1
|
Lopez S, Komarova NL. An optimal network that promotes the spread of an advantageous variant in an SIR epidemic. J Theor Biol 2025; 605:112095. [PMID: 40107346 DOI: 10.1016/j.jtbi.2025.112095] [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: 11/14/2024] [Revised: 02/01/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
In the course of epidemics, the pathogen may mutate to acquire a higher fitness. At the same time, such a mutant is automatically in an unfavorable position because the resident virus has a head start in accessing the pool of susceptible individuals. We considered a class of tunable small-world networks, where a parameter, p (the rewiring probability), characterizes the prevalence of non-local connections, and we asked, whether the underlying network can influence the fate of a mutant virus. Under an SIR model, we considered two measures of mutant success: the expected height of the peak of mutant infected individuals, and the total number of recovered from mutant individuals at the end of the epidemic. Using these measures, we have found the existence of an optimal (for an advantageous mutant virus) rewiring probability that promotes a larger infected maximum and a larger total recovered population corresponding to the advantageous pathogen strain. This optimal rewiring probability decreases as mean degree and the infectivity of the wild type are increased, and it increases with the mutant advantage. The non-monotonic behavior of the advantageous mutant as a function of rewiring probability may shed light into some of the complex patterns in the size of mutant peaks experienced by different countries during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Samuel Lopez
- Department of Mathematics, University of California Irvine, Irvine, CA, 92617, United States
| | - Natalia L Komarova
- Department of Mathematics, University of California San Diego, La Jolla, CA, 92093, United States.
| |
Collapse
|
2
|
Kumar N. P, P.M. A, Saini P, S. AR, Suresh A, Samuel P, R. B, Mathew J, T. S, K.P. A, K. H. K. R, A. V, A. S, T. B, Kumar A. Is the Zika virus re-emerging as a distinct genetic lineage in India? Access Microbiol 2025; 7:000857.v4. [PMID: 40376273 PMCID: PMC12078788 DOI: 10.1099/acmi.0.000857.v4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 02/11/2025] [Indexed: 05/18/2025] Open
Abstract
An outbreak of Zika fever occurred in Thiruvananthapuram City, Kerala, India, during 2021. At the request of the Kerala state health administration, we investigated the same, towards proposing requisite containment strategies for the disease outbreak. Epidemiological investigations indicated a clustering pattern of Zika fever cases with the presumed index case from a multi-speciality hospital in the city. Preliminary reports on the same had been already reported elsewhere during 2021. Further, entomological surveys carried out evinced the predominant mosquito species in the city, viz. Aedes albopictus (65.55%), Aedes aegypti (22.0%) and Aedes vittatus (12.0%) were naturally infected with Zika virus (ZIKV), the minimum infection rates being 17.9, 7.8 and 3.6, respectively. Also, trans-ovarian transmission was recorded in both Ae. aegypti and Ae. albopictus. This is the first report on the detection of ZIKV from Ae. albopictus in India. Analysis of phylogenetically informative genes of the ZIKV genome indicated the emergence of a distinct lineage of the Asian strain of virus, with five unique non-synonymous mutations, viz. 'A22T' and 'I160M' (pre-membrane) and 'D348N', 'T470A' and 'V473L' (envelope), that were involved in the outbreak. The altered gene expression pattern and evolutionary implications of these unique mutations remain to be investigated. Genetic analysis of the virus isolates from this and other investigations carried out on sporadic outbreaks of ZIKV in the country subsequently indicated that ZIKV is re-emerging as a distinct genetic lineage in India. These findings and other recent reports on ZIKV outbreaks warrant an urgent need for a systematic countrywide surveillance strategy, towards the prevention/preparedness/containment of a massive outbreak of this emerging neurovirulent arboviral disease.
Collapse
Affiliation(s)
- Pradeep Kumar N.
- ICMR-Vector Control Research Centre, Field Station, Kottayam, Kerala, India
| | - Ajithlal P.M.
- ICMR-Vector Control Research Centre, Field Station, Kottayam, Kerala, India
| | - Prasanta Saini
- ICMR-Vector Control Research Centre, Indira Nagar, Puducherry, India
| | - Aiswarya R. S.
- ICMR-Vector Control Research Centre, Field Station, Kottayam, Kerala, India
| | - Abidha Suresh
- ICMR-Vector Control Research Centre, Field Station, Kottayam, Kerala, India
| | - Philip Samuel
- ICMR-Vector Control Research Centre, Field Station, Madurai, India
| | | | - Jessu Mathew
- ICMR-Vector Control Research Centre, Field Station, Kottayam, Kerala, India
| | - Sonia T.
- ICMR-Vector Control Research Centre, Field Station, Kottayam, Kerala, India
| | - Amju K.P.
- ICMR-Vector Control Research Centre, Field Station, Kottayam, Kerala, India
| | - Raju K. H. K.
- ICMR-Vector Control Research Centre, Indira Nagar, Puducherry, India
| | - Veerapathiran A.
- ICMR-Vector Control Research Centre, Field Station, Madurai, India
| | - Selvam A.
- ICMR-Vector Control Research Centre, Field Station, Madurai, India
| | - Balaji T.
- ICMR-Vector Control Research Centre, Field Station, Madurai, India
| | - Ashwani Kumar
- ICMR-Vector Control Research Centre, Indira Nagar, Puducherry, India
| |
Collapse
|
3
|
Kimita G, Collins J, Mutai B, Omuseni E, Masakhwe C, Ocholla S, Lemtudo A, Awinda G, Githii R, Kellar G, Waitumbi J. Detection of zika virus disease caused by the Asian lineage of the zika virus at Kenya's Lamu island. Int J Infect Dis 2025; 154:107862. [PMID: 40010630 DOI: 10.1016/j.ijid.2025.107862] [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: 11/25/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025] Open
Abstract
OBJECTIVES We report the first confirmed cases of Zika virus (ZIKV) disease (ZVD) in Kenya and provide the associated genomic sequences. METHODS The study is part of a prospective febrile illness surveillance study in Kenya. A cluster of ZVD cases was identified in patients with fever presenting at King Fahd Referral Hospital, Lamu Island. Clinical signs resembled those caused by dengue or malaria and included fever, chills, muscle and joint pains, and headache. The Biofire Film Array Global fever panel (BioFire Diagnostics, UT, USA) identified ZIKV in three of five blood samples. RNA extracted from plasma was used for sequence library preparation and subsequent viral enrichment using the VirCapSeq-VERT probe set designed at the Center for Infection and Immunity, Columbia University. Enriched libraries were sequenced on the NextSeq 2000 (Illumina, San Diego, CA, USA). Assembled genomes were classified into lineages and a spatio-temporal phylogeny inferred with Nextstrain v8.5.3. RESULTS Assembled genomes produced contigs covering between 3363 and 10,950 nt that mapped to the Asian lineage of ZIKV and branched with genomes from India. Mutation profiling revealed viral strains with potentially enhanced transmission and immune evasion capabilities. CONCLUSIONS The presence of the more transmissible Asian lineage ZIKV that has previously caused large outbreaks of ZVD in the Americas raises significant public health concern.
Collapse
Affiliation(s)
- Gathii Kimita
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - John Collins
- Global Alliance for Preventing Pandemics, Columbia University Mailman School of Public Health, New York, USA
| | - Beth Mutai
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - Esther Omuseni
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - Clement Masakhwe
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - Stephen Ocholla
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - Allan Lemtudo
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - George Awinda
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - Rachel Githii
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - Gerald Kellar
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya
| | - John Waitumbi
- Kenya Medical Research Institute/Walter Reed Army Institute of Research-Africa, Basic Science Laboratory, Kisumu, Kenya.
| |
Collapse
|
4
|
Deshpande GR, Sapkal GN, Salunke A, Gunjikar R, Tadkalkar N, Shinde P, Daga N, Gopale M, Ramdasi A, Hundekar S, Lole K, Roy RR, Jenish JA, Srivastava R, Parmar S, Pawara P, Jarande K, Vidhate S, Khutwad K. An outbreak of Zika virus in western India in the metropolis of Pune in the monsoon of 2024. J Infect Public Health 2025; 18:102720. [PMID: 40043420 DOI: 10.1016/j.jiph.2025.102720] [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: 11/07/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Sporadic cases and rare outbreaks of Zika Virus (ZIKV) have been reported in India since 2016. Between June to early September 2024; the city of Pune in western India was affected with a large outbreak of ZIKV. Here we report the analysis of epidemiological; clinical and genomic characteristics of the outbreak. METHODS Suspected ZIKV cases and pregnant women within 3 km radius of ZIKV positive cases and household contacts were included in the study. The study area included parts of the Pune metropolitan region. ZIKV RNA in serum and urine samples was detected by a single-plex Realtime RT-PCR assay. Representative samples were characterized by whole-genome sequencing. RESULTS Among clinically suspected ZIKV cases 63/433 (14.55 %); and 45/1615 (2.78 %) pregnant women surveyed; were found positive for acute ZIKV infection. Majority of positive cases were asymptomatic or had mild symptoms. Two cases reported complications of pyelonephritis and encephalitis respectively. There was no evidence of microcephaly in the fetuses of the pregnant women surveyed. The ZIKV strains were found to belong to the Asian lineage and clustered close to the Rajasthan strain reported previously in India. Mutation linked with microcephaly was not found in these sequences. Follow-up of pregnant women till October 2024 indicated 22 % (10/45) of women delivered healthy babies. CONCLUSION The ZIKV strain associated with the present outbreak did not have mutations linked with microcephaly; but had mutations linked with enhanced transmission. Symptoms such as thrombocytopenia; bleeding manifestations; retro-orbital-pain; in a small subset of patients were unique; which were not reported in previous outbreaks in India. The findings highlight the urgent need for enhanced surveillance to plan strategies for public health measures to control the disease spread. Study highlights the need for monitoring the mutations and follow up of ZIKV infected pregnant women and their children to confirm absence of congenital anomalies.
Collapse
Affiliation(s)
- Gururaj Rao Deshpande
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Gajanan N Sapkal
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India.
| | - Asha Salunke
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Rashmi Gunjikar
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Nitali Tadkalkar
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Pradnya Shinde
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Nidhi Daga
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Manjusha Gopale
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Ashwini Ramdasi
- Hepatitis Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Supriya Hundekar
- Hepatitis Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Kavita Lole
- Hepatitis Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Rohan Raj Roy
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Jose Antony Jenish
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Rashi Srivastava
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Shivani Parmar
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Pooja Pawara
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Kajal Jarande
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Shankar Vidhate
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| | - Kirtee Khutwad
- Diagnostic Virology Group, ICMR-National Institute of Virology, MCC 130/1 Sus Road, Pashan, Pune, Maharashtra 411021, India
| |
Collapse
|
5
|
Shang J, Zhou C, He M, Huang XY, Qin CF, Wu A. Mutation S139N on Zika virus prM protein shifts immune response from Asian to contemporary strain. Brain Behav Immun 2025; 126:247-259. [PMID: 39986659 DOI: 10.1016/j.bbi.2025.02.012] [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: 08/11/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025] Open
Abstract
Zika virus (ZIKV) has been associated with neurological diseases like microcephaly and Guillain-Barré syndrome. The S139N single mutation on the prM protein of the FSS13025 Asian strain increases the mortality rate in mice. Therefore, it is a valuable tool for studying the impact of immune responses on neural damage. Here, we used single-cell sequencing technology to systematically assess the immune response induced by three ZIKV strains: Asian ancestral strain FSS13025/2010, FSS13025 strain with S139N mutation (FSS13025-S139N), and contemporary strain GZ01/2016. By infecting 1-day-old mice, we observed that the immune spectrum elicited by FSS13025-S139N mutant resembled that induced by the contemporary strain. The FSS13025-S139N strain induces the proliferation of inflammatory microglial cells earlier than the FSS13025 strain, similar to GZ01. A specific cell cluster, Microglia_Ccr7, was induced by the S139N mutant strain and GZ01 strain, which suppresses T cell activation through the PDCD1LG2-PDCD1 signaling pathway. Furthermore, the proliferation of CD8+ T cells was weakened and prolonged in S139N strain-infected samples. Finally, we found that the S139N mutant strain causes more apoptosis of neurons compared to the FSS13025 strain. These results indicate that the S139N mutation plays an important role in the immune response pattern of ZIKV and prolongs the duration of neuroinflammation.
Collapse
Affiliation(s)
- Jingzhe Shang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123 Jiangsu, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Chao Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Mengjiao He
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Xing-Yao Huang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences (AMMS), Beijing 100071, China.
| | - Aiping Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123 Jiangsu, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
| |
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
|
Roy RR, Tadkalkar N, Deshpande GR, Atre NM, Shil P, Sapkal G. Identification of B-cell epitopes of Indian Zika virus strains using immunoinformatics. Front Immunol 2025; 16:1534737. [PMID: 40083545 PMCID: PMC11903408 DOI: 10.3389/fimmu.2025.1534737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction The Zika virus is an emerging Flavivirus known to cause Zika infection in humans. It is associated with severe health problems such as microcephaly and Guillain-Barré syndrome post the Brazilian epidemic in 2015-16. The spread of the Zika virus to the Asian subcontinent, especially to India is a matter of great concern. Two recent co-circulating Indian Zika virus strains such as Rajasthan and Maharashtra detected in 2018 and 2021 were studied to identify B-cell epitopes in the envelope and non-structural 1 protein as these epitopes are major indicators of robust humoral immune response. The study aimed at identifying novel epitopes, followed by molecular docking with potent Zika virus-specific monoclonal antibodies. The novel epitopes identified in this study shall be essential in designing multi-epitope vaccines capable of inducing antibody response against Zika virus infection. Methods ABCpred, BepiPred 2.0 and Kolaskar-Tongaonkar methods were used for predicting the linear B-cell epitopes, and Discotope 2.0 and ElliPro were used for the prediction of conformational epitopes. Linear epitopes were further checked for protective antigenicity, allergenicity and toxicity. Based on the stringent study design criteria, only the novel epitopes were considered for molecular docking with complementary determining regions of potent Zika virus-specific monoclonal antibodies. Results Nineteen linear and five conformational epitopes were shortlisted based on protective potential, non-allergic and non-toxic properties for Zika virus E protein, from which nine linear and three conformational epitopes were identified as novel. Molecular docking studies revealed that the novel linear epitopes, one each from EDIII, EDII, EDI and EDI/DIII hinge were involved in epitope-CDR interactions with potent neutralizing Zika virus E-specific mouse monoclonal antibody ZV-67. Moreover, the novel EDII epitope was exclusively engaged in epitope-CDR interactions of potent neutralizing Zika virus E-specific human monoclonal antibody Z3L1. None of the linear epitopes of Zika virus NS1 were ascertained as novel based on our study criteria. Conformational epitopes were identified as novel for NS1 protein. Conclusion This study identified Zika virus-specific novel epitopes of envelope and non-structural -1 proteins in the currently co-circulating Indian strains. Furthermore, in-silico validation through molecular docking added insight into antigen-antibody interactions, paving way for future in vitro and in vivo studies.
Collapse
Affiliation(s)
- Rohan Raj Roy
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Nitali Tadkalkar
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Gururaj Rao Deshpande
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Nitin M. Atre
- Bioinformatics and Data Management, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Pratip Shil
- Bioinformatics and Data Management, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| | - Gajanan Sapkal
- Diagnostic Virology Group, Indian Council of Medical Research (ICMR) - National Institute of Virology, Pune, India
| |
Collapse
|
8
|
Lei Z, Gu Y, Liu Y, Liu H, Lu X, Chen W, Zhou L, Pan P, Chen Z, Yue Z, Ruan J, Zhu L, Li G, Xia X, Yu Y, Dai J, Chen X. Identification of antiviral RNAi regulators, ILF3/DHX9, recruit at ZIKV stem loop B to protect against ZIKV induced microcephaly. Nat Commun 2025; 16:1991. [PMID: 40011444 PMCID: PMC11865596 DOI: 10.1038/s41467-025-56859-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
Zika virus (ZIKV) is a member of the Flaviviridae family and causes congenital microcephaly and Guillain-Barré syndrome. Currently, there is a lack of approved vaccines or therapies against ZIKV infection. In this study, we profile vRNA‒host protein interactomes at ZIKV stem‒loop B (SLB) and reveal that interleukin enhancer binding factor 3 (ILF3) and DEAH-box helicase 9 (DHX9) form positive regulators of antiviral RNA inference in undifferentiated human neuroblastoma cells and induced pluripotent stem cell-derived human neural stem cells (iPSC-NSCs). Functionally, ablation of ILF3 in brain organoids and Nestin-Cre ILF3 cKO foetal mice significantly enhance ZIKV replication and aggravated ZIKV-induced microcephalic phenotypes. Mechanistically, ILF3/DHX9 enhance DICER processing of ZIKV vRNA-derived siRNAs (vsiR-1 and vsiR-2) to exert anti-flavivirus activity. VsiR-1 strongly inhibits ZIKV NS5 polymerase activity and RNA translation. Treatment with the vsiR-1 mimic inhibits ZIKV replication in vitro and in vivo and protected mice from ZIKV-induced microcephaly. Overall, we propose a novel therapeutic strategy to combat flavivirus infection.
Collapse
Affiliation(s)
- Zhiwei Lei
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Department of Gastroenterology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Yu Gu
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Ying Liu
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Hailiang Liu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaohua Lu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Weijie Chen
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Lu Zhou
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Pan Pan
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhuohong Chen
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Zhaoyang Yue
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Jinhui Ruan
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Leqing Zhu
- Guangzhou Laboratory, Bioland, Guangzhou, China
| | - Guangqiang Li
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Xichun Xia
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yang Yu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Jianfeng Dai
- Jiangsu Key Laboratory of Infection and Immunity, Institute of Biology and Medical Sciences, Soochow University, Suzhou, China.
| | - Xin Chen
- Guangdong Provincial Key Laboratory of Virology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China.
| |
Collapse
|
9
|
Darmuzey M, Touret F, Slowikowski E, Gladwyn-Ng I, Ahuja K, Sanchez-Felipe L, de Lamballerie X, Verfaillie C, Marques PE, Neyts J, Kaptein SJF. Epidemic Zika virus strains from the Asian lineage induce an attenuated fetal brain pathogenicity. Nat Commun 2024; 15:10870. [PMID: 39738084 PMCID: PMC11686291 DOI: 10.1038/s41467-024-55155-4] [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: 01/11/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
The 2015-2016 Zika virus (ZIKV) outbreak in the Americas revealed the ability of ZIKV from the Asian lineage to cause birth defects, generically called congenital Zika syndrome (CZS). Notwithstanding the long circulation history of Asian ZIKV, no ZIKV-associated CZS cases were reported prior to the outbreaks in French Polynesia (2013) and Brazil (2015). Whether the sudden emergence of CZS resulted from an evolutionary event of Asian ZIKV has remained unclear. We performed a comparative analysis of the pathogenicity of pre-epidemic and epidemic Asian ZIKV strains in mouse embryonic brains using a female immunocompetent intraplacental infection mouse model. All studied Asian ZIKV strains are neurovirulent, but pre-epidemic strains are consistently more pathogenic in the embryos than their epidemic equivalents. Pathogenicity is not directly linked to viral replication. By contrast, an influx of macrophages/microglial cells is noted in infected fetal brains for both pre-epidemic and epidemic ZIKV strains. Moreover, all tested ZIKV strains trigger an immunological response, whereby the intensity of the response differs between strains, and with epidemic ZIKV strains generally mounting a more attenuated immunostimulatory response. Our study reveals that Asian ZIKV strains evolved towards pathogenic attenuation, potentially resulting in CZS emergence in neonates rather than premature death in utero.
Collapse
Affiliation(s)
- Maïlis Darmuzey
- Virology and Immunology Unit, GIGA-Infection, Immunity and Inflammation, University of Liège, Liège, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium
| | - Franck Touret
- Unité Des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Emily Slowikowski
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, Leuven, Belgium
| | - Ivan Gladwyn-Ng
- Department of Application Scientists, Taconic Biosciences, Leverkusen, Germany
| | - Karan Ahuja
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium
| | - Xavier de Lamballerie
- Unité Des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Pedro E Marques
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium.
| | - Suzanne J F Kaptein
- KU Leuven Department of Microbiology, Immunology and Transplantation, Virology, Antiviral Drug & Vaccine Research Group, Rega Institute for Medical Research, Leuven, Belgium.
| |
Collapse
|
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: 3] [Impact Index Per Article: 3.0] [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
|
Zadra N, Rizzoli A, Rota-Stabelli O. Comprehensive phylogenomic analysis of Zika virus: Insights into its origin, past evolutionary dynamics, and global spread. Virus Res 2024; 350:199490. [PMID: 39489463 PMCID: PMC11583807 DOI: 10.1016/j.virusres.2024.199490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Zika virus (ZIKV), a Flaviviridae family member, has been linked to severe neurological disorders. Despite detailed studies on recent outbreaks, the early evolutionary history of ZIKV remains partially unclear. This study elucidates ZIKV origin and evolutionary dynamics, focusing on recombination events, early lineage diversification, and virus spread across continents. METHODS We assessed recombination using multiple methods. We conducted Bayesian phylogenetic analyses to understand the evolutionary relationships and timing of key diversification events. Model selection was carried out to determine the most appropriate evolutionary model for our dataset. RESULTS Our phylogenies revealed recent recombination between Singaporean and African lineages, indicating the co-circulation of diverse lineages during outbreaks. Thailand was identified as a crucial hub in the spread across Asia. The phylogenetic analysis suggests that the ZIKV lineage dates back to the eleventh century, with the first significant diversification occurring in the nineteenth century. The timing of the re-introduction of the Asian lineage into Africa and the delay between probable introduction and outbreak onset were also determined. CONCLUSIONS This study provides novel insights into ZIKV's origin and early evolutionary dynamics, highlighting Thailand's role in the spread of the virus in Asia and recent recombination events between distant lineages. These findings emphasize the need for continuous surveillance and a better understanding of ZIKV biology to forecast and mitigate future outbreaks.
Collapse
Affiliation(s)
- Nicola Zadra
- Conservation Genomics Research Unit, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Trento, Italy; NBFC, National Biodiversity Future Center, Palermo 90133, Italy.
| | - Annapaola Rizzoli
- Applied Ecology Research Unit, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Trento, Italy; NBFC, National Biodiversity Future Center, Palermo 90133, Italy
| | - Omar Rota-Stabelli
- Center Agriculture Food Environment (C3A), University of Trento, 38010, San Michele all'Adige, TN, Italy
| |
Collapse
|
13
|
Ning X, Xia B, Wang J, Gao R, Ren H. Host-adaptive mutations in Chikungunya virus genome. Virulence 2024; 15:2401985. [PMID: 39263937 PMCID: PMC11404619 DOI: 10.1080/21505594.2024.2401985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/08/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Chikungunya virus (CHIKV) is the causative agent of chikungunya fever (CHIKF), and its primary vectors are the mosquitoes Aedes aegypti and Aedes albopictus. CHIKV was initially endemic to Africa but has spread globally in recent years and affected millions of people. According to a risk assessment by the World Health Organization, CHIKV has the potential seriously impact public health. A growing body of research suggests that mutations in the CHIKV gene that enhance viral fitness in the host are contributing to the expansion of the global CHIKF epidemic. In this article, we review the host-adapted gene mutations in CHIKV under natural evolution and laboratory transmission conditions, which can help improve our understanding of the adaptive evolution of CHIKV and provide a basis for monitoring and early warning of future CHIKV outbreaks.
Collapse
Affiliation(s)
- Xinhang Ning
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| | - Jiaqi Wang
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| | - Rong Gao
- Department of Respiratory Medicine, The People’s Liberation Army Joint Logistic Support Force 943 Hospital, Wuwei, Gansu, People’s Republic of China
| | - Hao Ren
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
14
|
Bindu, Pandey HS, Seth P. Interplay Between Zika Virus-Induced Autophagy and Neural Stem Cell Fate Determination. Mol Neurobiol 2024; 61:9927-9944. [PMID: 37910284 DOI: 10.1007/s12035-023-03704-1] [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: 04/14/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023]
Abstract
The Zika virus (ZIKV) outbreaks and its co-relation with microcephaly have become a global health concern. It is primarily transmitted by a mosquito, but can also be transmitted from an infected mother to her fetus causing impairment in brain development, leading to microcephaly. However, the underlying molecular mechanism of ZIKV-induced microcephaly is poorly understood. In this study, we explored the role of ZIKV non-structural protein NS4A and NS4B in ZIKV pathogenesis in a well-characterized primary culture of human fetal neural stem cells (fNSCs). We observed that the co-transfection of NS4A and NS4B altered the neural stem cell fate by arresting proliferation and inducing premature neurogenesis. NS4A + NS4B transfection in fNSCs increased autophagy and dysregulated notch signaling. Further, it also altered the regulation of downstream genes controlling cell proliferation. Additionally, we reported that 3 methyl-adenine (3-MA), a potent autophagy inhibitor, attenuated the deleterious effects of NS4A and NS4B as evidenced by the rescue in Notch1 expression, enhanced proliferation, and reduced premature neurogenesis. Our attempts to understand the mechanism of autophagy induction indicate the involvement of mitochondrial fission and ROS. Collectively, our findings highlight the novel role of NS4A and NS4B in mediating NSC fate alteration through autophagy-mediated notch degradation. The study also helps to advance our understanding of ZIKV-induced neuropathogenesis and suggests autophagy as a potential target for anti-ZIKV therapeutic intervention.
Collapse
Affiliation(s)
- Bindu
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, 122052, India
| | - Hriday Shanker Pandey
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, 122052, India
| | - Pankaj Seth
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, 122052, India.
| |
Collapse
|
15
|
Ren Y, Liu Y, Pang R, Xu G, Lei Y, Kwok HF, Wu Y, Cao Z. ZIKV prM hijacks PIM1 kinase for phosphorylation to prevent ubiquitin-mediated degradation and facilitate viral replication. Front Cell Infect Microbiol 2024; 14:1502770. [PMID: 39679197 PMCID: PMC11638163 DOI: 10.3389/fcimb.2024.1502770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Viral infection usually stimulates a variety of host cell factors to modulate the life cycle of the virus. PIM1, a serine/threonine protein kinase widely involved in cell proliferation, survival, differentiation and apoptosis, was recently reported to be upregulated by Zika virus (ZIKV) infection. However, how ZIKV-PIM1 interactions affect the viral life cycle are not fully understood. Methods and results Here, we demonstrated that ZIKV replication was suppressed by the PIM1 kinase inhibitor SGI-1776 in both wt and Ifnar1-/- murine peritoneal macrophages, indicating that PIM1 functions independently of type I IFN signaling. Co-immunoprecipitation and GST pull-down assays revealed that the ZIKV structural protein precursor membrane (prM) interacted with PIM1. Moreover, we found that prM protein stability was enhanced by PIM1, which was attributed to its kinase activity. Mechanistically, we revealed that prM can undergo ubiquitin‒mediated proteolysis and the E3 ubiquitin ligase AMFR can target prM for degradation. Importantly, PIM1 catalyzed phosphorylation of prM at Ser101 and Thr107, and this phosphorylation prevented the proteasome-dependent degradation of prM by impairing its association with AMFR. Therefore, the S101/T107-D phosphorylation mimic mutant of prM was more resistant to PIM1-induced increases in cellular abundance. Discussion These findings revealed PIM1 as a critical host factor that is advantageous to ZIKV and revealed that targeting the PIM1‒prM axis is a conducive strategy for controlling ZIKV infection.
Collapse
Affiliation(s)
- Yingying Ren
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
- Shenzhen Research Institute, Wuhan University, Shenzhen, Guangdong, China
| | - Yishuo Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Rui Pang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Gang Xu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei, China
| | - Yining Lei
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei, China
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, Guangdong, China
| | - Hang Fai Kwok
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
- Shenzhen Research Institute, Wuhan University, Shenzhen, Guangdong, China
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei, China
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Fan XX, Li RT, Zhu YB, Chen Q, Li XF, Cao TS, Zhao H, Cheng G, Qin CF. An accumulated mutation gained in mosquito cells enhances Zika virus virulence and fitness in mice. J Virol 2024; 98:e0125124. [PMID: 39412258 PMCID: PMC11575407 DOI: 10.1128/jvi.01251-24] [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: 07/18/2024] [Accepted: 09/18/2024] [Indexed: 11/20/2024] Open
Abstract
Zika virus (ZIKV) remains a significant public health threat worldwide. A number of adaptive mutations have accumulated within the genome of ZIKV during global transmission, some of which have been linked to specific phenotypes. ZIKV maintains an alternating cycle of replication between mosquitoes and vertebrate hosts, but the role of mosquito-specific adaptive mutations in ZIKV has not been well investigated. In this study, we demonstrated that serial passaging of ZIKV in mosquito Aag2 cells led to the emergence of critical amino acid substitutions, including A94V in the prM protein and V153D and H401Y in the E protein. Further characterization via reverse genetics revealed that the H401Y substitution in the E protein did not augment viral replication in mosquitoes but significantly enhanced neurovirulence and lethality compared with those of the wild-type (WT) virus in mice. More importantly, the H401Y mutant maintained its virulence phenotype in mice after propagation in mosquitoes in mosquito-mouse cycle model. In particular, recombinant ZIKV harboring the H401Y substitution showed enhanced competitive fitness over WT ZIKV in various mammalian cells and mouse brains, but not in mosquito cells. Notably, the H401Y substitution in the ZIKV E protein has been detected in recent isolates derived from both mosquitoes and humans in Asia and the Americas. In summary, our findings not only identify a novel virulence determinant of ZIKV but also highlight the complexity of the relationship between the evolution of vector-borne viruses and their clinical outcome in nature. IMPORTANCE Zika virus (ZIKV) is an important arbovirus with a global impact. Experimental evolution by serial passaging of ZIKV in susceptible cells has led to the identification of a panel of critical amino acid substitutions with specific functions. Herein, we identified a mosquito cell-derived substitution, H401Y, in the ZIKV E protein via experimental evolution. The H401Y substitution significantly enhanced viral virulence and fitness in mammal cells and mice. Notably, the H401Y substitution has been detected in recent mosquito and human isolates from regions spanning Asia to the Americas. Our work elucidates unrecognized virulence determinant in the ZIKV genome that warrants urgent attention. Moreover, the findings underscore the critical need for extensive molecular surveillance and rigorous clinical observation to establish the potential impact in natural circulation. These endeavors are crucial for unraveling the potential of mutation to act as a catalyst for future epidemics, thereby preempting the public health challenges it may pose.
Collapse
Affiliation(s)
- Xiao-Xuan Fan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Rui-Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yi-Bin Zhu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Qi Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Tian-Shu Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Hui Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Rivera-Franco N, López-Alvarez D, Castillo A, Aristizabal E, Puiu D, Salzberg SL, Pardo CA, Parra B. Genomic variability in Zika virus in GBS cases in Colombia. PLoS One 2024; 19:e0313545. [PMID: 39561198 PMCID: PMC11575819 DOI: 10.1371/journal.pone.0313545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024] Open
Abstract
Major clusters of Guillain-Barré Syndrome (GBS) emerged during the Zika virus (ZIKV) outbreaks in the South Pacific and the Americas from 2014 to 2016. The factors contributing to GBS susceptibility in ZIKV infection remain unclear, although considerations of viral variation, patient susceptibility, environmental influences, and other potential factors have been hypothesized. Studying the role of viral genetic factors has been challenging due to the low viral load and rapid viral clearance from the blood after the onset of Zika symptoms. The prolonged excretion of ZIKV in urine by the time of GBS onset, when the virus is no longer present in the blood, provides an opportunity to unravel whether specific ZIKV mutations are related to the development of GBS in certain individuals. This study aimed to investigate the association between specific ZIKV genotypes and the development of GBS, taking advantage of a unique collection of ZIKV-positive urine samples obtained from GBS cases and controls during the 2016 ZIKV outbreak in Colombia. Utilizing Oxford-Nanopore technology, we conducted complete genome sequencing of ZIKV in biological samples from 15 patients with GBS associated with ZIKV and 17 with ZIKV infection without neurological complications. ZIKV genotypes in Colombia exhibited distribution across three clades (average bootstrap of 90.9±14.9%), with two clades dominating the landscape. A comparative analysis of ZIKV genomes from GBS and non-neurological complications, alongside 1368 previously reported genomes, revealed no significant distinctions between the two groups. Both genotypes were similarly distributed among observed clades in Colombia. Furthermore, no variations were identified in the amino acid composition of the viral genome between the two groups. Our findings suggest that GBS in ZIKV infection is perhaps associated with patient susceptibility and/or other para- or post-infectious immune-mediated mechanisms rather than with specific ZIKV genome variations.
Collapse
Affiliation(s)
- Nelson Rivera-Franco
- Laboratorio de Técnicas y Análisis Ómicos-TAOLab/CiBioFi, Facultad de Ciencias Naturales y Exactas, Universidad del Valle, Cali, Valle del Cauca, Colombia
- Grupo VIREM-Virus Emergentes y Enfermedad, Escuela de Ciencias Básicas, Facultad de Salud, Universidad del Valle, Cali, Valle del Cauca, Colombia
- Department of Neurology & Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Diana López-Alvarez
- Laboratorio de Técnicas y Análisis Ómicos-TAOLab/CiBioFi, Facultad de Ciencias Naturales y Exactas, Universidad del Valle, Cali, Valle del Cauca, Colombia
- Grupo VIREM-Virus Emergentes y Enfermedad, Escuela de Ciencias Básicas, Facultad de Salud, Universidad del Valle, Cali, Valle del Cauca, Colombia
- Departamento de Ciencias Biológicas, Facultad de Ciencias Agropecuarias, Universidad Nacional de Colombia, Palmira, Valle del Cauca, Colombia
| | - Andrés Castillo
- Laboratorio de Técnicas y Análisis Ómicos-TAOLab/CiBioFi, Facultad de Ciencias Naturales y Exactas, Universidad del Valle, Cali, Valle del Cauca, Colombia
| | - Erica Aristizabal
- Grupo VIREM-Virus Emergentes y Enfermedad, Escuela de Ciencias Básicas, Facultad de Salud, Universidad del Valle, Cali, Valle del Cauca, Colombia
| | - Daniela Puiu
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Steven L Salzberg
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Carlos A Pardo
- Department of Neurology & Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Beatriz Parra
- Grupo VIREM-Virus Emergentes y Enfermedad, Escuela de Ciencias Básicas, Facultad de Salud, Universidad del Valle, Cali, Valle del Cauca, Colombia
| |
Collapse
|
18
|
Cabral AP, Maia FPDS, Magliano DC, Graceli JB, Soares P, Morris EAR, Miranda-Alves L. Pyriproxyfen, villain or good guy? A brief review. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240154. [PMID: 39876972 PMCID: PMC11771759 DOI: 10.20945/2359-4292-2024-0154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/02/2024] [Indexed: 01/31/2025]
Abstract
Pyriproxyfen (PPF) acts as a juvenile growth regulator, interfering with normal metamorphosis and blocking the development of insects into adulthood. Although the World Health Organization (WHO) considers the use of PPF at a concentration of 0.01 mg/L as unlikely to pose health risks, recent studies have unveiled potential risks associated with PPF exposure to non-target organisms. Exposure to PPF disrupts insect development primarily by mimicking juvenile hormones; therefore, concerns linger over its impact on unintended species. Studies have highlighted the adverse effects of PPF on aquatic invertebrates, fish, and amphibians and revealed mortality and developmental abnormalities in non-target mosquito species exposed to PPF-treated water. Moreover, PPF may act as an endocrine disruptor, interfering with hormonal pathways crucial for growth, reproduction, and behavior in exposed organisms. Amphibians, for instance, display altered reproductive physiology and developmental abnormalities due to disruptions in endocrine signaling pathways caused by PPF. The ecological ramifications of PPF extend beyond direct toxicity to non-target species. Indirect effects include shifts in food web dynamics and ecosystem functioning. Reductions in insect populations, induced by PPF, can disrupt food availability for higher trophic levels, potentially destabilizing community structure and ecosystem equilibrium. Given mounting evidence of unintended consequences, robust risk assessment and regulatory oversight are imperative. Accurate classification of PPF by regulatory bodies is essential to balancing its role in disease control and pest management benefits with the need to safeguard non-target species and maintain ecosystem health. Future research must prioritize comprehensive assessments of PPF's ecological impact across various habitats and taxa to inform evidence-based policymaking.
Collapse
Affiliation(s)
- Andressa Pereira Cabral
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasLaboratório de Endocrinologia ExperimentalRio de JaneiroRJBrasilLaboratório de Endocrinologia Experimental (LEEx), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasRio de JaneiroRJBrasilPrograma de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Fabrício Pereira dos Santos Maia
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasLaboratório de Endocrinologia ExperimentalRio de JaneiroRJBrasilLaboratório de Endocrinologia Experimental (LEEx), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - D’Angelo Carlo Magliano
- Universidade Federal FluminenseCentro de Morfologia e MetabolismoNiteróiRJBrasilCentro de Morfologia e Metabolismo, Universidade Federal Fluminense, Niterói, RJ, Brasil
| | - Jones Bernardes Graceli
- Universidade Federal do Espírito SantoLaboratório de Endocrinologia e Toxicologia CelularDepartamento de MorfologiaEspírito SantoESBrasilLaboratório de Endocrinologia e Toxicologia Celular, Departamento de Morfologia, Universidade Federal do Espírito Santo, Espírito Santo, ES, Brasil
| | - Paula Soares
- Universidade do PortoInstituto de Investigação e Inovação em SaúdeGrupo de Sinalização e Metabolismo CelularPortoPortugalGrupo de Sinalização e Metabolismo Celular, i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Eduardo Andrés Rios Morris
- Universidade Federal do Rio de JaneiroPrograma de Pós-graduação em EndocrinologiaFaculdade de MedicinaRio de JaneiroRJBrasilPrograma de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Leandro Miranda-Alves
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasLaboratório de Endocrinologia ExperimentalRio de JaneiroRJBrasilLaboratório de Endocrinologia Experimental (LEEx), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasRio de JaneiroRJBrasilPrograma de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
- Universidade do PortoInstituto de Investigação e Inovação em SaúdeGrupo de Sinalização e Metabolismo CelularPortoPortugalGrupo de Sinalização e Metabolismo Celular, i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Universidade Federal do Rio de JaneiroPrograma de Pós-graduação em EndocrinologiaFaculdade de MedicinaRio de JaneiroRJBrasilPrograma de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
19
|
Heydarifard Z, Shapshak P, Zandi M. Underlying Factors Predisposing to Viral-Induced Neurological Diseases. Rev Med Virol 2024; 34:e2587. [PMID: 39327640 DOI: 10.1002/rmv.2587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Zahra Heydarifard
- Department of Virology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Paul Shapshak
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Milad Zandi
- Department of Microbiology, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
20
|
Li Q, Yang RH, Hu Y, Tang BB, Jiang YJ, Zheng CB, Song TZ. Zika virus infection induces glycometabolic disorder in northern pig-tailed macaques. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2527-2529. [PMID: 39235558 DOI: 10.1007/s11427-024-2663-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/24/2024] [Indexed: 09/06/2024]
Affiliation(s)
- Qing Li
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ren-Hua Yang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Yan Hu
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bei-Bei Tang
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- KIZ-SU Joint Laboratory of Animal Model and Drug Development, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215021, China
| | - Ying-Jie Jiang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China.
| | - Tian-Zhang Song
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
21
|
Lefèvre C, Cook GM, Dinan AM, Torii S, Stewart H, Gibbons G, Nicholson AS, Echavarría-Consuegra L, Meredith LW, Lulla V, McGovern N, Kenyon JC, Goodfellow I, Deane JE, Graham SC, Lakatos A, Lambrechts L, Brierley I, Irigoyen N. Zika viruses encode 5' upstream open reading frames affecting infection of human brain cells. Nat Commun 2024; 15:8822. [PMID: 39394194 PMCID: PMC11470053 DOI: 10.1038/s41467-024-53085-9] [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: 06/07/2023] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
Zika virus (ZIKV), an emerging mosquito-borne flavivirus, is associated with congenital neurological complications. Here, we investigate potential pathological correlates of virus gene expression in representative ZIKV strains through RNA sequencing and ribosome profiling. In addition to the single long polyprotein found in all flaviviruses, we identify the translation of unrecognised upstream open reading frames (uORFs) in the genomic 5' region. In Asian/American strains, ribosomes translate uORF1 and uORF2, whereas in African strains, the two uORFs are fused into one (African uORF). We use reverse genetics to examine the impact on ZIKV fitness of different uORFs mutant viruses. We find that expression of the African uORF and the Asian/American uORF1 modulates virus growth and tropism in human cortical neurons and cerebral organoids, suggesting a potential role in neurotropism. Although the uORFs are expressed in mosquito cells, we do not see a measurable effect on transmission by the mosquito vector in vivo. The discovery of ZIKV uORFs sheds new light on the infection of the human brain cells by this virus and raises the question of their existence in other neurotropic flaviviruses.
Collapse
Affiliation(s)
- Charlotte Lefèvre
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Georgia M Cook
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Adam M Dinan
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Shiho Torii
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Hazel Stewart
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - George Gibbons
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Alex S Nicholson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | | | - Luke W Meredith
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Valeria Lulla
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Naomi McGovern
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Julia C Kenyon
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Janet E Deane
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Stephen C Graham
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - András Lakatos
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, Cambridge, UK
| | - Louis Lambrechts
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Ian Brierley
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
22
|
Mantena S, Pillai PP, Petros BA, Welch NL, Myhrvold C, Sabeti PC, Metsky HC. Model-directed generation of artificial CRISPR-Cas13a guide RNA sequences improves nucleic acid detection. Nat Biotechnol 2024:10.1038/s41587-024-02422-w. [PMID: 39394482 DOI: 10.1038/s41587-024-02422-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/04/2024] [Indexed: 10/13/2024]
Abstract
CRISPR guide RNA sequences deriving exactly from natural sequences may not perform optimally in every application. Here we implement and evaluate algorithms for designing maximally fit, artificial CRISPR-Cas13a guides with multiple mismatches to natural sequences that are tailored for diagnostic applications. These guides offer more sensitive detection of diverse pathogens and discrimination of pathogen variants compared with guides derived directly from natural sequences and illuminate design principles that broaden Cas13a targeting.
Collapse
Affiliation(s)
- Sreekar Mantena
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | | | - Brittany A Petros
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Cambridge, MA, USA
- MD-PhD Program, Harvard/Massachusetts Institute of Technology, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Pardis C Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | | |
Collapse
|
23
|
Egloff C, Fovet CM, Denis J, Pascal Q, Bossevot L, Luccantoni S, Leonec M, Dereuddre-Bosquet N, Leparc-Goffart I, Le Grand R, Durand GA, Badaut C, Picone O, Roques P. Fetal Zika virus inoculation in macaques revealed control of the fetal viral load during pregnancy. Virol J 2024; 21:209. [PMID: 39227837 PMCID: PMC11373269 DOI: 10.1186/s12985-024-02468-x] [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: 12/18/2023] [Accepted: 08/13/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Early pregnancy Zika virus (ZIKV) infection is associated with major brain damage in fetuses, leading to microcephaly in 0.6-5.0% of cases, but the underlying mechanisms remain largely unknown. METHODS To understand the kinetics of ZIKV infection during fetal development in a nonhuman primate model, four cynomolgus macaque fetuses were exposed in utero through echo-guided intramuscular inoculation with 103 PFU of ZIKV at 70-80 days of gestation, 2 controls were mock inoculated. Clinical, immuno-virological and ultrasound imaging follow-ups of the mother/fetus pairs were performed until autopsy after cesarean section 1 or 2 months after exposure (n = 3 per group). RESULTS ZIKV was transmitted from the fetus to the mother and then replicate in the peripheral blood of the mother from week 1 to 4 postexposure. Infected fetal brains tended to be smaller than those of controls, but not the femur lengths. High level of viral RNA ws found after the first month in brain tissues and placenta. Thereafter, there was partial control of the virus in the fetus, resulting in a decreased number of infected tissue sections and a decreased viral load. Immune cellular and humoral responses were effectively induced. CONCLUSIONS ZIKV infection during the second trimester of gestation induces short-term brain injury, and although viral genomes persist in tissues, most of the virus is cleared before delivery.
Collapse
Affiliation(s)
- Charles Egloff
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
- Service de gynécologie-obstétrique, Hôpital Louis Mourier, AP-HP, IAME INSERM U1137, Université de PARIS, Paris, France
| | - Claire-Maëlle Fovet
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
| | - Jessica Denis
- Unité interactions hôtes-pathogènes, Institut de Recherche Biomédicale des Armées, 91223, Brétigny-sur-Orge, France
| | - Quentin Pascal
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
| | - Laetitia Bossevot
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
| | - Sophie Luccantoni
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
| | - Marco Leonec
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
| | - Isabelle Leparc-Goffart
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-Corsica Univ-IRD 190-Inserm 1207-IRBA), 13005, Marseille, France
- National Reference Center for Arboviruses, INSERM-Institut de Recherche Biomédicale des Armées, 13005, Marseille, France
| | - Roger Le Grand
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France
| | - Guillaume André Durand
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-Corsica Univ-IRD 190-Inserm 1207-IRBA), 13005, Marseille, France
- National Reference Center for Arboviruses, INSERM-Institut de Recherche Biomédicale des Armées, 13005, Marseille, France
| | - Cyril Badaut
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-Corsica Univ-IRD 190-Inserm 1207-IRBA), 13005, Marseille, France
- Unité de Virologie, Institut de Recherche Biomédicale des Armées, 91223, Brétigny-sur-Orge, France
| | - Olivier Picone
- Service de gynécologie-obstétrique, Hôpital Louis Mourier, AP-HP, IAME INSERM U1137, Université de PARIS, Paris, France
| | - Pierre Roques
- Center for Immunology of Viral, Auto-Immune, Hematological and Viral Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, 92265, Fontenay aux Roses, France.
- Virology Unit, Institut Pasteur de Guinée (IPGui), BP4416, Conakry, Guinea.
| |
Collapse
|
24
|
Kim WJ, Lee AR, Hong SY, Kim SH, Kim JD, Kim SJ, Oh JS, Shim SM, Seo SU. Characterization of a Small Plaque Variant Derived from Genotype V Japanese Encephalitis Virus Clinical Isolate K15P38. J Microbiol Biotechnol 2024; 34:1592-1598. [PMID: 39081248 PMCID: PMC11380520 DOI: 10.4014/jmb.2404.04054] [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: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 08/29/2024]
Abstract
Genotype V (GV) Japanese encephalitis virus (JEV) has been predominantly reported in the Republic of Korea (ROK) since 2010. GV JEV exhibits higher virulence and distinct antigenicity compared to other genotypes, which results in reduced efficacy of existing vaccines. Research on GV JEV is essential to minimize its clinical impact, but the only available clinical strain in the ROK is K15P38, isolated from the cerebrospinal fluid of a patient in 2015. We obtained this virus from National Culture Collection for Pathogens (NCCP) and isolated a variant forming small plaques during our research. We identified that this variant has one amino acid substitution each in the PrM and NS5 proteins compared to the reported K15P38. Additionally, we confirmed that this virus exhibits delayed propagation in vitro and an attenuated phenotype in mice. The isolation of this variant is a critical reference for researchers intending to study K15P38 obtained from NCCP, and the mutations in the small plaque-forming virus are expected to be useful for studying the pathology of GV JEV.
Collapse
Affiliation(s)
- Woo-Jin Kim
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Ah-Ra Lee
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Su-Yeon Hong
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang-Hyun Kim
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jae-Deog Kim
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sung Jae Kim
- Vaxdigm Co., Ltd., Seoul 04798, Republic of Korea
- Bio & Living Engineering Major, Global Leaders College, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae Sang Oh
- Department of Neurosurgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang-Mu Shim
- Division of Acute Viral Disease, Center for Emerging Virus Research, National Institute of Infectious Diseases, Korea National Institute of Health, Cheongju 28159, Republic of Korea
| | - Sang-Uk Seo
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
25
|
Yan Y, Yang H, Yang Y, Wang J, Zhou Y, Tang C, Li B, Huang Q, An R, Liang X, Lin D, Yu W, Fan C, Lu S. The inoculum dose of Zika virus can affect the viral replication dynamics, cytokine responses and survival rate in immunocompromised AG129 mice. MOLECULAR BIOMEDICINE 2024; 5:30. [PMID: 39095588 PMCID: PMC11297010 DOI: 10.1186/s43556-024-00195-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
Zika virus, a mosquito-borne arbovirus, has repeatedly caused large pandemics with symptoms worsening from mild and self-limiting diseases to Guillain-Barré syndrome in adults and fetal microcephaly in newborns. In recent years, Zika virus diseases have posed a serious threat to human health. The shortage of susceptible small animal models makes it difficult to study pathogenic mechanisms and evaluate potential therapies for Zika virus infection. Therefore, we chose immunocompromised mice (AG129 mice) deficient in IFN-α/β and IFN-γ receptors, which can abolish the innate immune system that prevents Zika virus infection early. AG129 mice were infected with the Zika virus, and this mouse model exhibited replication dynamics, tissue tropism, pathological lesion and immune activation of the Zika virus. Our results suggest that the inoculum dose of Zika virus can affect the viral replication dynamics, cytokine responses and survival rate in AG129 mice. By testing the potential antiviral drug favipiravir, several critical indicators, including replication dynamics and survival rates, were identified in AG129 mice after Zika virus infection. It is suggested that the model is reliable for drug evaluation. In brief, this model provides a potential platform for studies of the infectivity, virulence, and pathogenesis of the Zika virus. Moreover, the development of an accessible mouse model of Zika virus infection will expedite the research and deployment of therapeutics and vaccines.
Collapse
Affiliation(s)
- Yuhuan Yan
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Hao Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Junbin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yanan Zhou
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Cong Tang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Bai Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Qing Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Ran An
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Xiaoming Liang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Dongdong Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.
| | - Changfa Fan
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China.
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Kunming, China.
| |
Collapse
|
26
|
Saivish MV, Nogueira ML, Rossi SL, Vasilakis N. Exploring Iguape Virus-A Lesser-Known Orthoflavivirus. Viruses 2024; 16:960. [PMID: 38932252 PMCID: PMC11209261 DOI: 10.3390/v16060960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Brazil has earned the moniker "arbovirus hotspot", providing an ideal breeding ground for a multitude of arboviruses thriving in various zoonotic and urban cycles. As the planet warms and vectors expand their habitat range, a nuanced understanding of lesser-known arboviruses and the factors that could drive their emergence becomes imperative. Among these viruses is the Iguape virus (IGUV), a member of the Orthoflavivirus aroaense species, which was first isolated in 1979 from a sentinel mouse in the municipality of Iguape, within the Vale do Ribeira region of São Paulo State. While evidence suggests that IGUV circulates among birds, wild rodents, marsupials, bats, and domestic birds, there is no information available on its pathogenesis in both humans and animals. The existing literature on IGUV spans decades, is outdated, and is often challenging to access. In this review, we have curated information from the known literature, clarifying its elusive nature and investigating the factors that may influence its emergence. As an orthoflavivirus, IGUV poses a potential threat, which demands our attention and vigilance, considering the serious outbreaks that the Zika virus, another neglected orthoflavivirus, has unleashed in the recent past.
Collapse
Affiliation(s)
- Marielena V. Saivish
- Laboratórios de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto 15090-000, SP, Brazil; (M.V.S.); (M.L.N.)
- Brazilian Biosciences National Laboratory, Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas 13083-100, SP, Brazil
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
| | - Maurício L. Nogueira
- Laboratórios de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto 15090-000, SP, Brazil; (M.V.S.); (M.L.N.)
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
| | - Shannan L. Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX 77555-0609, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX 77555-0610, USA
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX 77555-0609, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX 77555-0610, USA
| |
Collapse
|
27
|
Kai I, Kobayashi D, Itokawa K, Sanjoba C, Itoyama K, Isawa H. Evaluation of long-term preservation methods for viral RNA in mosquitoes at room temperature. J Virol Methods 2024; 325:114887. [PMID: 38237867 DOI: 10.1016/j.jviromet.2024.114887] [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: 11/17/2023] [Revised: 01/05/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Mosquitoes are important vectors of various pathogenic viruses. Almost all viruses transmitted by mosquitoes are RNA viruses. Therefore, to detect viral genes, mosquito samples must be kept at low temperatures to prevent RNA degradation. However, prolonged transport from the field to laboratory can pose challenges for temperature control. The aim of this study was to evaluate methods for preserving viral RNA in mosquito bodies at room temperature. Virus-infected mosquito samples were immersed in ethanol, propylene glycol, and a commercially available nucleic acid preservation reagent at room temperature, and viral RNA stability was compared. As a result, for the two RNA viruses (San Gabriel mononegavirus and dengue virus 1) subjected to this experiment, no significant decrease in the viral RNA was observed for at least eight weeks after immersion in the reagents, and the amount of RNA remaining was equivalent to that of samples stored at - 80 °C. These results indicate that immersion storage in these reagents used in this study is effective in preserving viral RNA in mosquitoes under room temperature conditions and is expected to be implemented in epidemiologic surveillance that is not limited by the cold chain from the field to the laboratory.
Collapse
Affiliation(s)
- Izumi Kai
- Graduate school of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, Japan; Department of Medical Entomology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, Japan
| | - Daisuke Kobayashi
- Department of Medical Entomology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, Japan; Management Department of Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, Japan.
| | - Kentaro Itokawa
- Department of Medical Entomology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, Japan
| | - Chizu Sanjoba
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Kyo Itoyama
- Graduate school of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, Japan
| | - Haruhiko Isawa
- Department of Medical Entomology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
28
|
Peng J, Zhang M, Wang G, Zhang D, Zheng X, Li Y. Biased virus transmission following sequential coinfection of Aedes aegypti with dengue and Zika viruses. PLoS Negl Trop Dis 2024; 18:e0012053. [PMID: 38557981 PMCID: PMC10984552 DOI: 10.1371/journal.pntd.0012053] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Mosquito-borne arboviruses are expanding their territory and elevating their infection prevalence due to the rapid climate change, urbanization, and increased international travel and global trade. Various significant arboviruses, including the dengue virus, Zika virus, Chikungunya virus, and yellow fever virus, are all reliant on the same primary vector, Aedes aegypti. Consequently, the occurrence of arbovirus coinfection in mosquitoes is anticipated. Arbovirus coinfection in mosquitoes has two patterns: simultaneous and sequential. Numerous studies have demonstrated that simultaneous coinfection of arboviruses in mosquitoes is unlikely to exert mutual developmental influence on these viruses. However, the viruses' interplay within a mosquito after the sequential coinfection seems intricated and not well understood. METHODOLOGY/PRINCIPAL FINDINGS We conducted experiments aimed at examining the phenomenon of arbovirus sequential coinfection in both mosquito cell line (C6/36) and A. aegypti, specifically focusing on dengue virus (DENV, serotype 2) and Zika virus (ZIKV). We firstly observed that DENV and ZIKV can sequentially infect mosquito C6/36 cell line, but the replication level of the subsequently infected ZIKV was significantly suppressed. Similarly, A. aegypti mosquitoes can be sequentially coinfected by these two arboviruses, regardless of the order of virus exposure. However, the replication, dissemination, and the transmission potential of the secondary virus were significantly inhibited. We preliminarily explored the underlying mechanisms, revealing that arbovirus-infected mosquitoes exhibited activated innate immunity, disrupted lipid metabolism, and enhanced RNAi pathway, leading to reduced susceptibility to the secondary arbovirus infections. CONCLUSIONS/SIGNIFICANCE Our findings suggest that, in contrast to simultaneous arbovirus coinfection in mosquitoes that can promote the transmission and co-circulation of these viruses, sequential coinfection appears to have limited influence on arbovirus transmission dynamics. However, it is important to note that more experimental investigations are needed to refine and expand upon this conclusion.
Collapse
Affiliation(s)
- Jiameng Peng
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Meichun Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Gang Wang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Dongjing Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Xiaoying Zheng
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Yongjun Li
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Srikrishna D. Pentagon Found Daily, Metagenomic Detection of Novel Bioaerosol Threats to Be Cost-Prohibitive: Can Virtualization and AI Make It Cost-Effective? Health Secur 2024; 22:108-129. [PMID: 38625036 DOI: 10.1089/hs.2023.0048] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
In 2022, the Pentagon Force Protection Agency found threat agnostic detection of novel bioaerosol threats to be "not feasible for daily operations" due to the cost of reagents used for metagenomics, cost of sequencing instruments, and cost of labor for subject matter experts to analyze bioinformatics. Similar operational difficulties might extend to many of the 280,000 buildings (totaling 2.3 billion square feet) at 5,000 secure US Department of Defense military sites, 250 Navy ships, as well as many civilian buildings. These economic barriers can still be addressed in a threat agnostic manner by dynamically pooling samples from dry filter units, called spike-triggered virtualization, whereby pooling and sequencing depth are automatically modulated based on novel biothreats in the sequencing output. By running at a high average pooling factor, the daily and annual cost per dry filter unit can be reduced by 10 to 100 times depending on the chosen trigger thresholds. Artificial intelligence can further enhance the sensitivity of spike-triggered virtualization. The risk of infection during the 12- to 24-hour window between a bioaerosol incident and its detection remains, but in some cases it can be reduced by 80% or more with high-speed indoor air cleaning exceeding 12 air changes per hour, which is similar to the rate of air cleaning in passenger airplanes in flight. That level of air changes per hour or higher is likely to be cost-prohibitive using central heating ventilation and air conditioning systems, but it can be achieved economically by using portable air filtration in rooms with typical ceiling heights (less than 10 feet) for a cost of approximately $0.50 to $1 per square foot for do-it-yourself units and $2 to $5 per square foot for high-efficiency particulate air filters.
Collapse
|
30
|
Elliott KC, Mattapallil JJ. Zika Virus-A Reemerging Neurotropic Arbovirus Associated with Adverse Pregnancy Outcomes and Neuropathogenesis. Pathogens 2024; 13:177. [PMID: 38392915 PMCID: PMC10892292 DOI: 10.3390/pathogens13020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/07/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Zika virus (ZIKV) is a reemerging flavivirus that is primarily spread through bites from infected mosquitos. It was first discovered in 1947 in sentinel monkeys in Uganda and has since been the cause of several outbreaks, primarily in tropical and subtropical areas. Unlike earlier outbreaks, the 2015-2016 epidemic in Brazil was characterized by the emergence of neurovirulent strains of ZIKV strains that could be sexually and perinatally transmitted, leading to the Congenital Zika Syndrome (CZS) in newborns, and Guillain-Barre Syndrome (GBS) along with encephalitis and meningitis in adults. The immune response elicited by ZIKV infection is highly effective and characterized by the induction of both ZIKV-specific neutralizing antibodies and robust effector CD8+ T cell responses. However, the structural similarities between ZIKV and Dengue virus (DENV) lead to the induction of cross-reactive immune responses that could potentially enhance subsequent DENV infection, which imposes a constraint on the development of a highly efficacious ZIKV vaccine. The isolation and characterization of antibodies capable of cross-neutralizing both ZIKV and DENV along with cross-reactive CD8+ T cell responses suggest that vaccine immunogens can be designed to overcome these constraints. Here we review the structural characteristics of ZIKV along with the evidence of neuropathogenesis associated with ZIKV infection and the complex nature of the immune response that is elicited by ZIKV infection.
Collapse
Affiliation(s)
- Kenneth C. Elliott
- Department of Microbiology & Immunology, The Henry M Jackson Foundation for Military Medicine, Uniformed Services University, Bethesda, MD 20814, USA
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - Joseph J. Mattapallil
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
31
|
Link N, Harnish JM, Hull B, Gibson S, Dietze M, Mgbike UE, Medina-Balcazar S, Shah PS, Yamamoto S. A Zika virus protein expression screen in Drosophila to investigate targeted host pathways during development. Dis Model Mech 2024; 17:dmm050297. [PMID: 38214058 PMCID: PMC10924231 DOI: 10.1242/dmm.050297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 12/29/2023] [Indexed: 01/13/2024] Open
Abstract
In the past decade, Zika virus (ZIKV) emerged as a global public health concern. Although adult infections are typically mild, maternal infection can lead to adverse fetal outcomes. Understanding how ZIKV proteins disrupt development can provide insights into the molecular mechanisms of disease caused by this virus, which includes microcephaly. In this study, we generated a toolkit to ectopically express ZIKV proteins in vivo in Drosophila melanogaster in a tissue-specific manner using the GAL4/UAS system. We used this toolkit to identify phenotypes and potential host pathways targeted by the virus. Our work identified that expression of most ZIKV proteins caused scorable phenotypes, such as overall lethality, gross morphological defects, reduced brain size and neuronal function defects. We further used this system to identify strain-dependent phenotypes that may have contributed to the increased pathogenesis associated with the outbreak of ZIKV in the Americas in 2015. Our work demonstrates the use of Drosophila as an efficient in vivo model to rapidly decipher how pathogens cause disease and lays the groundwork for further molecular study of ZIKV pathogenesis in flies.
Collapse
Affiliation(s)
- Nichole Link
- Department of Neurobiology, University of Utah, Salt Lake City, UT, 84112, USA
- Howard Hughes Medical Institute, Baylor College of Medicine (BCM), Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, BCM, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - J. Michael Harnish
- Department of Molecular and Human Genetics, BCM, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Brooke Hull
- Department of Molecular and Human Genetics, BCM, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Postbaccalaureate Research Education Program (PREP), Houston, TX, 77030, USA
| | - Shelley Gibson
- Department of Molecular and Human Genetics, BCM, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Miranda Dietze
- Department of Neurobiology, University of Utah, Salt Lake City, UT, 84112, USA
| | | | - Silvia Medina-Balcazar
- Department of Molecular and Human Genetics, BCM, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Priya S. Shah
- Department of Chemical Engineering, Department of Microbiology and Molecular Genetics, University of California, Davis, CA, 95616, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, BCM, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Postbaccalaureate Research Education Program (PREP), Houston, TX, 77030, USA
- Department of Neuroscience, BCM, Houston, TX, 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX, 77030, USA
| |
Collapse
|
32
|
Caetano CCS, Azamor T, Meyer NM, Onwubueke C, Calabrese CM, Calabrese LH, Visperas A, Piuzzi NS, Husni ME, Foo SS, Chen W. Mechanistic insights into bone remodelling dysregulation by human viral pathogens. Nat Microbiol 2024; 9:322-335. [PMID: 38316931 PMCID: PMC11045166 DOI: 10.1038/s41564-023-01586-6] [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: 05/13/2022] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
Bone-related diseases (osteopathologies) associated with human virus infections have increased around the globe. Recent findings have highlighted the intricate interplay between viral infection, the host immune system and the bone remodelling process. Viral infections can disrupt bone homeostasis, contributing to conditions such as arthritis and soft tissue calcifications. Osteopathologies can occur after arbovirus infections such as chikungunya virus, dengue virus and Zika virus, as well as respiratory viruses, such as severe acute respiratory syndrome coronavirus 2 and enteroviruses such as Coxsackievirus B. Here we explore how human viruses dysregulate bone homeostasis, detailing viral factors, molecular mechanisms, host immune response changes and bone remodelling that ultimately result in osteopathologies. We highlight model systems and technologies to advance mechanistic understanding of viral-mediated bone alterations. Finally, we propose potential prophylactic and therapeutic strategies, introduce 'osteovirology' as a research field highlighting the underestimated roles of viruses in bone-related diseases, and discuss research avenues for further investigation.
Collapse
Affiliation(s)
- Camila C S Caetano
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tamiris Azamor
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nikki M Meyer
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chineme Onwubueke
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Cassandra M Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Leonard H Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Anabelle Visperas
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Nicolas S Piuzzi
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - M Elaine Husni
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Suan-Sin Foo
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| | - Weiqiang Chen
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
33
|
Yao M, Cheng Z, Li X, Li Y, Ye W, Zhang H, Liu H, Zhang L, Lei Y, Zhang F, Lv X. N6-methyladenosine modification positively regulate Japanese encephalitis virus replication. Virol J 2024; 21:23. [PMID: 38243270 PMCID: PMC10799421 DOI: 10.1186/s12985-023-02275-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024] Open
Abstract
N6-methyladenosine (m6A) is present in diverse viral RNA and plays important regulatory roles in virus replication and host antiviral innate immunity. However, the role of m6A in regulating JEV replication has not been investigated. Here, we show that the JEV genome contains m6A modification upon infection of mouse neuroblast cells (neuro2a). JEV infection results in a decrease in the expression of m6A writer METTL3 in mouse brain tissue. METTL3 knockdown by siRNA leads to a substantial decrease in JEV replication and the production of progeny viruses at 48 hpi. Mechanically, JEV triggered a considerable increase in the innate immune response of METTL3 knockdown neuro2a cells compared to the control cells. Our study has revealed the distinctive m6A signatures of both the virus and host in neuro2a cells infected with JEV, illustrating the positive role of m6A modification in JEV infection. Our study further enhances understanding of the role of m6A modification in Flaviviridae viruses.
Collapse
Affiliation(s)
- Min Yao
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhirong Cheng
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
- College of Life Science, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Xueyun Li
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
- College of Basic Medicine, Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Yuexiang Li
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Wei Ye
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Hui Zhang
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - He Liu
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Liang Zhang
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Yingfeng Lei
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China
| | - Fanglin Zhang
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China.
| | - Xin Lv
- Department of Microbiology, Airforce Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
34
|
Sabir AJ, Singh PP, Trus I, Le NPK, Karniychuk U. Asian Zika virus can acquire generic African-lineage mutations during in utero infection. Emerg Microbes Infect 2023; 12:2263592. [PMID: 37747060 PMCID: PMC10561574 DOI: 10.1080/22221751.2023.2263592] [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: 07/13/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
The Zika virus 2015 epidemic showed an unusual phenotype for human flaviviruses, specifically fetal infection. We previously showed that in utero inoculation with the Asian Zika virus isolated from the human sample causes persistent infection in porcine fetuses. Here, we characterized the evolution of the Asian Zika virus in the fetal brain and placenta. Interestingly, the Asian Zika virus acquired generic African lineage K101R (A408G) and R1609 K (G4932A) mutations during in utero infection. Both African mutations were nonsynonymous and had a high frequency of nearly 100% in the fetal brain. Then, we synthetically generated the wild-type Asian variant and fetal brain-specific variant with generic African-lineage K101R and R1609 K mutations. In mosquito C6/36 cells, but not in human and pig cells, the fetal brain-specific variant showed higher virus loads compared to the Asian wild-type prototype. While in utero infection with both variants caused comparable virus loads in the placenta and amniotic fluids, fetuses injected with the fetal brain-specific variant had the trend to higher virus loads in lymph nodes. Also, introduced K101R and R1609 K mutations were stable and had high nearly 100% frequency at 28 days after in utero inoculation in both directly injected and trans-infected fetuses. These findings evoke concerns because Zika persists in pig herds and mosquitoes on farms in Mexico. It will be essential to identify how persistent in utero infection affects virus evolution and whether in utero-emerged Zika variants have the potential for shedding into the environment, more efficient transmission, and more aggressive infection phenotypes.
Collapse
Affiliation(s)
- Ahmad Jawad Sabir
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, Canada
| | - Prince Pal Singh
- School of Public Health, University of Saskatchewan, Saskatoon, Canada
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Ivan Trus
- Dioscuri Centre for RNA-Protein Interactions in Human Health and Disease, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Nguyen Phuong Khanh Le
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Uladzimir Karniychuk
- School of Public Health, University of Saskatchewan, Saskatoon, Canada
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
35
|
Lange RE, Dupuis Ii AP, Prusinski MA, Maffei JG, Koetzner CA, Ngo K, Backenson B, Oliver J, Vogels CBF, Grubaugh ND, Kramer LD, Ciota AT. Identification and characterization of novel lineage 1 Powassan virus strains in New York State. Emerg Microbes Infect 2023; 12:2155585. [PMID: 36503411 PMCID: PMC9788702 DOI: 10.1080/22221751.2022.2155585] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
Powassan virus (POWV, family Flaviviridae) is a reemerging tick-borne virus endemic in North America and Russia. In 1997, a POWV-like agent was isolated from Ixodes scapularis in New England and determined to be genetically distinct from the original POWV isolate. This revealed the existence of two lineages: lineage 1, prototype Powassan virus (POWV-1) and lineage 2, deer tick virus (DTV). POWV-1 is thought to be primarily maintained in a cycle between I. cookei and woodchucks and I. marxi and squirrels, while DTV is primarily maintained in a cycle between I. scapularis and small mammal hosts. Recent tick, mammalian, and human isolates from New York State (NYS) have been identified as DTV, but for the first time in 45 years, we detected four POWV-1 isolates, including the first reported isolation of POWV-1 from I. scapularis. We aimed to investigate genotypic and phenotypic characteristics of recent NYS isolates through sequence analysis and evaluation of replication kinetics in vitro and in vivo. Our sequencing revealed genetic divergence between NYS POWV-1 isolates, with two distinct foci. We found that POWV-1 isolates displayed variable replication kinetics in nymphal ticks but not in cell culture. POWV-1 isolated from I. scapularis displayed increased fitness in experimentally infected I. scapularis as compared to historic and recent POWV-1 isolates from I. cookei. These data suggest the emergence of divergent POWV-1 strains in alternate tick hosts and maintenance of genetically and phenotypically discrete POWV-1 foci.
Collapse
Affiliation(s)
- Rachel E Lange
- Department of Biomedical Sciences, State University of New York University at Albany School of Public Health, Albany, NY, USA
- New York State Department of Health, The Arbovirus Laboratory, Wadsworth Center, Slingerlands, NY, USA
| | - Alan P Dupuis Ii
- New York State Department of Health, The Arbovirus Laboratory, Wadsworth Center, Slingerlands, NY, USA
| | - Melissa A Prusinski
- New York State Department of Health, Bureau of Communicable Disease Control, Vector Ecology Laboratory, Albany, NY, USA
| | - Joseph G Maffei
- New York State Department of Health, The Arbovirus Laboratory, Wadsworth Center, Slingerlands, NY, USA
| | - Cheri A Koetzner
- New York State Department of Health, The Arbovirus Laboratory, Wadsworth Center, Slingerlands, NY, USA
| | - Kiet Ngo
- New York State Department of Health, The Arbovirus Laboratory, Wadsworth Center, Slingerlands, NY, USA
| | - Bryon Backenson
- New York State Department of Health, Bureau of Communicable Disease Control, Vector Ecology Laboratory, Albany, NY, USA
| | - JoAnne Oliver
- New York State Department of Health, Bureau of Communicable Disease Control, Syracuse, NY, USA
| | - Chantal B F Vogels
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Laura D Kramer
- Department of Biomedical Sciences, State University of New York University at Albany School of Public Health, Albany, NY, USA
- New York State Department of Health, The Arbovirus Laboratory, Wadsworth Center, Slingerlands, NY, USA
| | - Alexander T Ciota
- Department of Biomedical Sciences, State University of New York University at Albany School of Public Health, Albany, NY, USA
- New York State Department of Health, The Arbovirus Laboratory, Wadsworth Center, Slingerlands, NY, USA
| |
Collapse
|
36
|
Khongwichit S, Chuchaona W, Vongpunsawad S, Poovorawan Y. Molecular epidemiology, clinical analysis, and genetic characterization of Zika virus infections in Thailand (2020-2023). Sci Rep 2023; 13:21030. [PMID: 38030715 PMCID: PMC10687007 DOI: 10.1038/s41598-023-48508-4] [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: 11/02/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
To investigate the clinical and molecular characteristics and evolution of the Zika virus (ZIKV) in Thailand from March 2020 to March 2023. In all, 751 serum samples from hospitalized patients in Bangkok and the surrounding areas were screened for ZIKV using real-time RT-PCR. Demographic data and clinical variables were evaluated. Phylogenetic and molecular clock analysis determined the genetic relationships among the ZIKV strains, emergence timing, and their molecular characteristics. Among the 90 confirmed ZIKV cases, there were no significant differences in infection prevalence when comparing age groups and sexes. Rash was strongly associated with ZIKV infection. Our ZIKV Thai isolates were categorized into two distinct clades: one was related to strains from Myanmar, Vietnam, Oceania, and various countries in the Americas, and the other was closely related to previously circulating strains in Thailand, one of which shared a close relation to a neurovirulent ZIKV strain from Cambodia. Moreover, ZIKV Thai strains could be further classified into multiple sub-clades, each exhibiting specific mutations suggesting the genetic diversity among the circulating strains of ZIKV in Thailand. Understanding ZIKV epidemiology and genetic diversity is crucial for tracking the virus's evolution and adapting prevention and control strategies.
Collapse
Affiliation(s)
- Sarawut Khongwichit
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Pathumwan, Bangkok, 10330, Thailand
| | - Watchaporn Chuchaona
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Pathumwan, Bangkok, 10330, Thailand
| | - Sompong Vongpunsawad
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Pathumwan, Bangkok, 10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
37
|
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
|
38
|
Song GY, Huang XY, He MJ, Zhou HY, Li RT, Tian Y, Wang Y, Cheng ML, Chen X, Zhang RR, Zhou C, Zhou J, Fang XY, Li XF, Qin CF. A single amino acid substitution in the capsid protein of Zika virus contributes to a neurovirulent phenotype. Nat Commun 2023; 14:6832. [PMID: 37884553 PMCID: PMC10603150 DOI: 10.1038/s41467-023-42676-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Increasing evidence shows the African lineage Zika virus (ZIKV) displays a more severe neurovirulence compared to the Asian ZIKV. However, viral determinants and the underlying mechanisms of enhanced virulence phenotype remain largely unknown. Herein, we identify a panel of amino acid substitutions that are unique to the African lineage of ZIKVs compared to the Asian lineage by phylogenetic analysis and sequence alignment. We then utilize reverse genetic technology to generate recombinant ZIKVs incorporating these lineage-specific substitutions based on an infectious cDNA clone of Asian ZIKV. Through in vitro characterization, we discover a mutant virus with a lysine to arginine substitution at position 101 of capsid (C) protein (termed K101R) displays a larger plaque phenotype, and replicates more efficiently in various cell lines. Moreover, K101R replicates more efficiently in mouse brains and induces stronger inflammatory responses than the wild type (WT) virus in neonatal mice. Finally, a combined analysis reveals the K101R substitution promotes the production of mature C protein without affecting its binding to viral RNA. Our study identifies the role of K101R substitution in the C protein in contributing to the enhanced virulent phenotype of the African lineage ZIKV, which expands our understanding of the complexity of ZIKV proteins.
Collapse
Affiliation(s)
- Guang-Yuan Song
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Xing-Yao Huang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Meng-Jiao He
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Hang-Yu Zhou
- Suzhou Institute of System Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 215123, Suzhou, Jiangsu, China
| | - Rui-Ting Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Ying Tian
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Yan Wang
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Meng-Li Cheng
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Xiang Chen
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Rong-Rong Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Chao Zhou
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Jia Zhou
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China
| | - Xian-Yang Fang
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China.
| | - Cheng-Feng Qin
- School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, Anhui, China.
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China.
| |
Collapse
|
39
|
Gupta Y, Baranwal M, Chudasama B. Immunoinformatics-Based Identification of the Conserved Immunogenic Peptides Targeting of Zika Virus Precursor Membrane Protein. Viral Immunol 2023; 36:503-519. [PMID: 37486711 DOI: 10.1089/vim.2023.0015] [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] [Indexed: 07/25/2023] Open
Abstract
Zika virus infections lead to neurological complications such as congenital Zika syndrome and Guillain-Barré syndrome. Rising Zika infections in newborns and adults have triggered the need for vaccine development. In the current study, the precursor membrane (prM) protein of the Zika virus is explored for its functional importance and design of epitopes enriched conserved peptides with the usage of different immunoinformatics approach. Phylogenetic and mutational analyses inferred that the prM protein is highly conserved. Three conserved peptides containing multiple T and B cell epitopes were designed by employing different epitope prediction algorithms. IEDB population coverage analysis of selected peptides in six different continents has shown the population coverage of 60-99.8% (class I HLA) and 80-100% (class II HLA). Molecular docking of selected peptides/epitopes was carried out with each of class I and II HLA alleles using HADDOCK. A majority of peptide-HLA complex (pHLA) have HADDOCK scores found to be comparable and more than native-HLA complex representing the good binding interaction of peptides to HLA. Molecular dynamics simulation with best docked pHLA complexes revealed that pHLA complexes are stable with RMSD <5.5Å. Current work highlights the importance of prM as a strong antigenic protein and selected peptides have the potential to elicit humoral and cell-mediated immune responses.
Collapse
Affiliation(s)
- Yogita Gupta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Bhupendra Chudasama
- School of Physics & Materials Science, Thapar Institute of Engineering and Technology, Patiala, India
| |
Collapse
|
40
|
Evans AB, Winkler CW, Anzick SL, Ricklefs SM, Sturdevant DE, Peterson KE. Zika virus diversity in mice is maintained during early vertical transmission from placenta to fetus, but reduced in fetal bodies and brains at late stages of infection. PLoS Negl Trop Dis 2023; 17:e0011657. [PMID: 37796973 PMCID: PMC10581492 DOI: 10.1371/journal.pntd.0011657] [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: 05/12/2023] [Revised: 10/17/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
Since emerging in French Polynesia and Brazil in the 2010s, Zika virus (ZIKV) has been associated with fetal congenital disease. Previous studies have compared ancestral and epidemic ZIKV strains to identify strain differences that may contribute to vertical transmission and fetal disease. However, within-host diversity in ZIKV populations during vertical transmission has not been well studied. Here, we used the established anti-interferon treated Rag1-/- mouse model of ZIKV vertical transmission to compare genomic variation within ZIKV populations in matched placentas, fetal bodies, and fetal brains via RNASeq. At early stages of vertical transmission, the ZIKV populations in the matched placentas and fetal bodies were similar. Most ZIKV single nucleotide variants were present in both tissues, indicating little to no restriction in transmission of ZIKV variants from placenta to fetus. In contrast, at later stages of fetal infection there was a sharp reduction in ZIKV diversity in fetal bodies and fetal brains. All fetal brain ZIKV populations were comprised of one of two haplotypes, containing either a single variant or three variants together, as largely homogenous populations. In most cases, the dominant haplotype present in the fetal brain was also the dominant haplotype present in the matched fetal body. However, in two of ten fetal brains the dominant ZIKV haplotype was undetectable or present at low frequencies in the matched placenta and fetal body ZIKV populations, suggesting evidence of a strict selective bottleneck and possible selection for certain variants during neuroinvasion of ZIKV into fetal brains.
Collapse
Affiliation(s)
- Alyssa B. Evans
- Laboratory of Neurological Infections and Immunity, Neuroimmunology Section; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases (NIAID); National Institutes of Health (NIH); Hamilton, Montana, United States of America
| | - Clayton W. Winkler
- Laboratory of Neurological Infections and Immunity, Neuroimmunology Section; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases (NIAID); National Institutes of Health (NIH); Hamilton, Montana, United States of America
| | - Sarah L. Anzick
- Genomics Research Section, Research Technologies Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases (NIAID); National Institutes of Health (NIH); Hamilton, Montana, United States of America
| | - Stacy M. Ricklefs
- Genomics Research Section, Research Technologies Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases (NIAID); National Institutes of Health (NIH); Hamilton, Montana, United States of America
| | - Dan E. Sturdevant
- Genomics Research Section, Research Technologies Branch; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases (NIAID); National Institutes of Health (NIH); Hamilton, Montana, United States of America
| | - Karin E. Peterson
- Laboratory of Neurological Infections and Immunity, Neuroimmunology Section; Rocky Mountain Laboratories; National Institute of Allergy and Infectious Diseases (NIAID); National Institutes of Health (NIH); Hamilton, Montana, United States of America
| |
Collapse
|
41
|
Mantena S, Pillai PP, Petros BA, Welch NL, Myhrvold C, Sabeti PC, Metsky HC. Model-directed generation of CRISPR-Cas13a guide RNAs designs artificial sequences that improve nucleic acid detection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.557569. [PMID: 37786711 PMCID: PMC10541601 DOI: 10.1101/2023.09.20.557569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Generating maximally-fit biological sequences has the potential to transform CRISPR guide RNA design as it has other areas of biomedicine. Here, we introduce model-directed exploration algorithms (MEAs) for designing maximally-fit, artificial CRISPR-Cas13a guides-with multiple mismatches to any natural sequence-that are tailored for desired properties around nucleic acid diagnostics. We find that MEA-designed guides offer more sensitive detection of diverse pathogens and discrimination of pathogen variants compared to guides derived directly from natural sequences, and illuminate interpretable design principles that broaden Cas13a targeting.
Collapse
Affiliation(s)
- Sreekar Mantena
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | | | - Brittany A. Petros
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/Massachusetts Institute of Technology, MD-PhD Program, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Pardis C. Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | |
Collapse
|
42
|
Tian Z, Zhang H, Yu R, Du J, Gao S, Wang Q, Guan G, Yin H. The GTPase activity and isoprenylation of Swine GBP1 are critical for inhibiting the production of Japanese Encephalitis Virus. Vet Microbiol 2023; 284:109843. [PMID: 37540998 DOI: 10.1016/j.vetmic.2023.109843] [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: 11/27/2022] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/06/2023]
Abstract
Japanese encephalitis virus (JEV) is a flavivirus that cause severe neurological deficits. The guanylate-binding protein 1 (GBP1) gene is an interferon-stimulated gene and exerts antiviral functions on many RNA and DNA viruses via diverse mechanisms, however, the roles and the action modes of GBP1 in the antiviral effect on the production of JEV RNA and infectious virions remain to be clarified. In this study, we found that the RNA levels of swine GBP1 (sGBP1) in PK15 cells were up-regulated at the late stage of JEV infection. The overexpression of sGBP1 significantly inhibited the production of JEV while the knockdown of sGBP1 promoted the production of JEV. The GTPase activity and isoprenylation of sGBP1 both are critical for anti-JEV activity. The GTPase activity of sGBP1 is responsible for inhibiting the production of JEV genomic RNA. The isoprenylation of sGBP1 inhibited the expression and cleavage of JEV prM to decrease the yields of infectious virions, which may be associated with the interaction between sGBP1 and cellular proprotein convertase furin. Taken together, the study dissected the action modes of sGBP1with potent anti-JEV activity in more details.
Collapse
Affiliation(s)
- Zhancheng Tian
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China.
| | - Hongge Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Ruiming Yu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Junzheng Du
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Shandian Gao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Qiongjie Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China
| | - Hong Yin
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China
| |
Collapse
|
43
|
Recaioglu H, Kolk SM. Developing brain under renewed attack: viral infection during pregnancy. Front Neurosci 2023; 17:1119943. [PMID: 37700750 PMCID: PMC10493316 DOI: 10.3389/fnins.2023.1119943] [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: 12/09/2022] [Accepted: 04/26/2023] [Indexed: 09/14/2023] Open
Abstract
Living in a globalized world, viral infections such as CHIKV, SARS-COV-2, and ZIKV have become inevitable to also infect the most vulnerable groups in our society. That poses a danger to these populations including pregnant women since the developing brain is sensitive to maternal stressors including viral infections. Upon maternal infection, the viruses can gain access to the fetus via the maternofetal barrier and even to the fetal brain during which factors such as viral receptor expression, time of infection, and the balance between antiviral immune responses and pro-viral mechanisms contribute to mother-to-fetus transmission and fetal infection. Both the direct pro-viral mechanisms and the resulting dysregulated immune response can cause multi-level impairment in the maternofetal and brain barriers and the developing brain itself leading to dysfunction or even loss of several cell populations. Thus, maternal viral infections can disturb brain development and even predispose to neurodevelopmental disorders. In this review, we discuss the potential contribution of maternal viral infections of three relevant relative recent players in the field: Zika, Chikungunya, and Severe Acute Respiratory Syndrome Coronavirus-2, to the impairment of brain development throughout the entire route.
Collapse
Affiliation(s)
| | - Sharon M. Kolk
- Faculty of Science, Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
44
|
Bayandin RB, Makenov MT, Boumbaly S, Stukolova OA, Gladysheva AV, Shipovalov AV, Skarnovich MO, Camara O, Toure AH, Svyatchenko VA, Shvalov AN, Ternovoi VA, Boiro MY, Agafonov AP, Karan LS. The First Case of Zika Virus Disease in Guinea: Description, Virus Isolation, Sequencing, and Seroprevalence in Local Population. Viruses 2023; 15:1620. [PMID: 37631963 PMCID: PMC10459603 DOI: 10.3390/v15081620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/29/2023] Open
Abstract
The Zika virus (ZIKV) is a widespread mosquito-borne pathogen. Phylogenetically, two lineages of ZIKV are distinguished: African and Asian-American. The latter became the cause of the 2015-2016 pandemic, with severe consequences for newborns. In West African countries, the African lineage was found, but there is evidence of the emergence of the Asian-American lineage in Cape Verde and Angola. This highlights the need to not only monitor ZIKV but also sequence the isolates. In this article, we present a case report of Zika fever in a pregnant woman from Guinea identified in 2018. Viral RNA was detected through qRT-PCR in a serum sample. In addition, the seroconversion of anti-Zika IgM and IgG antibodies was detected in repeated blood samples. Subsequently, the virus was isolated from the C6/36 cell line. The detected ZIKV belonged to the African lineage, the Nigerian sublineage. The strains with the closest sequences were isolated from mosquitoes in Senegal in 2011 and 2015. In addition, we conducted the serological screening of 116 blood samples collected from patients presenting to the hospital of Faranah with fevers during the period 2018-2021. As a result, it was found that IgM-positive patients were identified each year and that the seroprevalence varied between 5.6% and 17.1%.
Collapse
Affiliation(s)
- Roman B. Bayandin
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | - Marat T. Makenov
- Central Research Institute of Epidemiology, Novogireevskaya St. 3A, 111123 Moscow, Russia
| | - Sanaba Boumbaly
- Virology Research Center/Laboratory of Viral Hemorrhagic Fevers, Conakry, Guinea
| | - Olga A. Stukolova
- Central Research Institute of Epidemiology, Novogireevskaya St. 3A, 111123 Moscow, Russia
| | - Anastasia V. Gladysheva
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | - Andrey V. Shipovalov
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | - Maksim O. Skarnovich
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | | | - Aboubacar Hady Toure
- Research Institute of Applied Biology of Guinea, Pastoria, CREMS, Kindia, Guinea
| | - Victor A. Svyatchenko
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | - Alexander N. Shvalov
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | - Vladimir A. Ternovoi
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | - Mamadou Y. Boiro
- Research Institute of Applied Biology of Guinea, Pastoria, CREMS, Kindia, Guinea
| | - Alexander P. Agafonov
- State Research Center of Virology and Biotechnology «Vector», 630559 Kol’tsovo, Novosibirsk Oblast, Russia (M.O.S.)
| | - Lyudmila S. Karan
- Central Research Institute of Epidemiology, Novogireevskaya St. 3A, 111123 Moscow, Russia
| |
Collapse
|
45
|
Huang Y, Li Q, Kang L, Li B, Ye H, Duan X, Xie H, Jiang M, Li S, Zhu Y, Tan Q, Chen L. Mitophagy Activation Targeting PINK1 Is an Effective Treatment to Inhibit Zika Virus Replication. ACS Infect Dis 2023; 9:1424-1436. [PMID: 37300493 DOI: 10.1021/acsinfecdis.3c00196] [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] [Indexed: 06/12/2023]
Abstract
Mitophagy is a selective degradation mechanism that maintains mitochondrial homeostasis by eliminating damaged mitochondria. Many viruses manipulate mitophagy to promote their infection, but its role in Zika virus (ZIKV) is unclear. In this study, we investigated the effect of mitophagy activation on ZIKV replication by the mitochondrial uncoupling agent niclosamide. Our results demonstrate that niclosamide-induced mitophagy inhibits ZIKV replication by eliminating fragmented mitochondria, both in vitro and in a mouse model of ZIKV-induced necrosis. Niclosamide induces autophosphorylation of PTEN-induced putative kinase 1 (PINK1), leading to the recruitment of PRKN/Parkin to the outer mitochondrial membrane and subsequent phosphorylation of ubiquitin. Knockdown of PINK1 promotes ZIKV infection and rescues the anti-ZIKV effect of mitophagy activation, confirming the role of ubiquitin-dependent mitophagy in limiting ZIKV replication. These findings demonstrate the role of mitophagy in the host response in limiting ZIKV replication and identify PINK1 as a potential therapeutic target in ZIKV infection.
Collapse
Affiliation(s)
- Yike Huang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
| | - Qingyuan Li
- North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Lan Kang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
| | - Bin Li
- Joint Laboratory on Transfusion-transmitted Infectious Diseases between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Nanning City, Nanning 530007, Guangxi, China
| | - Haiyan Ye
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
| | - Xiaoqiong Duan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
| | - He Xie
- The Hospital of Xidian Group, Xian 710077, Shaanxi, China
| | - Man Jiang
- Department of Pharmacology, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150000, Heilongjiang, China
| | - Shilin Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
| | - Ya Zhu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
| | - Qi Tan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
| | - Limin Chen
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu 610052, Sichuan, China
- Joint Laboratory on Transfusion-transmitted Infectious Diseases between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Nanning City, Nanning 530007, Guangxi, China
- The Hospital of Xidian Group, Xian 710077, Shaanxi, China
| |
Collapse
|
46
|
Tian L, Liang C, Huang X, Liu Z, Su J, Guo C, Zhu G, Sun J. Genomic epidemiology of dengue in Shantou, China, 2019. Front Public Health 2023; 11:1035060. [PMID: 37522010 PMCID: PMC10374217 DOI: 10.3389/fpubh.2023.1035060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Objectives Dengue has been endemic in Southeast Asian countries for decades. There are few reports tracing the dynamics of dengue in real time. In this study, we generated hundreds of pathogen genomes to understand the genomic epidemiology of an outbreak in a hyper-endemic area of dengue. Methods We leveraged whole-genome short-read sequencing (PE150) to generate genomes of the dengue virus and investigated the genomic epidemiology of a dengue virus transmission in a mesoscale outbreak in Shantou, China, in 2019. Results The outbreak was sustained from July to December 2019. The total accumulated number of laboratory-confirmed cases was 944. No gender bias or fatalities were recorded. Cambodia and Singapore were the main sources of imported dengue cases (74.07%, n = 20). A total of 284 dengue virus strains were isolated, including 259 DENV-1, 24 DENV-2, and 1 DENV-3 isolates. We generated the entire genome of 252 DENV isolates (229 DENV-1, 22 DENV-2, and 1 DENV-3), which represented 26.7% of the total cases. Combined epidemiological and phylogenetic analyses indicated multiple independent introductions. The internal transmission evaluations and transmission network reconstruction supported the inference of phylodynamic analysis, with high Bayes factor support in BSSVS analysis. Two expansion founders and transmission chains were detected in CCH and LG of Shantou. Conclusions We observed the instant effects of genomic epidemiology in monitoring the dynamics of DENV and highlighted its prospects for real-time tracing of outbreaks of other novel agents in the future.
Collapse
Affiliation(s)
- Lina Tian
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Provincial Institute of Public Health, Guangzhou, China
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
- School of Mathematics and Computing Science, Guilin University of Electronic Technology, Guilin, China
| | - Chumin Liang
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Provincial Institute of Public Health, Guangzhou, China
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Xiaorong Huang
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Provincial Institute of Public Health, Guangzhou, China
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhe Liu
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Provincial Institute of Public Health, Guangzhou, China
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Juan Su
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Chuan Guo
- Center for Disease Control and Prevention of Shantou City, Shantou, Guangdong, China
| | - Guanghu Zhu
- School of Mathematics and Computing Science, Guilin University of Electronic Technology, Guilin, China
| | - Jiufeng Sun
- Guangdong Provincial Center for Disease Control and Prevention, Guangdong Provincial Institute of Public Health, Guangzhou, China
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
- School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Álvarez-Díaz DA, Usme-Ciro JA, Corchuelo S, Naizaque JR, Rivera JA, Castiblanco-Martínez HD, Torres-Fernández O, Rengifo AC. 5'/3' RACE method for sequencing the 5' and 3' untranslated regions of Zika virus. Arch Virol 2023; 168:204. [PMID: 37428234 DOI: 10.1007/s00705-023-05820-8] [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: 09/12/2022] [Accepted: 05/19/2023] [Indexed: 07/11/2023]
Abstract
The spread of Zika virus (ZIKV) from the African continent to the Americas promoted its molecular evolution, as reflected by mutations in its RNA genome. Most of the ZIKV genome sequences in the GenBank database have incomplete 5' and 3' UTR sequences, reflecting the deficiency of whole-genome sequencing technologies to resolve the sequences of the genome ends. We modified a protocol for rapid amplification of cDNA ends (RACE) to determine the complete sequences of the 5' and 3' UTRs of a previously reported ZIKV isolate (GenBank no. MH544701.1). This strategy is useful for determining 5' and 3' UTR sequences of ZIKV isolates and will be useful for comparative genomics applications.
Collapse
Affiliation(s)
- Diego Alejandro Álvarez-Díaz
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá, D.C., Colombia
- Grupo de Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá, D.C., Colombia
| | - José Aldemar Usme-Ciro
- Centro de Investigación en Salud para el Trópico-CIST, Universidad Cooperativa de Colombia, Santa Marta, Colombia
| | - Sheryll Corchuelo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá, D.C., Colombia
| | - Julián Ricardo Naizaque
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá, D.C., Colombia
| | - Jorge Alonso Rivera
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá, D.C., Colombia
| | | | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá, D.C., Colombia
| | - Aura Caterine Rengifo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud, Bogotá, D.C., Colombia.
| |
Collapse
|
48
|
Barbosa MD, Costa A, Prieto-Oliveira P, Andreata-Santos R, Peter CM, Zanotto PMA, Janini LMR. Proposal of Model for Evaluation of Viral Kinetics of African/Asian/Brazilian- Zika virus Strains (Step Growth Curve) in Trophoblastic Cell Lines. Viruses 2023; 15:1446. [PMID: 37515134 PMCID: PMC10386092 DOI: 10.3390/v15071446] [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: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
The Zika virus (ZIKV) epidemic brought new discoveries regarding arboviruses, especially flaviviruses, as ZIKV was described as sexually and vertically transmitted. The latter shows severe consequences for the embryo/fetus, such as congenital microcephaly and deficiency of the neural system, currently known as Congenital ZIKV Syndrome (CZS). To better understand ZIKV dynamics in trophoblastic cells present in the first trimester of pregnancy (BeWo, HTR-8, and control cell HuH-7), an experiment of viral kinetics was performed for African MR766 low passage and Asian-Brazilian IEC ZIKV lineages. The results were described independently and demonstrated that the three placental cells lines are permissive and susceptible to ZIKV. We noticed cytopathic effects that are typical in in vitro viral infection in BeWo and HTR-8. Regarding kinetics, MR766lp showed peaks of viral loads in 24 and 48 hpi for all cell types tested, as well as marked cells death after peak production. On the other hand, the HTR-8 lineage inoculated with ZIKV-IEC exhibited increased viral production in 144 hpi, with a peak between 24 and 96 hpi. Furthermore, IEC had peak variations of viral production for BeWo in 144 hpi. Considering such in vitro results, the hypothesis that maternal fetal transmission is probably a way of virus transmission between the mother and the embryo/fetus is maintained.
Collapse
Affiliation(s)
- Márcia Duarte Barbosa
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-000, Brazil
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Anderson Costa
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Paula Prieto-Oliveira
- Department of Bioinformatics and Genomics, College of Computing and Informatics, University of North Carolina at Charlotte, 9331 Robert D. Snyder Rd., Charlotte, NC 28223, USA
| | - Robert Andreata-Santos
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Cristina M Peter
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Paolo M A Zanotto
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Luiz Mario Ramos Janini
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| |
Collapse
|
49
|
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
|
50
|
Santamaría G, Rengifo AC, Torres-Fernández O. NeuN distribution in brain structures of normal and Zika-infected suckling mice. J Mol Histol 2023:10.1007/s10735-023-10128-7. [PMID: 37199896 DOI: 10.1007/s10735-023-10128-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023]
Abstract
Microcephaly is the more severe brain malformation because of Zika virus infection. Increased vulnerability of neural stem and progenitor cells to Zika infection during prenatal neurodevelopment impairs the complete formation of cortical layers. Normal development of cerebellum is also affected. However, the follow-up of apparently healthy children born to Zika exposed mothers during pregnancy has revealed other neurological sequelae. This suggests Zika infection susceptibility remains in nervous tissue after neurogenesis end, when differentiated neuronal populations predominate. The neuronal nuclear protein (NeuN) is an exclusive marker of postmitotic neurons. Changes in NeuN expression are associated with neuronal degeneration. We have evaluated immunohistochemical expression of NeuN protein in cerebral cortex, hippocampus, and cerebellum of normal and Zika-infected neonatal Balb/c mice. The highest NeuN immunoreactivity was found mainly in neurons of all cortical layers, pyramidal layer of hippocampus, granular layer of dentate gyrus and in internal granular layer of cerebellum. Viral infection caused marked loss of NeuN immunostaining in all these brain areas. This suggests neurodegenerative effects of Zika virus infection during postmitotic neuron maturation and contribute to interpretation of neuropathogenic mechanisms of Zika.
Collapse
Affiliation(s)
- Gerardo Santamaría
- Grupo de Morfología Celular, Instituto Nacional de Salud (INS), Av. Calle 26 No. 51-20, Bogotá, 111321, DC, Colombia
| | - Aura Caterine Rengifo
- Grupo de Morfología Celular, Instituto Nacional de Salud (INS), Av. Calle 26 No. 51-20, Bogotá, 111321, DC, Colombia
| | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Instituto Nacional de Salud (INS), Av. Calle 26 No. 51-20, Bogotá, 111321, DC, Colombia.
| |
Collapse
|