023; 20:218. [PMID: 37775774 PMCID: PMC10542253 DOI: 10.1186/s12974-023-02898-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023] Open
Abstract
Flaviviruses are arthropod-borne RNA viruses found worldwide that, when introduced into the human body, cause diseases, including neuroinfections, that can lead to serious metabolic consequences and even death. Some of the diseases caused by flaviviruses occur continuously in certain regions, while others occur intermittently or sporadically, causing epidemics. Some of the most common flaviviruses are West Nile virus, dengue virus, tick-borne encephalitis virus, Zika virus and Japanese encephalitis virus. Since all the above-mentioned viruses are capable of penetrating the blood-brain barrier through different mechanisms, their actions also affect the central nervous system (CNS). Like other viruses, flaviviruses, after entering the human body, contribute to redox imbalance and, consequently, to oxidative stress, which promotes inflammation in skin cells, in the blood and in CNS. This review focuses on discussing the effects of oxidative stress and inflammation resulting from pathogen invasion on the metabolic antiviral response of the host, and the ability of viruses to evade the consequences of metabolic changes or exploit them for increased replication and further progression of infection, which affects the development of sequelae and difficulties in therapy.
Collapse
Affiliation(s)
- Marta Dobrzyńska
- Department of Analytical Chemistry, Medical University of Białystok, Białystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540, Bialystok, Poland.
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
32
|
Shukla D, Alanazi AM, Panda SP, Dwivedi VD, Kamal MA. Unveiling the antiviral potential of Plant compounds from the Meliaceae family against the Zika virus through QSAR modeling and MD simulation analysis. J Biomol Struct Dyn 2023; 42:11064-11079. [PMID: 37728536 DOI: 10.1080/07391102.2023.2259498] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/11/2023] [Indexed: 09/21/2023]
Abstract
Zika virus (ZIKV) is a flavivirus transmitted by mosquitoes, causing neurological disorders and congenital malformations. RNA-dependent RNA polymerase (RdRp) is one of its essential enzymes and a promising drug target for antiviral therapy due to its involvement in the growth and multiplication of the virus. In this study, we conducted a QSAR-based chemical library screening from the Meliaceae family to identify potential RdRp inhibitors. The QSAR model was built using the known inhibitors of RdRp NS5 of ZIKV and their biological activity (EC50), along with the structural and chemical characteristics of the compounds. The top two hit compounds were selected from QSAR screening for further analysis using molecular docking to evaluate their binding energies and intermolecular interactions with RdRp, including the critical residue Trp485. Furthermore, molecular dynamics (MD) simulations were performed to evaluate their binding stability and flexibility upon binding to RdRp. The MD results showed that the selected compounds formed stable complexes with RdRp, and their binding interactions were similar to those observed for the native ligand. The binding energies of the top two hits (-8.6 and -7.7 kcal/mole) were comparable to those of previously reported ZIKV RdRp inhibitors (-8.9 kcal/mole). The compound IMPHY009135 showed the strongest binding affinity with RdRp, forming multiple hydrogen bonds and hydrophobic interactions with key residues. However, compound IMPHY009276 showed the most stable and consistent RMSD, which was similar to the native ligand. Our findings suggest that IMPHY009135 and IMPHY009276 are potential lead compounds for developing novel antiviral agents against ZIKV.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Divyanshi Shukla
- Department of Chemistry, Lajpat Rai PG College, Sahibabad, Ghaziabad, India
- Computational Chemistry & Drug Discovery Division, Quanta Calculus, Greater Noida, India
| | - Amer M Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar, India
| | - Vivek Dhar Dwivedi
- Computational Chemistry & Drug Discovery Division, Quanta Calculus, Greater Noida, India
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Thandalam, Tamil Nadu, India
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
| |
Collapse
|
33
|
da Costa HHM, Bielavsky M, Orts DJB, Araujo S, Adriani PP, Nogueira JS, Astray RM, Pandey RP, Lancellotti M, Cunha-Junior JP, Prudencio CR. Production of Recombinant Zika Virus Envelope Protein by Airlift Bioreactor as a New Subunit Vaccine Platform. Int J Mol Sci 2023; 24:13955. [PMID: 37762254 PMCID: PMC10531330 DOI: 10.3390/ijms241813955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 09/29/2023] Open
Abstract
The Zika Virus (ZIKV) is an emerging arbovirus of great public health concern, particularly in the Americas after its last outbreak in 2015. There are still major challenges regarding disease control, and there is no ZIKV vaccine currently approved for human use. Among many different vaccine platforms currently under study, the recombinant envelope protein from Zika Virus (rEZIKV) constitutes an alternative option for vaccine development and has great potential for monitoring ZIKV infection and antibody response. This study describes a method to obtain a bioactive and functional rEZIKV using an E. coli expression system, with the aid of a 5-L airlift bioreactor and following an automated fast protein liquid chromatography (FPLC) protocol, capable of obtaining high yields of approximately 20 mg of recombinant protein per liter of bacterium cultures. The purified rEZIKV presented preserved antigenicity and immunogenicity. Our results show that the use of an airlift bioreactor for the production of rEZIKV is ideal for establishing protocols and further research on ZIKV vaccines bioprocess, representing a promising system for the production of a ZIKV envelope recombinant protein-based vaccine candidate.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Monica Bielavsky
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Diego J. B. Orts
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Laboratory of Cardiobiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, São Paulo 04023-062, Brazil
| | - Sergio Araujo
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Patrícia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Renato M. Astray
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
- Multi-Purpose Laboratory Butantan Institute, São Paulo 05503-900, Brazil
| | - Ramendra P. Pandey
- School of Health Sciences and Technology, UPES University, Dehradun 248007, Uttarakhand, India
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences—FCF, University of Campinas—UNICAMP, Campinas 13083-871, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
34
|
Andrade MA, Mottin M, Sousa BKDP, Barbosa JARG, Dos Santos Azevedo C, Lasse Silva C, Gonçalves de Andrade M, Motta FN, Maulay-Bailly C, Amand S, Santana JMD, Horta Andrade C, Grellier P, Bastos IMD. Identification of novel Zika virus NS3 protease inhibitors with different inhibition modes by integrative experimental and computational approaches. Biochimie 2023; 212:143-152. [PMID: 37088408 DOI: 10.1016/j.biochi.2023.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/14/2023] [Accepted: 04/07/2023] [Indexed: 04/25/2023]
Abstract
Zika virus (ZIKV) infection is associated with severe neurological disorders and congenital malformation. Despite efforts to eradicate the disease, there is still neither vaccine nor approved drugs to treat ZIKV infection. The NS2B-NS3 protease is a validated drug target since it is essential to polyprotein virus maturation. In the present study, we describe an experimental screening of 2,320 compounds from the chemical library of the Muséum National d'Histoire Naturelle of Paris on ZIKV NS2B-NS3 protease. A total of 96 hits were identified with 90% or more of inhibitory activity at 10 μM. Amongst the most active compounds, five were analyzed for their inhibitory mechanisms by kinetics assays and computational approaches such as molecular docking. 2-(3-methoxyphenoxy) benzoic acid (compound 945) show characteristics of a competitive inhibition (Ki = 0.49 μM) that was corroborated by its molecular docking at the active site of the NS2B-NS3 protease. Taxifolin (compound 2292) behaves as an allosteric inhibitor whereas 3,8,9-trihydroxy-2-methyl-1H-phenalen-1-one (compound 128), harmol (compound 368) and anthrapurpurin (compound 1499) show uncompetitive inhibitions. These new NS2B-NS3 protease inhibitors are valuable hits to further hit-to-lead optimization.
Collapse
Affiliation(s)
- Milene Aparecida Andrade
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Melina Mottin
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil; Laboratory for Molecular Modeling and Drug Design - LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Bruna K de P Sousa
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Clênia Dos Santos Azevedo
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Camila Lasse Silva
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | | | - Flávia Nader Motta
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil; Faculdade de Ceilândia, Universidade de Brasília, Brasília, Brazil
| | - Christine Maulay-Bailly
- UMR 7245 MCAM, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Séverine Amand
- UMR 7245 MCAM, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Jaime Martins de Santana
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Philippe Grellier
- UMR 7245 MCAM, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France.
| | - Izabela M D Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil.
| |
Collapse
|
35
|
Sankhala RS, Dussupt V, Donofrio G, Gromowski GD, De La Barrera RA, Larocca RA, Mendez-Rivera L, Lee A, Choe M, Zaky W, Mantus G, Jensen JL, Chen WH, Gohain N, Bai H, McCracken MK, Mason RD, Leggat D, Slike BM, Tran U, Jian N, Abbink P, Peterson R, Mendes EA, Freitas de Oliveira Franca R, Calvet GA, Bispo de Filippis AM, McDermott A, Roederer M, Hernandez M, Albertus A, Davidson E, Doranz BJ, Rolland M, Robb ML, Lynch RM, Barouch DH, Jarman RG, Thomas SJ, Modjarrad K, Michael NL, Krebs SJ, Joyce MG. Zika-specific neutralizing antibodies targeting inter-dimer envelope epitopes. Cell Rep 2023; 42:112942. [PMID: 37561630 PMCID: PMC10775418 DOI: 10.1016/j.celrep.2023.112942] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 06/09/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Zika virus (ZIKV) is an emerging pathogen that causes devastating congenital defects. The overlapping epidemiology and immunologic cross-reactivity between ZIKV and dengue virus (DENV) pose complex challenges to vaccine design, given the potential for antibody-dependent enhancement of disease. Therefore, classification of ZIKV-specific antibody targets is of notable value. From a ZIKV-infected rhesus macaque, we identify ZIKV-reactive B cells and isolate potent neutralizing monoclonal antibodies (mAbs) with no cross-reactivity to DENV. We group these mAbs into four distinct antigenic groups targeting ZIKV-specific cross-protomer epitopes on the envelope glycoprotein. Co-crystal structures of representative mAbs in complex with ZIKV envelope glycoprotein reveal envelope-dimer epitope and unique dimer-dimer epitope targeting. All four specificities are serologically identified in convalescent humans following ZIKV infection, and representative mAbs from all four groups protect against ZIKV replication in mice. These results provide key insights into ZIKV-specific antigenicity and have implications for ZIKV vaccine, diagnostic, and therapeutic development.
Collapse
Affiliation(s)
- Rajeshwer S Sankhala
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Vincent Dussupt
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gina Donofrio
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gregory D Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Rafael A De La Barrera
- Pilot Bioproduction Facility, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Letzibeth Mendez-Rivera
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Anna Lee
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Misook Choe
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Weam Zaky
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Grace Mantus
- George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Jaime L Jensen
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Wei-Hung Chen
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Neelakshi Gohain
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Hongjun Bai
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Michael K McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | - David Leggat
- Vaccine Research Center, NIH, Bethesda, MD 20852, USA
| | - Bonnie M Slike
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Ursula Tran
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Ningbo Jian
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rebecca Peterson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erica Araujo Mendes
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - Guilherme Amaral Calvet
- Oswaldo Cruz Foundation, Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, RJ 21040-360, Brazil
| | | | | | | | | | | | | | | | - Morgane Rolland
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Merlin L Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Rebecca M Lynch
- George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Richard G Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Stephen J Thomas
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Kayvon Modjarrad
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Nelson L Michael
- Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Shelly J Krebs
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| | - M Gordon Joyce
- Emerging Infectious Disease Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA; U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
36
|
Kollepara PK, Chisholm RH, Miller JC. Heterogeneity in network structure switches the dominant transmission mode of infectious diseases. PNAS NEXUS 2023; 2:pgad227. [PMID: 37533729 PMCID: PMC10393287 DOI: 10.1093/pnasnexus/pgad227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
Several recent emerging diseases have exhibited both sexual and nonsexual transmission modes (Ebola, Zika, and mpox). In the recent mpox outbreaks, transmission through sexual contacts appears to be the dominant mode of transmission. Motivated by this, we use an SIR-like model to argue that an initially dominant sexual transmission mode can be overtaken by casual transmission at later stages, even if the basic casual reproduction number is less than one. Our results highlight the risk of intervention designs which are informed only by the early dynamics of the disease.
Collapse
Affiliation(s)
- Pratyush K Kollepara
- Department of Mathematical and Physical Sciences, La Trobe University, Plenty Rd and Kingsbury Dr, Melbourne, 3086 VIC, Australia
| | - Rebecca H Chisholm
- Department of Mathematical and Physical Sciences, La Trobe University, Plenty Rd and Kingsbury Dr, Melbourne, 3086 VIC, Australia
- Melbourne School of Population and Global Health, The University of Melbourne, Grattan St, Melbourne, 3010 VIC, Australia
| | | |
Collapse
|
37
|
Ferreira AP, Santana DS, Figueiredo ERL, Simões MC, de Morais DF, Tavares VB, de Sousa JG, Silva MJA, de Campos Gomes F, de Melo Neto JS. Sociodemographic and Clinical Factors for Microcephaly Secondary to Teratogenic Infections in Brazil: An Ecological Study. Viruses 2023; 15:1675. [PMID: 37632018 PMCID: PMC10457789 DOI: 10.3390/v15081675] [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: 06/24/2023] [Revised: 07/17/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Microcephaly is a neurological condition characterized by anomalies in the growth of the cranial circumference. This study aims to examine the association between sociodemographic and clinical variables and the occurrence of secondary microcephaly in newborns in Brazil. It also aims to investigate the association between this congenital anomaly and teratogenic infections. This research adopts an observational approach with an ecological, descriptive, and analytical design. The sample includes infants aged ≤28 days and registered in the country's Live Births Information System from January 2015 to December 2021. Newborns were categorized into G1, consisting of newborns with one of the three infections (Zika, toxoplasmosis, or syphilis), and G2, consisting of newborns with two of the three infections. A total of 1513 samples were analyzed and divided into two groups: one infection (syphilis n = 423; toxoplasmosis n = 295; or Zika n = 739) and two infections (n = 56). The northeastern region of Brazil has the highest prevalence of microcephaly. Regarding the population profile, the Zika virus infection is more common among white mothers, while the syphilis infection is more common among black mothers. Among newborns with microcephaly, boys have a lower prevalence of toxoplasmosis infection, while girls have a lower prevalence of Zika virus infection. This study provides pertinent information on each infection and contributes to the epidemiologic understanding of the association between teratogenic infections and microcephaly.
Collapse
Affiliation(s)
- Arlison Pereira Ferreira
- Unidade de Pesquisa Clínica e Experimental do Sistema Urogenital (UPCEURG), Instituto de Ciências da Saúde, Universidade Federal do Pará (UFPA), Belém 66075-110, PA, Brazil; (A.P.F.); (D.S.S.); (E.R.L.F.); (V.B.T.); (J.G.d.S.)
| | - Davi Silva Santana
- Unidade de Pesquisa Clínica e Experimental do Sistema Urogenital (UPCEURG), Instituto de Ciências da Saúde, Universidade Federal do Pará (UFPA), Belém 66075-110, PA, Brazil; (A.P.F.); (D.S.S.); (E.R.L.F.); (V.B.T.); (J.G.d.S.)
| | - Eric Renato Lima Figueiredo
- Unidade de Pesquisa Clínica e Experimental do Sistema Urogenital (UPCEURG), Instituto de Ciências da Saúde, Universidade Federal do Pará (UFPA), Belém 66075-110, PA, Brazil; (A.P.F.); (D.S.S.); (E.R.L.F.); (V.B.T.); (J.G.d.S.)
| | - Marcelo Coelho Simões
- Programa de Pós-Graduação em Ciências Ambientais, Universidade do Estado do Pará (UEPA), Belém 66095-100, PA, Brazil;
| | - Dionei Freitas de Morais
- Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil;
| | - Victória Brioso Tavares
- Unidade de Pesquisa Clínica e Experimental do Sistema Urogenital (UPCEURG), Instituto de Ciências da Saúde, Universidade Federal do Pará (UFPA), Belém 66075-110, PA, Brazil; (A.P.F.); (D.S.S.); (E.R.L.F.); (V.B.T.); (J.G.d.S.)
| | - Juliana Gonçalves de Sousa
- Unidade de Pesquisa Clínica e Experimental do Sistema Urogenital (UPCEURG), Instituto de Ciências da Saúde, Universidade Federal do Pará (UFPA), Belém 66075-110, PA, Brazil; (A.P.F.); (D.S.S.); (E.R.L.F.); (V.B.T.); (J.G.d.S.)
| | | | | | - João Simão de Melo Neto
- Unidade de Pesquisa Clínica e Experimental do Sistema Urogenital (UPCEURG), Instituto de Ciências da Saúde, Universidade Federal do Pará (UFPA), Belém 66075-110, PA, Brazil; (A.P.F.); (D.S.S.); (E.R.L.F.); (V.B.T.); (J.G.d.S.)
| |
Collapse
|
38
|
Lai ZZ, Shen HH, Lee YM. Inhibitory effect of β-escin on Zika virus infection through the interruption of viral binding, replication, and stability. Sci Rep 2023; 13:10014. [PMID: 37340032 DOI: 10.1038/s41598-023-36871-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
β-Escin is a mixture of triterpenoid saponins extracted from horse chestnut seeds that have diverse pharmacological activities, including anti-inflammation, anti-edematous, venotonic, and antiviral effects. In the clinical setting, β-escin is primarily used to treat venous insufficiency and blunt trauma injuries. The anti-Zika virus (ZIKV) activity of β-escin has not been explored. This study investigated the antiviral efficacy of β-escin on ZIKV and dengue virus (DENV) in vitro and then elucidated the underlying mechanism. The inhibitory effects of β-escin on viral RNA synthesis, protein levels, and infection ability were determined using qRT-PCR, Western blotting, and immunofluorescence assays, respectively. To further characterize how β-escin interferes with the viral life cycle, the time-of-addition experiment was performed. An inactivation assay was performed to determine whether β-escin affects ZIKV virion stability. To broaden these findings, the antiviral effects of β-escin on different DENV serotypes were assessed using dose-inhibition and time-of-addition assays. The results showed that β-escin exhibits anti-ZIKV activity by decreasing viral RNA levels, protein expression, progeny yield, and virion stability. β-Escin inhibited ZIKV infection by disrupting viral binding and replication. Furthermore, β-escin demonstrated antiviral activities against four DENV serotypes in a Vero cell model and prophylactic protection against ZIKV and DENV 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
- Department of Pharmacy Practice, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
| | - Hsin-Hsuen Shen
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, 114, Taiwan
| | - Yen-Mei Lee
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, 114, Taiwan.
| |
Collapse
|
39
|
Hu H, Feng Y, He ML. Targeting Type I Interferon Induction and Signaling: How Zika Virus Escapes from Host Innate Immunity. Int J Biol Sci 2023; 19:3015-3028. [PMID: 37416780 PMCID: PMC10321277 DOI: 10.7150/ijbs.83056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/23/2023] [Indexed: 07/08/2023] Open
Abstract
Zika virus (ZIKV) infection causes neurological disorders and draws great attention. ZIKV infection can elicit a wide range of immune response. Type I interferons (IFNs) as well as its signaling cascade play crucial role in innate immunity against ZIKV infection and in turn ZIKV can antagonize them. ZIKV genome are mainly recognized by Toll-like receptors 3 (TLR3), TLR7/8 and RIG-I-like receptor 1 (RIG-1), which induces the expression of Type I IFNs and interferon-stimulated genes (ISGs). ISGs exert antiviral activity at different stages of the ZIKV life cycle. On the other hand, ZIKV takes multiple strategies to antagonize the Type Ⅰ IFN induction and its signaling pathway to establish a pathogenic infection, especially by using the viral nonstructural (NS) proteins. Most of the NS proteins can directly interact with the factors in the pathways to escape the innate immunity. In addition, structural proteins also participate in the innate immune evasion and activation of antibody-binding of blood dendritic cell antigen 2 (BDCA2) or inflammasome also be used to enhance ZIKV replication. In this review, we summarize the recent findings about the interaction between ZIKV infection and type I IFNs pathways and suggest potential strategies for antiviral drug development.
Collapse
Affiliation(s)
- Huan Hu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yaxiu Feng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
40
|
Lu AY, Gustin A, Newhouse D, Gale M. Viral Protein Accumulation of Zika Virus Variants Links with Regulation of Innate Immunity for Differential Control of Viral Replication, Spread, and Response to Interferon. J Virol 2023; 97:e0198222. [PMID: 37162358 PMCID: PMC10231147 DOI: 10.1128/jvi.01982-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
Asian lineage Zika virus (ZIKV) strains emerged globally, causing outbreaks linked with critical clinical disease outcomes unless the virus is effectively restricted by host immunity. We have previously shown that retinoic acid-inducible gene-I (RIG-I) senses ZIKV to trigger innate immunity to direct interferon (IFN) production and antiviral responses that can control ZIKV infection. However, ZIKV proteins have been demonstrated to antagonize IFN. Here, we conducted in vitro analyses to assess how divergent prototypic ZIKV variants differ in virologic properties, innate immune regulation, and infection outcome. We comparatively assessed African lineage ZIKV/Dakar/1984/ArD41519 (ZIKV/Dakar) and Asian lineage ZIKV/Malaysia/1966/P6740 (ZIKV/Malaysia) in a human epithelial cell infection model. De novo viral sequence determination identified amino acid changes within the ZIKV/Dakar genome compared to ZIKV/Malaysia. Viral growth analyses revealed that ZIKV/Malaysia accumulated viral proteins and genome copies earlier and to higher levels than ZIKV/Dakar. Both ZIKV strains activated RIG-I/IFN regulatory factor (IRF3) and NF-κB pathways to induce inflammatory cytokine expression and types I and III IFNs. However, ZIKV/Malaysia, but not ZIKV/Dakar, potently blocked downstream IFN signaling. Remarkably, ZIKV/Dakar protein accumulation and genome replication were rescued in RIG-I knockout (KO) cells late in acute infection, resulting in ZIKV/Dakar-mediated blockade of IFN signaling. We found that RIG-I signaling specifically restricts viral protein accumulation late in acute infection where early accumulation of viral proteins in infected cells confers enhanced ability to limit IFN signaling, promoting viral replication and spread. Our results demonstrate that RIG-I-mediated innate immune signaling imparts restriction of ZIKV protein accumulation, which permits IFN signaling and antiviral actions controlling ZIKV infection. IMPORTANCE ZIKV isolates are classified under African or Asian lineages. Infection with emerging Asian lineage-derived ZIKV strains is associated with increased incidence of neurological symptoms that were not previously reported during infection with African or preemergent Asian lineage viruses. In this study, we utilized in vitro models to compare the virologic properties of and innate immune responses to two prototypic ZIKV strains from distinct lineages: African lineage ZIKV/Dakar and Asian lineage ZIKV/Malaysia. Compared to ZIKV/Dakar, ZIKV/Malaysia accumulates viral proteins earlier, replicates to higher levels, and robustly blocks IFN signaling during acute infection. Early accumulation of ZIKV/Malaysia NS5 protein confers enhanced ability to antagonize IFN signaling, dampening innate immune responses to promote viral spread. Our data identify the kinetics of viral protein accumulation as a major regulator of host innate immunity, influencing host-mediated control of ZIKV replication and spread. Importantly, these findings provide a novel framework for evaluating the virulence of emerging variants.
Collapse
Affiliation(s)
- Amy Y. Lu
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Andrew Gustin
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Daniel Newhouse
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
41
|
Pereira RS, Santos FCP, Campana PRV, Costa VV, de Pádua RM, Souza DG, Teixeira MM, Braga FC. Natural Products and Derivatives as Potential Zika virus Inhibitors: A Comprehensive Review. Viruses 2023; 15:v15051211. [PMID: 37243296 DOI: 10.3390/v15051211] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/30/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Zika virus (ZIKV) is an arbovirus whose infection in humans can lead to severe outcomes. This article reviews studies reporting the anti-ZIKV activity of natural products (NPs) and derivatives published from 1997 to 2022, which were carried out with NPs obtained from plants (82.4%) or semisynthetic/synthetic derivatives, fungi (3.1%), bacteria (7.6%), animals (1.2%) and marine organisms (1.9%) along with miscellaneous compounds (3.8%). Classes of NPs reported to present anti-ZIKV activity include polyphenols, triterpenes, alkaloids, and steroids, among others. The highest values of the selectivity index, the ratio between cytotoxicity and antiviral activity (SI = CC50/EC50), were reported for epigallocatechin gallate (SI ≥ 25,000) and anisomycin (SI ≥ 11,900) obtained from Streptomyces bacteria, dolastane (SI = 1246) isolated from the marine seaweed Canistrocarpus cervicorni, and the flavonol myricetin (SI ≥ 862). NPs mostly act at the stages of viral adsorption and internalization in addition to presenting virucidal effect. The data demonstrate the potential of NPs for developing new anti-ZIKV agents and highlight the lack of studies addressing their molecular mechanisms of action and pre-clinical studies of efficacy and safety in animal models. To the best of our knowledge, none of the active compounds has been submitted to clinical studies.
Collapse
Affiliation(s)
- Rosângela Santos Pereira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Françoise Camila Pereira Santos
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | | | - Vivian Vasconcelos Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Rodrigo Maia de Pádua
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Daniele G Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Fernão Castro Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
42
|
Leiva S, Bugnon Valdano M, Gardiol D. Unravelling the epidemiological diversity of Zika virus by analyzing key protein variations. Arch Virol 2023; 168:115. [PMID: 36943525 DOI: 10.1007/s00705-023-05726-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/19/2023] [Indexed: 03/23/2023]
Abstract
The consequences of Zika virus (ZIKV) infections were limited to sporadic mild diseases until almost a decade ago, when epidemic outbreaks took place, with quick spread into the Americas. Simultaneously, novel severe neurological manifestations of ZIKV infections were identified, including congenital microcephaly. However, why the epidemic strains behave differently is not yet completely understood, and many questions remain about the actual significance of genetic variations in the epidemiology and biology of ZIKV. In this study, we analysed a large number of viral sequences to identify genes with different levels of variability and patterns of genomic variations that could be associated with ZIKV diversity. We compared numerous epidemic strains with pre-epidemic strains, using the BWA-mem algorithm, and we also examined specific variations among the epidemic ZIKV strains derived from microcephaly cases. We identified several viral genes with dissimilar mutation rates among the ZIKV strain groups and novel protein variation profiles that might be associated with epidemiological particularities. Finally, we assessed the impact of the detected changes on the structure and stability of the NS1, NS5, and E proteins using the I-TASSER, trRosetta, and RaptorX modelling algorithms, and we found some interesting variations that might help to explain the heterogeneous features of the diverse ZIKA strains. This work contributes to the identification of genetic differences in the ZIKV genome that might have a phenotypic impact, providing a basis for future experimental analysis to elucidate the genetic causes of the recent ZIKV emergency.
Collapse
Affiliation(s)
- Santiago Leiva
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Marina Bugnon Valdano
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina.
| | - Daniela Gardiol
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina.
| |
Collapse
|
43
|
Bhardwaj U, Singh SK. Zika virus NS1 suppresses VE-cadherin via hsa-miR-29b-3p/DNMT3b/MMP-9 pathway in human brain microvascular endothelial cells. Cell Signal 2023; 106:110659. [PMID: 36948479 DOI: 10.1016/j.cellsig.2023.110659] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/14/2023] [Accepted: 03/19/2023] [Indexed: 03/24/2023]
Abstract
Zika virus infection has been reported to cause microcephaly in newborns. ZIKV exploits various strategies to cross the blood-brain barrier. ZIKV NS1 may compromise the barrier integrity of endothelial cells by regulating expression of junctional proteins. MicroRNAs play an important role in post-transcriptional gene regulations. We demonstrated that ZIKV-NS1 affected the adherence junction protein in human brain microvascular endothelial cells via hsa-miR-29b-3p/DNMT3b/MMP-9 pathway. The hCMEC/D3 cells were exposed to ZIKV-NS1 with different doses (500 ng/mL and 1000 ng/mL) for 24 h. The expression pattern of DNTM3b, MMP-9, and VE-cadherin were studied using immunoblotting and the distribution of DNMT3b and MMP-9 were studied using immunofluorescence. The quantification of hsa-miR-29b-3p was done through qRT-PCR. Direct regulation of DNMT3b by hsa-miR-29b-3p was demonstrated by overexpression of hsa-miR-29b-3p using hsa-miR-29b-3p mimic, and knockdown of hsa-miR-29b-3p by using hsa-miR-29b-3p inhibitors. The ZIKV-NS1 affected the barrier function of endothelial cells through the increased expression of hsa-miR29b-3p, which suppressed the DNMT3b, thus enhanced expression of MMP-9, which finally suppressed the expression of VE-cadherin. These findings suggested that ZIKV-NS1 alters the expression of Adherens Junction protein in human brain microvascular endothelial cells through hsa-miR-29b-3p/DNMT3b/MMP-9 pathway, which compromised the barrier function of human brain microvascular endothelial cells.
Collapse
Affiliation(s)
- Utkarsh Bhardwaj
- Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Sunit K Singh
- Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
44
|
Standardized evaluation of Zika nucleic acid tests used in clinical settings and blood screening. PLoS Negl Trop Dis 2023; 17:e0011157. [PMID: 36930653 PMCID: PMC10072466 DOI: 10.1371/journal.pntd.0011157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 04/04/2023] [Accepted: 02/09/2023] [Indexed: 03/18/2023] Open
Abstract
Early detection of Zika virus (ZIKV) transmission within geographic regions informs implementation of community mitigation measures such as vector reduction strategies, travel advisories, enhanced surveillance among pregnant women, and possible implementation of blood and organ donor screening or deferral. Standardized, comparative assessments of ZIKV assay and testing lab performance are important to develop optimal approaches to ZIKV diagnostic testing and surveillance. We conducted an expanded blinded panel study to characterize and compare the analytical performance of fifteen diagnostic and blood screening ZIKV NAT assays, including detection among single- and multiplex assays detecting ZIKV, dengue virus (DENV) and chikungunya virus (CHIKV). A 300 member blinded panel was constructed, consisting of 11 serial half-log dilutions ranging from ~104 to 10-1 genome equivalents/mL in 25 replicates each of the Tahitian Asian ZIKV isolate in ZIKV-negative human serum. Additionally, clinical samples from individuals with DENV-like syndrome or suspected ZIKV infection in Brazil were evaluated. The majority of assays demonstrated good specificity. Analytical sensitivities varied 1-2 logs, with a substantially higher limit of detection (LOD) in one outlier. Similar analytical sensitivity for ZIKV RNA detection in singleplex and multiplex assays of the Grifols and ThermoFisher tests were observed. Coefficient of Assay Efficiency (CE), calculated to characterize assays' RNA extraction and amplification efficiency, ranged from 0.13 for the Certest VIASURE multiplex and 0.75 for the Grifols multiplex assays. In general, assays using transcription mediated amplification (TMA) technology had greater CE compared to assays using conventional PCR technology. Donor screening NAT assays were significantly more sensitive than diagnostic RT-qPCR assays, primarily attributable to higher sample input volumes. However, ideal assays to maximize sensitivity and throughput may not be a viable option in all contexts, with other factors such as cost, instrumentation, and regulatory approval status influencing assay availability and selection, particularly in resource constrained settings.
Collapse
|
45
|
Mac PA, Kroeger A, Daehne T, Anyaike C, Velayudhan R, Panning M. Zika, Flavivirus and Malaria Antibody Cocirculation in Nigeria. Trop Med Infect Dis 2023; 8:tropicalmed8030171. [PMID: 36977172 PMCID: PMC10059970 DOI: 10.3390/tropicalmed8030171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Introduction. Arboviruses and malaria pose a growing threat to public health, affecting not only the general population but also immunocompromised individuals and pregnant women. Individuals in vulnerable groups are at a higher risk of severe complications from the co-circulation and transmission of ZIKV, malaria, and FLAVI fever. In sub-Saharan countries, such as Nigeria, these mosquito-borne infections have clinical presentations that overlap with other diseases (dengue, West Nile virus, and Japanese encephalitis, chikungunya, and O’nyong o’nyong virus), making them a diagnostic challenge for clinicians in regions where they co-circulate. Vertical transmission can have a devastating impact on maternal health and fetal outcomes, including an increased risk of fetal loss and premature birth. Despite the global recognition of the burden of malaria and arboviruses, particularly ZIKV and other flaviviruses, there is limited data on their prevalence in Nigeria. In urban settings, where these diseases are endemic and share common biological, ecological, and economic factors, they may impact treatment outcomes and lead to epidemiological synergy. Hence, it is imperative to conduct sero-epidemiological and clinical studies to better understand the disease burden and hidden endemicity, thereby enabling improved prevention and clinical management. Method. Serum samples collected from outpatients between December 2020 and November 2021 in three regions of Nigeria were tested for the presence of IgG antibody seropositivity against ZIKV and FLAVI using immunoblot serological assay. Results. The overall cohort co-circulation antibody seropositivity of ZIKV, FLAVI and malaria was 24.0% (209/871). A total of 19.2% (167/871) of the study participants had ZIKV-seropositive antibodies and 6.2% (54/871) were FLAVI-seropositive, while 40.0% (348/871) of the subjects had malaria parasite antigens. Regional analysis revealed that participants from the southern region had the highest antibody seropositivity against ZIKV (21.7% (33/152)) and FLAVI (8.6% (13/152)), whereas those from the central region had a higher malaria parasite antigen (68.5% (287/419)). Conclusions. This study represents the largest comparative cross-sectional descriptive sero-epidemiological investigation of ZIKV-FLAVI and malaria cocirculation in Nigeria. The findings of this study revealed increased antibody seropositivity, hidden endemicity, and the burden of ZIKV, FLAVI, and malaria co-circulating in Nigeria.
Collapse
Affiliation(s)
- Peter Asaga Mac
- Institute of Virologie Hermann Herder Strabe, Universitatsklinikum Freiburg, 79104 Freiburg, Germany
- Correspondence:
| | - Axel Kroeger
- Centre for Medicine and Society, University of Freiburg, 79085 Freiburg, Germany
| | - Theo Daehne
- Institute of Virologie Hermann Herder Strabe, Universitatsklinikum Freiburg, 79104 Freiburg, Germany
| | | | | | - Marcus Panning
- Institute of Virologie Hermann Herder Strabe, Universitatsklinikum Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
46
|
Astrocytes in the pathophysiology of neuroinfection. Essays Biochem 2023; 67:131-145. [PMID: 36562155 DOI: 10.1042/ebc20220082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Key homeostasis providing cells in the central nervous system (CNS) are astrocytes, which belong to the class of cells known as atroglia, a highly heterogeneous type of neuroglia and a prominent element of the brain defence. Diseases evolve due to altered homeostatic state, associated with pathology-induced astroglia remodelling represented by reactive astrocytes, astroglial atrophy and astrodegeneration. These features are hallmarks of most infectious insults, mediated by bacteria, protozoa and viruses; they are also prominent in the systemic infection. The COVID-19 pandemic revived the focus into neurotropic viruses such as SARS-CoV2 (Coronaviridae) but also the Flaviviridae viruses including tick-borne encephalitis (TBEV) and Zika virus (ZIKV) causing the epidemic in South America prior to COVID-19. Astrocytes provide a key response to neurotropic infections in the CNS. Astrocytes form a parenchymal part of the blood-brain barrier, the site of virus entry into the CNS. Astrocytes exhibit aerobic glycolysis, a form of metabolism characteristic of highly morphologically plastic cells, like cancer cells, hence a suitable milieu for multiplication of infectious agent, including viral particles. However, why the protection afforded by astrocytes fails in some circumstances is an open question to be studied in the future.
Collapse
|
47
|
Growth Velocity and Nutritional Status in Children Exposed to Zika Virus during Pregnancy from Amazonas Cohort, Brazil. Viruses 2023; 15:v15030662. [PMID: 36992371 PMCID: PMC10056230 DOI: 10.3390/v15030662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/18/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
The high incidence of Zika virus (ZIKV) infection in the period of 2015–2016 in Brazil may have affected linear height growth velocity (GV) in children exposed in utero to ZIKV. This study describes the growth velocity and nutritional status based on the World Organization (WHO) standards of children exposed to ZIKV during pregnancy and followed up in a tertiary unit, a reference for tropical and infectious diseases in the Amazon. Seventy-one children born between March 2016 and June 2018 were monitored for anthropometric indices: z-score for body mass index (BMI/A); weight (W/A); height (H/A) and head circumference (HC/A); and growth velocity. The mean age at the last assessment was 21.1 months (SD ± 8.93). Four children had congenital microcephaly and severe neurological impairment. The other 67 were non-microcephalic children (60 normocephalic and 7 macrocephalic); of these; 24.2% (16 children) had neurological alterations, and 28.8% (19 children) had altered neuropsychomotor development. Seventeen (24.2%) children had inadequate GV (low growth velocity). The frequencies of low growth among microcephalic and non-microcephalic patients are 25% (1 of 4 children) and 23.9% (16 of 67 children); respectively. Most children had normal BMI/A values during follow-up. Microcephalic patients showed low H/A and HC/A throughout the follow-up, with a significant reduction in the HC/A z-score. Non-microcephalic individuals are within the regular ranges for H/A; HC/A; and W/A, except for the H/A score for boys. This study showed low growth velocity in children with and without microcephaly, highlighting the need for continuous evaluation of all children born to mothers exposed to ZIKV during pregnancy.
Collapse
|
48
|
Santos CNO, Magalhães LS, Fonseca ABDL, Bispo AJB, Porto RLS, Alves JC, Dos Santos CA, de Carvalho JV, da Silva AM, Teixeira MM, de Almeida RP, Dos Santos PL, de Jesus AR. Association between genetic variants in TREM1, CXCL10, IL4, CXCL8 and TLR7 genes with the occurrence of congenital Zika syndrome and severe microcephaly. Sci Rep 2023; 13:3466. [PMID: 36859461 PMCID: PMC9975867 DOI: 10.1038/s41598-023-30342-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Congenital Zika syndrome (CZS) is a cluster of malformations induced by Zika virus (ZIKV) infection and the underline mechanisms involved in its occurrence are yet not fully understood. Along with epidemiological and environmental factors, the genetic host factors are suggested as important to the CZS occurrence and development, however, few studies have evaluated this. This study enrolled a total of 245 individuals in a case-control association study compound a cohort of high specific interest constituted by 75 mothers who had delivered CZS infants, their 76 infants, and 47 mothers that had delivered healthy infants, and their 47 infants. Sixteen single-nucleotide polymorphisms on TREM1, CXCL10, IL4, CXCL8, TLR3, TLR7, IFNR1, CXCR1, IL10, CCR2 and CCR5 genes were genotyped to investigate their association as risk factors to CZS. The results show an association between C allele at TREM1 rs2234246 and C allele at IL4 rs224325 in mothers infected with ZIKV during pregnancy, with the increased susceptibility to CZS occurrence in their infants and the SNP CXCL8 rs4073 and the G allele at CXCL10 rs4508917 with presence of CZS microcephaly in the infants. Furthermore, the T allele at CXCL8 rs4073 and TRL7 rs179008 SNPs were associated with the severity of microcephaly in children with CZS. These results suggest that these polymorphisms in genes of innate immune responses addressed here are associated to increased risk of occurrence and severity of CZS in pregnant mothers infected with ZIKV and their CZS infants.
Collapse
Affiliation(s)
- Camilla Natália Oliveira Santos
- Immunology and Molecular Biology Laboratory and Graduate Program in Health Sciences, University Hospital of Federal University of Sergipe, Aracaju, Brazil.
| | - Lucas Sousa Magalhães
- Immunology and Molecular Biology Laboratory and Graduate Program in Health Sciences, University Hospital of Federal University of Sergipe, Aracaju, Brazil
- Sector of Parasitology and Pathology, Biological and Health Sciences Institute, Federal University of Alagoas, Maceió, Brazil
| | | | | | | | - Juliana Cardoso Alves
- Immunology and Molecular Biology Laboratory and Graduate Program in Health Sciences, University Hospital of Federal University of Sergipe, Aracaju, Brazil
| | | | | | - Angela Maria da Silva
- Immunology and Molecular Biology Laboratory and Graduate Program in Health Sciences, University Hospital of Federal University of Sergipe, Aracaju, Brazil
- Department of Medicine of University Hospital, Federal University of Sergipe, Aracaju, Brazil
| | | | - Roque Pacheco de Almeida
- Immunology and Molecular Biology Laboratory and Graduate Program in Health Sciences, University Hospital of Federal University of Sergipe, Aracaju, Brazil
- Department of Medicine of University Hospital, Federal University of Sergipe, Aracaju, Brazil
| | - Priscila Lima Dos Santos
- Immunology and Molecular Biology Laboratory and Graduate Program in Health Sciences, University Hospital of Federal University of Sergipe, Aracaju, Brazil
| | - Amélia Ribeiro de Jesus
- Immunology and Molecular Biology Laboratory and Graduate Program in Health Sciences, University Hospital of Federal University of Sergipe, Aracaju, Brazil
- Department of Medicine of University Hospital, Federal University of Sergipe, Aracaju, Brazil
| |
Collapse
|
49
|
Huo HF, Fu T, Xiang H. Dynamics and optimal control of a Zika model with sexual and vertical transmissions. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:8279-8304. [PMID: 37161197 DOI: 10.3934/mbe.2023361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
A new transmission model of Zika virus with three transmission routes including human transmission by mosquito bites, sexual transmission between males and females and vertical transmission is established. The basic reproduction number $ R_{0} $ is derived. When $ R_{0} < 1 $, it is proved that the disease-free equilibrium is globally stable. Furthermore, the optimal control and mitigation methods for transmission of Zika virus are deduced and explored. The MCMC method is used to estimate the parameters and the reasons for the deviation between the actual infection cases and the simulated data are discussed. In addition, different strategies for controlling the spread of Zika virus are simulated and studied. The combination of mosquito control strategies and internal human control strategies is the most effective way in reducing the risk of Zika virus infection.
Collapse
Affiliation(s)
- Hai-Feng Huo
- Department of Applied Mathematics, Lanzhou University of Technology, Lanzhou, Gansu 730050, China
| | - Tian Fu
- Department of Applied Mathematics, Lanzhou University of Technology, Lanzhou, Gansu 730050, China
| | - Hong Xiang
- Department of Applied Mathematics, Lanzhou University of Technology, Lanzhou, Gansu 730050, China
| |
Collapse
|
50
|
Safadi DE, Lebeau G, Lagrave A, Mélade J, Grondin L, Rosanaly S, Begue F, Hoareau M, Veeren B, Roche M, Hoarau JJ, Meilhac O, Mavingui P, Desprès P, Viranaïcken W, Krejbich-Trotot P. Extracellular Vesicles Are Conveyors of the NS1 Toxin during Dengue Virus and Zika Virus Infection. Viruses 2023; 15:v15020364. [PMID: 36851578 PMCID: PMC9965858 DOI: 10.3390/v15020364] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Extracellular vesicles (EVs), produced during viral infections, are of emerging interest in understanding infectious processes and host-pathogen interactions. EVs and exosomes in particular have the natural ability to transport nucleic acids, proteins, and other components of cellular or viral origin. Thus, they participate in intercellular communication, immune responses, and infectious and pathophysiological processes. Some viruses are known to hijack the cell production and content of EVs for their benefit. Here, we investigate whether two pathogenic flaviviruses i.e., Zika Virus (ZIKV) and Dengue virus (DENV2) could have an impact on the features of EVs. The analysis of EVs produced by infected cells allowed us to identify that the non-structural protein 1 (NS1), described as a viral toxin, is associated with exosomes. This observation could be confirmed under conditions of overexpression of recombinant NS1 from each flavivirus. Using different isolation methods (i.e., exosome isolation kit, size exclusion chromatography, Polyethylene Glycol enrichment, and ELISA capture), we showed that NS1 was present as a dimer at the surface of excreted exosomes, and that this association could occur in the extracellular compartment. This finding could be of major importance in a physiological context. Indeed, this capacity of NS1 to address EVs and its implication in the pathophysiology during Dengue or Zika diseases should be explored. Furthermore, exosomes that have demonstrated a natural capacity to vectorize NS1 could serve as useful tools for vaccine development.
Collapse
Affiliation(s)
- Daed El Safadi
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Grégorie Lebeau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Alisé Lagrave
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Julien Mélade
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Lauriane Grondin
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Sarah Rosanaly
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Floran Begue
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Mathilde Hoareau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Bryan Veeren
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Marjolaine Roche
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Jean-Jacques Hoarau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Olivier Meilhac
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Patrick Mavingui
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Philippe Desprès
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Wildriss Viranaïcken
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
- Correspondence: (W.V.); (P.K.-T.)
| | - Pascale Krejbich-Trotot
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
- Correspondence: (W.V.); (P.K.-T.)
| |
Collapse
|