1
|
Mousavi MJ, Arefinia N, Azarsa M, Hoseinnezhad T, Behboudi E. MicroRNA profiles in Zika virus infection: Insights from diverse sources. Indian J Med Microbiol 2024; 51:100697. [PMID: 39103054 DOI: 10.1016/j.ijmmb.2024.100697] [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: 11/08/2023] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Zika virus (ZIKV) stands as one of the most significant reemerging viral pathogens, linked to neurological diseases such as meningoencephalitis and congenital microcephaly. Today there are no effective therapies for treating ZIKV-infected patients. MiRNAs play a critical role in regulating cellular signaling and physiological conditions, and alterations in their profiles can bear great significance in disease progression. OBJECTIVES Despite significant progress in understanding the interaction between the ZIKV and its host since the outbreak, a more comprehensive understanding on these interactions is imperative. This review aims to summarize the studies in the field and shed light on the intricate relationship between ZIKV and its host at the molecular level. CONTENT We found that in ZIKV-infected humans, over-expression of miR-431-5p and miR-30e-5p plays a crucial role in innate immune responses and contributes to neurological damage. Additionally, in ZIKA-infected mice, we observed upregulated expression of all the targets of miR-124-3p including CCL2, IL7, IRF1, and SBNO2. Notably, other targets of this miRNA include TLR6, TNF, STAT3, and NF-kB also exhibited upregulation in the central nervous system (CNS) of infected mice. Conversely, miR-654-3p levels were reduced, correlating with the upregulation of its predicted targets including FLT3LG, LITAF, CD69, and TLR2. In the case of insects, aae-miR-286a/b-3p was predicted to target all ZIKV genotypes. This specific miRNA is typically found in ovaries and can be transferred to embryos. In conclusion, our findings suggest that host microRNAs and ZIKV-encoded microRNAs hold promise as potential targets for the diagnosis of ZIKV infections and may even serve as a therapeutic approach for managing this infectious disease.
Collapse
Affiliation(s)
- Mohammad Javad Mousavi
- Department of Hematology, School of Para-Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Nasir Arefinia
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mohammad Azarsa
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Taraneh Hoseinnezhad
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Emad Behboudi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| |
Collapse
|
2
|
Bahojb Mahdavi SZ, Jebelli A, Aghbash PS, Baradaran B, Amini M, Oroojalian F, Pouladi N, Baghi HB, de la Guardia M, Mokhtarzadeh AA. A comprehensive overview on the crosstalk between microRNAs and viral pathogenesis and infection. Med Res Rev 2024. [PMID: 39185567 DOI: 10.1002/med.22073] [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: 11/06/2021] [Revised: 04/11/2023] [Accepted: 08/04/2024] [Indexed: 08/27/2024]
Abstract
Infections caused by viruses as the smallest infectious agents, pose a major threat to global public health. Viral infections utilize different host mechanisms to facilitate their own propagation and pathogenesis. MicroRNAs (miRNAs), as small noncoding RNA molecules, play important regulatory roles in different diseases, including viral infections. They can promote or inhibit viral infection and have a pro-viral or antiviral role. Also, viral infections can modulate the expression of host miRNAs. Furthermore, viruses from different families evade the host immune response by producing their own miRNAs called viral miRNAs (v-miRNAs). Understanding the replication cycle of viruses and their relation with host miRNAs and v-miRNAs can help to find new treatments against viral infections. In this review, we aim to outline the structure, genome, and replication cycle of various viruses including hepatitis B, hepatitis C, influenza A virus, coronavirus, human immunodeficiency virus, human papillomavirus, herpes simplex virus, Epstein-Barr virus, Dengue virus, Zika virus, and Ebola virus. We also discuss the role of different host miRNAs and v-miRNAs and their role in the pathogenesis of these viral infections.
Collapse
Affiliation(s)
- Seyedeh Zahra Bahojb Mahdavi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asiyeh Jebelli
- Department of Biological Science, Faculty of Basic Science, Higher Education Institute of Rab-Rashid, Tabriz, Iran
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Burjassot, Valencia, Spain
| | | |
Collapse
|
3
|
Oh SJ, Kumari P, Auroni TT, Stone S, Pathak H, Elsharkawy A, Natekar JP, Shin OS, Kumar M. Upregulation of Neuroinflammation-Associated Genes in the Brain of SARS-CoV-2-Infected Mice. Pathogens 2024; 13:528. [PMID: 39057755 PMCID: PMC11280415 DOI: 10.3390/pathogens13070528] [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: 11/01/2023] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Neurological manifestations are a significant complication of coronavirus disease 2019 (COVID-19), but the underlying mechanisms are yet to be understood. Recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced neuroinvasion and encephalitis were observed in K18-hACE2 mice, leading to mortality. Our goal in this study was to gain insights into the molecular pathogenesis of neurological manifestations in this mouse model. To analyze differentially expressed genes (DEGs) in the brains of mice following SARS-CoV-2 infection, we performed NanoString gene expression analysis using three individual animal samples at 1, 3, and 6 days post-infection. We identified the DEGs by comparing them to animals that were not infected with the virus. We found that genes upregulated at day 6 post-infection were mainly associated with Toll-like receptor (TLR) signaling, RIG-I-like receptor (RLR) signaling, and cell death pathways. However, downregulated genes were associated with neurodegeneration and synaptic signaling pathways. In correlation with gene expression profiles, a multiplexed immunoassay showed the upregulation of multiple cytokines and chemokines involved in inflammation and cell death in SARS-CoV-2-infected brains. Furthermore, the pathway analysis of DEGs indicated a possible link between TLR2-mediated signaling pathways and neuroinflammation, as well as pyroptosis and necroptosis in the brain. In conclusion, our work demonstrates neuroinflammation-associated gene expression profiles, which can provide key insight into the severe disease observed in COVID-19 patients.
Collapse
Affiliation(s)
- Soo-Jin Oh
- BK21 Graduate Program, Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea;
| | - Pratima Kumari
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Tabassum Tasnim Auroni
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Shannon Stone
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Heather Pathak
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Amany Elsharkawy
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Janhavi Prasad Natekar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| | - Ok Sarah Shin
- BK21 Graduate Program, Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul 08308, Republic of Korea;
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA; (P.K.); (T.T.A.); (S.S.); (H.P.); (A.E.); (J.P.N.)
| |
Collapse
|
4
|
Song Q, Fan Y, Zhang H, Wang N. Z-DNA binding protein 1 orchestrates innate immunity and inflammatory cell death. Cytokine Growth Factor Rev 2024; 77:15-29. [PMID: 38548490 DOI: 10.1016/j.cytogfr.2024.03.005] [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] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 06/22/2024]
Abstract
Innate immunity is not only the first line of host defense against microbial infections but is also crucial for the host responses against a variety of noxious stimuli. Z-DNA binding protein 1 (ZBP1) is a cytosolic nucleic acid sensor that can induce inflammatory cell death in both immune and nonimmune cells upon sensing of incursive virus-derived Z-form nucleic acids and self-nucleic acids via its Zα domain. Mechanistically, aberrantly expressed or activated ZBP1 induced by pathogens or noxious stimuli enables recruitment of TANK binding kinase 1 (TBK1), interferon regulatory factor 3 (IRF3), receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 to drive type I interferon (IFN-I) responses and activation of nuclear factor kappa B (NF-κB) signaling. Meanwhile, ZBP1 promotes the assembly of ZBP1- and absent in melanoma 2 (AIM2)-PANoptosome, which ultimately triggers PANoptosis through caspase 3-mediated apoptosis, mixed lineage kinase domain like pseudokinase (MLKL)-mediated necroptosis, and gasdermin D (GSDMD)-mediated pyroptosis. In response to damaged mitochondrial DNA, ZBP1 can interact with cyclic GMP-AMP synthase to augment IFN-I responses but inhibits toll like receptor 9-mediated inflammatory responses. This review summarizes the structure and expression pattern of ZBP1, discusses its roles in human diseases through immune-dependent (e.g., the production of IFN-I and pro-inflammatory cytokines) and -independent (e.g., the activation of cell death) functions, and highlights the attractive prospect of manipulating ZBP1 as a promising therapeutic target in diseases.
Collapse
Affiliation(s)
- Qixiang Song
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China
| | - Yuhang Fan
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China.
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 110 Xiangya Road, Changsha 410083, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, 110 Xiangya Road, Changsha 410083, China.
| |
Collapse
|
5
|
Zhang X, Li Y, Cao Y, Wu Y, Cheng G. The Role of Noncoding RNA in the Transmission and Pathogenicity of Flaviviruses. Viruses 2024; 16:242. [PMID: 38400018 PMCID: PMC10892091 DOI: 10.3390/v16020242] [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: 12/11/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Noncoding RNAs (ncRNAs) constitute a class of RNA molecules that lack protein-coding capacity. ncRNAs frequently modulate gene expression through specific interactions with target proteins or messenger RNAs, thereby playing integral roles in a wide array of cellular processes. The Flavivirus genus comprises several significant members, such as dengue virus (DENV), Zika virus (ZIKV), and yellow fever virus (YFV), which have caused global outbreaks, resulting in high morbidity and mortality in human populations. The life cycle of arthropod-borne flaviviruses encompasses their transmission between hematophagous insect vectors and mammalian hosts. During this process, a complex three-way interplay occurs among the pathogen, vector, and host, with ncRNAs exerting a critical regulatory influence. ncRNAs not only constitute a crucial regulatory mechanism that has emerged from the coevolution of viruses and their hosts but also hold potential as antiviral targets for controlling flavivirus epidemics. This review introduces the biogenesis of flavivirus-derived ncRNAs and summarizes the regulatory roles of ncRNAs in viral replication, vector-mediated viral transmission, antiviral innate immunity, and viral pathogenicity. A profound comprehension of the interplay between ncRNAs and flaviviruses will help formulate efficacious prophylactic and therapeutic strategies against flavivirus-related diseases.
Collapse
Affiliation(s)
- Xianwen Zhang
- Shenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen 518000, China
| | - Yuhan Li
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; (Y.L.); (Y.C.)
| | - Yingyi Cao
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; (Y.L.); (Y.C.)
| | - Ying Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan 430072, China;
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; (Y.L.); (Y.C.)
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
- Southwest United Graduate School, Kunming 650092, China
| |
Collapse
|
6
|
Camacho-Concha N, Santana-Román ME, Sánchez NC, Velasco I, Pando-Robles V, Pedraza-Alva G, Pérez-Martínez L. Insights into Zika Virus Pathogenesis and Potential Therapeutic Strategies. Biomedicines 2023; 11:3316. [PMID: 38137537 PMCID: PMC10741857 DOI: 10.3390/biomedicines11123316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 12/24/2023] Open
Abstract
Zika virus (ZIKV) has emerged as a significant public health threat, reaching pandemic levels in 2016. Human infection with ZIKV can manifest as either asymptomatic or as an acute illness characterized by symptoms such as fever and headache. Moreover, it has been associated with severe neurological complications in adults, including Guillain-Barre syndrome, and devastating fetal abnormalities, like microcephaly. The primary mode of transmission is through Aedes spp. mosquitoes, and with half of the world's population residing in regions where Aedes aegypti, the principal vector, thrives, the reemergence of ZIKV remains a concern. This comprehensive review provides insights into the pathogenesis of ZIKV and highlights the key cellular pathways activated upon ZIKV infection. Additionally, we explore the potential of utilizing microRNAs (miRNAs) and phytocompounds as promising strategies to combat ZIKV infection.
Collapse
Affiliation(s)
- Nohemi Camacho-Concha
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Morelos, Mexico; (N.C.-C.); (M.E.S.-R.); (N.C.S.); (G.P.-A.)
| | - María E. Santana-Román
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Morelos, Mexico; (N.C.-C.); (M.E.S.-R.); (N.C.S.); (G.P.-A.)
| | - Nilda C. Sánchez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Morelos, Mexico; (N.C.-C.); (M.E.S.-R.); (N.C.S.); (G.P.-A.)
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Ciudad de México 14269, Mexico
| | - Victoria Pando-Robles
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Morelos, Mexico;
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Morelos, Mexico; (N.C.-C.); (M.E.S.-R.); (N.C.S.); (G.P.-A.)
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Morelos, Mexico; (N.C.-C.); (M.E.S.-R.); (N.C.S.); (G.P.-A.)
| |
Collapse
|
7
|
Gomes JA, Vieira IA, Sgarioni E, Terças-Tretell ACP, da Silva JH, Ribeiro BFR, Galera MF, de Oliveira TM, Carvalho de Andrade MDF, Carvalho IF, Schüler-Faccini L, Vianna FSL. Contribution of miR-124 rs531564 polymorphism to the occurrence of congenital Zika syndrome. Epigenetics 2023; 18:2145061. [PMID: 36411728 PMCID: PMC9980461 DOI: 10.1080/15592294.2022.2145061] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Zika virus (ZIKV) cause Congenital Zika Syndrome (CZS) in individuals exposed during pregnancy. Studies have shown that ZIKV infection positively regulates the miR-124 expression in neural cells, which leads to a decrease of TFRC, a gene targeted of this miRNA. Both miR-124 and TFRC exhibit a pivotal role in nervous system development. Therefore, in this study we aimed to investigate whether genetic variants that affect the expression of these genes could act together with ZIKV to increase the risk of individuals developing CZS. TFRC rs406271 and MIR-124-1 rs531564 polymorphisms were genotyped, using TaqMan® Genotyping Assays, in a sample of children who were exposed to ZIKV during pregnancy, of whom 40 were born with CZS and 48 without congenital anomalies. We identified that individuals with CZS presented a higher frequency of CG genotype of rs531564 polymorphism in MIR-124-1 (p=0.048), which is associated with increased expression of miR-124. Since ZIKV also upregulates the expression of this miRNA, the presence of CG genotype in individuals exposed to the virus could lead to a scenario of overexpression of miR-124 in the brain. Since teratogenesis is a multifactorial event, this genetic finding could partly explain why such individuals are more susceptible to CZS, considering both the downregulation of important neurodevelopment genes, as well as deregulation of the neurogenesis process. Thus, we provide preliminary evidence about a possible genetic risk factor to CZS and highlight the importance of analyzing functional polymorphisms related to epigenetic modulators of neurodevelopment genes in the context of ZIKV teratogenesis.
Collapse
Affiliation(s)
- Julia A Gomes
- Sistema Nacional de Informação sobre Agentes Teratogênicos (SIAT), Serviço de Genética Médica (SGM), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil,Instituto Nacional de Genética Médica Populacional (INAGEMP), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil,Laboratório de Medicina Genômica (LMG), Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Igor Araujo Vieira
- Laboratório de Medicina Genômica (LMG), Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil,Escola de Saúde, Universidade do Vale do Rio do Sinos (Unisinos), São Leopoldo, Brazil
| | - Eduarda Sgarioni
- Laboratório de Medicina Genômica (LMG), Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | | | - Juliana H da Silva
- Programa de Pós-graduação em Saúde Coletiva, Universidade Federal de Mato Grosso (UFMT), Cuiabá, Brazil,Secretaria Municipal de Saúde de Tangará da Serra, Tangará da Serra, Brazil
| | | | - Marcial F Galera
- Departamento de Pediatria, Faculdade de Medicina, Universidade Federal de Mato Grosso (UFMT), Cuiabá, Brazil
| | - Thalita M de Oliveira
- Hospital Universitário Júlio Müller (HUJM), Universidade Federal de Mato Grosso (UFMT), Empresa Brasileira de Serviços Hospitalares (EBSERH), Cuiabá, Brazil
| | | | | | - Lavínia Schüler-Faccini
- Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil,Sistema Nacional de Informação sobre Agentes Teratogênicos (SIAT), Serviço de Genética Médica (SGM), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil,Instituto Nacional de Genética Médica Populacional (INAGEMP), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Fernanda SL Vianna
- Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil,Sistema Nacional de Informação sobre Agentes Teratogênicos (SIAT), Serviço de Genética Médica (SGM), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil,Instituto Nacional de Genética Médica Populacional (INAGEMP), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil,Laboratório de Medicina Genômica (LMG), Centro de Pesquisa Experimental (CPE), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil,CONTACT Fernanda SL Vianna Programa de Pós-Graduação em Genética e Biologia Molecular (PPGBM), Departamento de Genética, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, ZIP91501-970, Brazil
| |
Collapse
|
8
|
Auroni TT, Arora K, Natekar JP, Pathak H, Elsharkawy A, Kumar M. The critical role of interleukin-6 in protection against neurotropic flavivirus infection. Front Cell Infect Microbiol 2023; 13:1275823. [PMID: 38053527 PMCID: PMC10694511 DOI: 10.3389/fcimb.2023.1275823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 12/07/2023] Open
Abstract
West Nile virus (WNV) and Japanese encephalitis virus (JEV) are emerging mosquito-borne flaviviruses causing encephalitis globally. No specific drug or therapy exists to treat flavivirus-induced neurological diseases. The lack of specific therapeutics underscores an urgent need to determine the function of important host factors involved in flavivirus replication and disease progression. Interleukin-6 (IL-6) upregulation has been observed during viral infections in both mice and humans, implying that it may influence the disease outcome significantly. Herein, we investigated the function of IL-6 in the pathogenesis of neurotropic flavivirus infections. First, we examined the role of IL-6 in flavivirus-infected human neuroblastoma cells, SK-N-SH, and found that IL-6 neutralization increased the WNV or JEV replication and inhibited the expression of key cytokines. We further evaluated the role of IL-6 by infecting primary mouse cells derived from IL-6 knockout (IL-6-/-) mice and wild-type (WT) mice with WNV or JEV. The results exhibited increased virus yields in the cells lacking the IL-6 gene. Next, our in vivo approach revealed that IL-6-/- mice had significantly higher morbidity and mortality after subcutaneous infection with the pathogenic WNV NY99 or JEV Nakayama strain compared to WT mice. The non-pathogenic WNV Eg101 strain did not cause mortality in WT mice but resulted in 60% mortality in IL-6-/- mice, indicating that IL-6 is required for the survival of mice after the peripheral inoculation of WNV or JEV. We also observed significantly higher viremia and brain viral load in IL-6-/- mice than in WT mice. Subsequently, we explored innate immune responses in WT and IL-6-/- mice after WNV NY99 infection. Our data demonstrated that the IL-6-/- mice had reduced levels of key cytokines in the serum during early infection but elevated levels of proinflammatory cytokines in the brain later, along with suppressed anti-inflammatory cytokines. In addition, mRNA expression of IFN-α and IFN-β was significantly lower in the infected IL-6-/- mice. In conclusion, these data suggest that the lack of IL-6 exacerbates WNV or JEV infection in vitro and in vivo by causing an increase in virus replication and dysregulating host immune response.
Collapse
Affiliation(s)
| | | | | | | | | | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
9
|
Ramphan S, Chumchanchira C, Sornjai W, Chailangkarn T, Jongkaewwattana A, Assavalapsakul W, Smith DR. Strain Variation Can Significantly Modulate the miRNA Response to Zika Virus Infection. Int J Mol Sci 2023; 24:16216. [PMID: 38003407 PMCID: PMC10671159 DOI: 10.3390/ijms242216216] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-transmitted virus that has emerged as a major public health concern due to its association with neurological disorders in humans, including microcephaly in fetuses. ZIKV infection has been shown to alter the miRNA profile in host cells, and these changes can contain elements that are proviral, while others can be antiviral in action. In this study, the expression of 22 miRNAs in human A549 cells infected with two different ZIKV isolates was investigated. All of the investigated miRNAs showed significant changes in expression at at least one time point examined. Markedly, 18 of the miRNAs examined showed statistically significant differences in expression between the two strains examined. Four miRNAs (miR-21, miR-34a, miR-128 and miR-155) were subsequently selected for further investigation. These four miRNAs were shown to modulate antiviral effects against ZIKV, as downregulation of their expression through anti-miRNA oligonucleotides resulted in increased virus production, whereas their overexpression through miRNA mimics reduced virus production. However, statistically significant changes were again seen when comparing the two strains investigated. Lastly, candidate targets of the miRNAs miR-34a and miR-128 were examined at the level of the mRNA and protein. HSP70 was identified as a target of miR-34a, but, again, the effects were strain type-specific. The two ZIKV strains used in this study differ by only nine amino acids, and the results highlight that consideration must be given to strain type variation when examining the roles of miRNAs in ZIKV, and probably other virus infections.
Collapse
Affiliation(s)
- Suwipa Ramphan
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand; (S.R.); (W.S.)
| | - Chanida Chumchanchira
- Department of Biology, Faculty of Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Wannapa Sornjai
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand; (S.R.); (W.S.)
| | - Thanathom Chailangkarn
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 12120, Thailand; (T.C.); (A.J.)
| | - Anan Jongkaewwattana
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 12120, Thailand; (T.C.); (A.J.)
| | - Wanchai Assavalapsakul
- Department of Microbiology, Faculty of Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Duncan R. Smith
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand; (S.R.); (W.S.)
| |
Collapse
|
10
|
Carneiro VCDS, Moreira ODC, Coelho WLDCNP, Rio BC, Sarmento DJDS, Salvio AL, Alves-Leon SV, de Paula VS, Leon LAA. miRNAs in Neurological Manifestation in Patients Co-Infected with SARS-CoV-2 and Herpesvírus 6 (HHV-6). Int J Mol Sci 2023; 24:11201. [PMID: 37446381 PMCID: PMC10342854 DOI: 10.3390/ijms241311201] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 07/15/2023] Open
Abstract
Human herpesviruses (HHVs) can establish latency and be reactivated, also are neurotropic viruses that can trigger neurological disorders. HHV-6 is a herpesvirus that is associated with neurological disorders. Studies have reported the detection of HHV-6 in patients with COVID-19 and neurological manifestations. However, specific diagnoses of the neurological disorders caused by these viruses tend to be invasive or difficult to interpret. This study aimed to establish a relationship between miRNA and neurological manifestations in patients co-infected with COVID-19 and HHV-6 and evaluate miRNAs as potential biomarkers. Serum samples from COVID-19 patients in the three cohorts were analyzed. miRNA analysis by real-time polymerase chain reaction (qPCR) revealed miRNAs associated with neuroinflammation were highly expressed in patients with neurological disorders and HHV-6 detection. When compared with the group of patients without detection of HHVs DNA and without neurological alterations, the group with detection of HHV-6 DNA and neurological alteration, displayed significant differences in the expression of mir-21, mir-146a, miR-155 and miR-let-7b (p < 0.01). Our results reinforce the involvement of miRNAs in neurological disorders and provide insights into their use as biomarkers for neurological disorders triggered by HHV-6. Furthermore, understanding the expression of miRNAs may contribute to therapeutic strategies.
Collapse
Affiliation(s)
- Vanessa Cristine de Souza Carneiro
- Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil (L.A.A.L.)
| | - Otacilio da Cruz Moreira
- Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Real Time PCR Platform RPT09A, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | | | - Beatriz Chan Rio
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil (L.A.A.L.)
| | | | - Andreza Lemos Salvio
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro 22290-240, Brazil
| | - Soniza Vieira Alves-Leon
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro 22290-240, Brazil
- Department of Neurology, Reference and Research Center for Multiple Sclerosis and Other Central Nervous System Idiopathic Demyelinating Inflammatory Diseases, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro 21941-617, Brazil
| | - Vanessa Salete de Paula
- Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Luciane Almeida Amado Leon
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil (L.A.A.L.)
| |
Collapse
|
11
|
Bhardwaj U, Singh SK. Zika virus NS1 suppresses VE-cadherin via hsa-miR-29b-3p/DNMT3b/MMP-9 pathway in human brain microvascular endothelial cells. Cell Signal 2023; 106:110659. [PMID: 36948479 DOI: 10.1016/j.cellsig.2023.110659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/14/2023] [Accepted: 03/19/2023] [Indexed: 03/24/2023]
Abstract
Zika virus infection has been reported to cause microcephaly in newborns. ZIKV exploits various strategies to cross the blood-brain barrier. ZIKV NS1 may compromise the barrier integrity of endothelial cells by regulating expression of junctional proteins. MicroRNAs play an important role in post-transcriptional gene regulations. We demonstrated that ZIKV-NS1 affected the adherence junction protein in human brain microvascular endothelial cells via hsa-miR-29b-3p/DNMT3b/MMP-9 pathway. The hCMEC/D3 cells were exposed to ZIKV-NS1 with different doses (500 ng/mL and 1000 ng/mL) for 24 h. The expression pattern of DNTM3b, MMP-9, and VE-cadherin were studied using immunoblotting and the distribution of DNMT3b and MMP-9 were studied using immunofluorescence. The quantification of hsa-miR-29b-3p was done through qRT-PCR. Direct regulation of DNMT3b by hsa-miR-29b-3p was demonstrated by overexpression of hsa-miR-29b-3p using hsa-miR-29b-3p mimic, and knockdown of hsa-miR-29b-3p by using hsa-miR-29b-3p inhibitors. The ZIKV-NS1 affected the barrier function of endothelial cells through the increased expression of hsa-miR29b-3p, which suppressed the DNMT3b, thus enhanced expression of MMP-9, which finally suppressed the expression of VE-cadherin. These findings suggested that ZIKV-NS1 alters the expression of Adherens Junction protein in human brain microvascular endothelial cells through hsa-miR-29b-3p/DNMT3b/MMP-9 pathway, which compromised the barrier function of human brain microvascular endothelial cells.
Collapse
Affiliation(s)
- Utkarsh Bhardwaj
- Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Sunit K Singh
- Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
12
|
Dong Y, Yan H, Li J, Bei L, Shi X, Zhu Y, Xie Z, Zhang R, Jiang S. miR-155-1 as a positive factor for novel duck reovirus replication by regulating SOCS5-mediated interferons. Virus Res 2023; 323:199003. [PMID: 36384170 PMCID: PMC10194143 DOI: 10.1016/j.virusres.2022.199003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 11/14/2022]
Abstract
Diseases caused by novel duck reovirus (NDRV) have brought considerable economic losses to the poultry industry. MicroRNAs (miRNAs) have an impact on virus replication and antiviral immunity. However, the miRNA profile upon NDRV infection in duck embryo fibroblasts (DEFs) remains to be discovered. In this study, small RNA (sRNA) sequencing was performed to decipher the cellular miRNA response to NDRV infection. Based on 26 differentially expressed miRNAs (19 upregulated and 7 downregulated miRNAs) obtained from sequencing data and their target genes predicted by software, GO and KEGG analyses were performed to elucidate the functions of miRNAs in NDRV invasion, replication, and virus spread. "FoxO signaling pathway", "autophagy", and "Toll-like receptor signaling pathway" might participate in NDRV replication as revealed by KEGG enrichment analysis. The miR-155-1 sequence was found to be identical to rno-miR-155-5p and was sharply increased with the progression of NDRV infection. Moreover, NDRV-induced miR-155-1 could act as a positive factor for virus replication in DEFs, which inhibited type I interferon (IFN-I) production. Luciferase assay confirmed that miR-155-1 disturbed the abundance of suppressor of cytokine signaling (SOCS) 5 by targeting 3'-UTR. SOCS5, which is linked to increased IRF7 expression, restricts IFN expression and promotes NDRV replication in DEFs. Therefore, this study proposed that miR-155-1 was used by NDRV to restrict SOCS5 expression, attenuating the production of IFN-I and creating a favorable environment for virus replication.
Collapse
Affiliation(s)
- Yu Dong
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China
| | - Hui Yan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China
| | - Jinman Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China
| | - Lei Bei
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China
| | - Xingxing Shi
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China
| | - Yanli Zhu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China
| | - Zhijin Xie
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China
| | - Ruihua Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China.
| | - Shijin Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Tai'an, China.
| |
Collapse
|
13
|
Hao Y, Yang B, Yang J, Shi X, Yang X, Zhang D, Zhao D, Yan W, Chen L, Zheng H, Zhang K, Liu X. ZBP1: A Powerful Innate Immune Sensor and Double-Edged Sword in Host Immunity. Int J Mol Sci 2022; 23:ijms231810224. [PMID: 36142136 PMCID: PMC9499459 DOI: 10.3390/ijms231810224] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/28/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Z-conformation nucleic acid binding protein 1 (ZBP1), a powerful innate immune sensor, has been identified as the important signaling initiation factor in innate immune response and the multiple inflammatory cell death known as PANoptosis. The initiation of ZBP1 signaling requires recognition of left-handed double-helix Z-nucleic acid (includes Z-DNA and Z-RNA) and subsequent signaling transduction depends on the interaction between ZBP1 and its adapter proteins, such as TANK-binding kinase 1 (TBK1), interferon regulatory factor 3 (IRF3), receptor-interacting serine/threonine-protein kinase 1 (RIPK1), and RIPK3. ZBP1 activated innate immunity, including type-I interferon (IFN-I) response and NF-κB signaling, constitutes an important line of defense against pathogenic infection. In addition, ZBP1-mediated PANoptosis is a double-edged sword in anti-infection, auto-inflammatory diseases, and tumor immunity. ZBP1-mediated PANoptosis is beneficial for eliminating infected cells and tumor cells, but abnormal or excessive PANoptosis can lead to a strong inflammatory response that is harmful to the host. Thus, pathogens and host have each developed multiplex tactics targeting ZBP1 signaling to maintain strong virulence or immune homeostasis. In this paper, we reviewed the mechanisms of ZBP1 signaling, the effects of ZBP1 signaling on host immunity and pathogen infection, and various antagonistic strategies of host and pathogen against ZBP1. We also discuss existent gaps regarding ZBP1 signaling and forecast potential directions for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Haixue Zheng
- Correspondence: (H.Z.); (K.Z.); Tel.: +86-15214078335 (K.Z.)
| | - Keshan Zhang
- Correspondence: (H.Z.); (K.Z.); Tel.: +86-15214078335 (K.Z.)
| | | |
Collapse
|
14
|
Involvement of host microRNAs in flavivirus-induced neuropathology: An update. J Biosci 2022. [PMID: 36222134 PMCID: PMC9425815 DOI: 10.1007/s12038-022-00288-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Flaviviruses are a spectrum of vector-borne RNA viruses that cause potentially severe diseases in humans including encephalitis, acute-flaccid paralysis, cognitive disorders and foetal abnormalities. Japanese encephalitis virus (JEV), Zika virus (ZIKV), West Nile virus (WNV) and Dengue virus (DENV) are globally emerging pathogens that lead to epidemics and outbreaks with continued transmission to newer geographical areas over time. In the past decade, studies have focussed on understanding the pathogenic mechanisms of these viruses in a bid to alleviate their disease burden. MicroRNAs (miRNAs) are short single-stranded RNAs that have emerged as master-regulators of cellular gene expression. The dynamics of miRNAs within a cell have the capacity to modulate hundreds of genes and, consequently, their physiological manifestation. Increasing evidence suggests their role in host response to disease and infection including cell survival, intracellular viral replication and immune activation. In this review, we aim to comprehensively update published evidence on the role of miRNAs in host cells infected with the common neurotropic flaviviruses, with an increased focus on neuropathogenic mechanisms. In addition, we briefly cover therapeutic advancements made in the context of miRNA-based antiviral strategies.
Collapse
|
15
|
Li K, Ji Q, Jiang S, Zhang N. Advancement in the Development of Therapeutics Against Zika Virus Infection. Front Cell Infect Microbiol 2022; 12:946957. [PMID: 35880081 PMCID: PMC9307976 DOI: 10.3389/fcimb.2022.946957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Zika virus (ZIKV), a re-emerging arbovirus, causes teratogenic effects on the fetus and normal nerve functions, resulting in harmful autoimmune responses, which call for the development of therapeutics against ZIKV infection. In this review, we introduce the pathogenesis of ZIKV infection and summarize the advancement in the development of therapeutics against ZIKV infection. It provides guidance for the development of effective therapeutics against ZIKV infection.
Collapse
Affiliation(s)
- Kangchen Li
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Qianting Ji
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE), National Health Commission (NHC) and Chinese Academy of Medical Sciences (CAMS), School of Basic Medical Sciences and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- *Correspondence: Shibo Jiang, ; Naru Zhang,
| | - Naru Zhang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
- *Correspondence: Shibo Jiang, ; Naru Zhang,
| |
Collapse
|
16
|
Carvalho-Silva AC, Da Silva Junior AR, Rigaud VOC, Martins WK, Coelho V, Pfrimer IAH, Kalil J, Fonseca SG, Cunha-Neto E, Ferreira LRP. A Major Downregulation of Circulating microRNAs in Zika Acutely Infected Patients: Potential Implications in Innate and Adaptive Immune Response Signaling Pathways. Front Genet 2022; 13:857728. [PMID: 35719399 PMCID: PMC9199004 DOI: 10.3389/fgene.2022.857728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/18/2022] [Indexed: 11/29/2022] Open
Abstract
Zika virus (ZIKV) is an arbovirus mainly transmitted by mosquitos of the genus Aedes. The first cases of ZIKV infection in South America occurred in Brazil in 2015. The infection in humans causes diverse symptoms from asymptomatic to a syndrome-like dengue infection with fever, arthralgia, and myalgia. Furthermore, ZIKV infection during pregnancy is associated with fetal microcephaly and neurological disorders. The identification of host molecular mechanisms responsible for the modulation of different signaling pathways in response to ZIKV is the first step to finding potential biomarkers and therapeutic targets and understanding disease outcomes. In the last decade, it has been shown that microRNAs (miRNAs) are important post-transcriptional regulators involved in virtually all cellular processes. miRNAs present in body fluids can not only serve as key biomarkers for diagnostics and prognosis of human disorders but also contribute to cellular signaling offering new insights into pathological mechanisms. Here, we describe for the first time ZIKV-induced changes in miRNA plasma levels in patients during the acute and recovery phases of infection. We observed that during ZIKV acute infection, among the dysregulated miRNAs (DMs), the majority is with decreased levels when compared to convalescent and control patients. We used systems biology tools to build and highlight biological interactions between miRNAs and their multiple direct and indirect target molecules. Among the 24 DMs identified in ZIKV + patients, miR-146, miR-125a-5p, miR-30-5p, and miR-142-3p were related to signaling pathways modulated during infection and immune response. The results presented here are an effort to open new vistas for the key roles of miRNAs during ZIKV infection.
Collapse
Affiliation(s)
- Ana Carolina Carvalho-Silva
- RNA Systems Biology Laboratory (RSBL), Departmento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Programa de Pós-Graduação em Biologia Celular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Almir Ribeiro Da Silva Junior
- Laboratory of Immunology, Heart Institute (InCor) School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology, iii-INCT (National Institute of Science and Technology), São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Waleska Kerllen Martins
- Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- Universidade Anhanguera, São Paulo, Brazil
| | - Verônica Coelho
- Laboratory of Immunology, Heart Institute (InCor) School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology, iii-INCT (National Institute of Science and Technology), São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor) School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology, iii-INCT (National Institute of Science and Technology), São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Simone Gonçalves Fonseca
- Institute for Investigation in Immunology, iii-INCT (National Institute of Science and Technology), São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor) School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology, iii-INCT (National Institute of Science and Technology), São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Ludmila Rodrigues Pinto Ferreira
- National Institute of Science and Technology for Vaccines (INCTV), Belo Horizonte, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Ludmila Rodrigues Pinto Ferreira,
| |
Collapse
|
17
|
Abstract
Influenza viruses cause respiratory tract infections, which lead to human disease outbreaks and pandemics. Influenza A virus (IAV) circulates in diverse animal species, predominantly aquatic birds. This often results in the emergence of novel viral strains causing severe human disease upon zoonotic transmission. Innate immune sensing of the IAV infection promotes host cell death and inflammatory responses to confer antiviral host defense. Dysregulated respiratory epithelial cell death and excessive proinflammatory responses drive immunopathology in highly pathogenic influenza infections. Here, we discuss the critical mechanisms regulating IAV-induced cell death and proinflammatory responses. We further describe the essential role of the Z-form nucleic acid sensor ZBP1/DAI and RIPK3 in triggering apoptosis, necroptosis, and pyroptosis during IAV infection and their impact on host defense and pathogenicity in vivo. We also discuss the functional importance of ZBP1-RIPK3 signaling in recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other viral infections. Understanding these mechanisms of RNA virus-induced cytopathic and pathogenic inflammatory responses is crucial for targeting pathogenic lung infections and human respiratory illness.
Collapse
|
18
|
Winter C, Camarão AAR, Steffen I, Jung K. Network meta-analysis of transcriptome expression changes in different manifestations of dengue virus infection. BMC Genomics 2022; 23:165. [PMID: 35220956 PMCID: PMC8882220 DOI: 10.1186/s12864-022-08390-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/15/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Several studies have been performed to study transcriptome profiles after dengue virus infections with partly different results. Due to slightly different settings of the individual studies, different genes and enriched gene sets are reported in these studies. The main aim of this network meta-analysis was to aggregate a selection of these studies to identify genes and gene sets that are more generally associated with dengue virus infection, i.e. with less dependence on the individual study settings.
Methods
We performed network meta-analysis by different approaches using publicly available gene expression data of five selected studies from the Gene Expression Omnibus database. The study network includes dengue fever (DF), hemorrhagic fever (DHF), shock syndrome (DSS) patients as well as convalescent and healthy control individuals. After data merging and missing value imputation, study-specific batch effects were removed. Pairwise differential expression analysis and subsequent gene-set enrichment analysis were performed between the five study groups. Furthermore, mutual information networks were derived from the top genes of each group comparison, and the separability between the three patient groups was studied by machine learning models.
Results
From the 10 possible pairwise group comparisons in the study network, six genes (IFI27, TPX2, CDT1, DTL, KCTD14 and CDCA3) occur with a noticeable frequency among the top listed genes of each comparison. Thus, there is an increased evidence that these genes play a general role in dengue virus infections. IFI27 and TPX2 have also been highlighted in the context of dengue virus infection by other studies. A few of the identified gene sets from the network meta-analysis overlap with findings from the original studies. Mutual information networks yield additional genes for which the observed pairwise correlation is different between the patient groups. Machine learning analysis shows a moderate separability of samples from the DF, DHF and DSS groups (accuracy about 80%).
Conclusions
Due to an increased sample size, the network meta-analysis could reveal additional genes which are called differentially expressed between the studied groups and that may help to better understand the molecular basis of this disease.
Collapse
|
19
|
Rothan HA, Kumari P, Stone S, Natekar JP, Arora K, Auroni TT, Kumar M. SARS-CoV-2 Infects Primary Neurons from Human ACE2 Expressing Mice and Upregulates Genes Involved in the Inflammatory and Necroptotic Pathways. Pathogens 2022; 11:pathogens11020257. [PMID: 35215199 PMCID: PMC8876293 DOI: 10.3390/pathogens11020257] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/05/2022] [Accepted: 02/15/2022] [Indexed: 02/01/2023] Open
Abstract
Transgenic mice expressing human angiotensin-converting enzyme 2 under the cytokeratin 18 promoter (K18-hACE2) have been extensively used to investigate the pathogenesis and tissue tropism of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. Neuroinvasion and the replication of SARS-CoV-2 within the central nervous system (CNS) of K18-hACE2 mice is associated with increased mortality; although, the mechanisms by which this occurs remain unclear. In this study, we generated primary neuronal cultures from K18-hACE2 mice to investigate the effects of a SARS-CoV-2 infection. We also evaluated the immunological response to SARS-CoV-2 infection in the CNS of K18-hACE2 mice and mouse neuronal cultures. Our data show that neuronal cultures obtained from K18-hACE2 mice are permissive to SARS-CoV-2 infection and support productive virus replication. Furthermore, SARS-CoV-2 infection upregulated the expression of genes involved in innate immunity and inflammation, including IFN-α, ISG-15, CXCL10, CCL2, IL-6 and TNF-α, in the neurons and mouse brains. In addition, we found that SARS-CoV-2 infection of neurons and mouse brains activates the ZBP1/pMLKL-regulated necroptosis pathway. Together, our data provide insights into the neuropathogenesis of SARS-CoV-2 infection in K18-hACE2 mice.
Collapse
|
20
|
Majumdar A, Basu A. Involvement of host microRNAs in flavivirus-induced neuropathology: An update. J Biosci 2022; 47:54. [PMID: 36222134 PMCID: PMC9425815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/17/2022] [Indexed: 09/07/2024]
Abstract
Flaviviruses are a spectrum of vector-borne RNA viruses that cause potentially severe diseases in humans including encephalitis, acute-flaccid paralysis, cognitive disorders and foetal abnormalities. Japanese encephalitis virus (JEV), Zika virus (ZIKV), West Nile virus (WNV) and Dengue virus (DENV) are globally emerging pathogens that lead to epidemics and outbreaks with continued transmission to newer geographical areas over time. In the past decade, studies have focussed on understanding the pathogenic mechanisms of these viruses in a bid to alleviate their disease burden. MicroRNAs (miRNAs) are short single-stranded RNAs that have emerged as master-regulators of cellular gene expression. The dynamics of miRNAs within a cell have the capacity to modulate hundreds of genes and, consequently, their physiological manifestation. Increasing evidence suggests their role in host response to disease and infection including cell survival, intracellular viral replication and immune activation. In this review, we aim to comprehensively update published evidence on the role of miRNAs in host cells infected with the common neurotropic flaviviruses, with an increased focus on neuropathogenic mechanisms. In addition, we briefly cover therapeutic advancements made in the context of miRNA-based antiviral strategies.
Collapse
Affiliation(s)
- Atreye Majumdar
- National Brain Research Centre, Manesar, Gurugram 122 052 India
| | - Anirban Basu
- National Brain Research Centre, Manesar, Gurugram 122 052 India
| |
Collapse
|
21
|
Zingale VD, Gugliandolo A, Mazzon E. MiR-155: An Important Regulator of Neuroinflammation. Int J Mol Sci 2021; 23:90. [PMID: 35008513 PMCID: PMC8745074 DOI: 10.3390/ijms23010090] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression at the post-transcriptional level and that play an important role in many cellular processes, including modulation of inflammation. MiRNAs are present in high concentrations in the central nervous system (CNS) and are spatially and temporally expressed in a specific way. Therefore, an imbalance in the expression pattern of these small molecules can be involved in the development of neurological diseases. Generally, CNS responds to damage or disease through the activation of an inflammatory response, but many neurological disorders are characterized by uncontrolled neuroinflammation. Many studies support the involvement of miRNAs in the activation or inhibition of inflammatory signaling and in the promotion of uncontrolled neuroinflammation with pathological consequences. MiR-155 is a pro-inflammatory mediator of the CNS and plays an important regulatory role. The purpose of this review is to summarize how miR-155 is regulated and the pathological consequences of its deregulation during neuroinflammatory disorders, including multiple sclerosis, Alzheimer's disease and other neuroinflammatory disorders. Modulation of miRNAs' expression could be used as a therapeutic strategy in the treatment of pathological neuroinflammation.
Collapse
Affiliation(s)
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (V.D.Z.); (E.M.)
| | | |
Collapse
|
22
|
Jafarzadeh A, Naseri A, Shojaie L, Nemati M, Jafarzadeh S, Bannazadeh Baghi H, Hamblin MR, Akhlagh SA, Mirzaei H. MicroRNA-155 and antiviral immune responses. Int Immunopharmacol 2021; 101:108188. [PMID: 34626873 DOI: 10.1016/j.intimp.2021.108188] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
The microRNA, miR-155 regulates both adaptive and innate immune responses. In viral infections, miR-155 can affect both innate immunity (interferon response, natural killer cell activity, and macrophage polarization) and adaptive immunity (including generation of anti-viral antibodies, CD8+ cytotoxic T lymphocytes, Th17, Th2, Th1, Tfh and Treg cells). In many viral infections, the proper and timely regulation of miR-155 expression is critical for the induction of an effective anti-virus immune response and viral clearance without any harmful immunopathologic consequences. MiR-155 may also exert pro-viral effects, mainly through the inhibition of the anti-viral interferon response. Thus, dysregulated expression of miR-155 can result in virus persistence and disruption of the normal response to viral infections. This review provides a thorough discussion of the role of miR-155 in immune responses and immunopathologic reactions during viral infections, and highlights its potential as a therapeutic target.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Alma Naseri
- Department of Immunology, Islamic Azadi university of Zahedan, Zahedan, Iran
| | - Layla Shojaie
- Research Center for Liver Diseases, Keck School of Medicine, Department of Medicine, University of Southern California, Los angeles, CA, USA
| | - Maryam Nemati
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
23
|
BAF45b Is Required for Efficient Zika Virus Infection of HAP1 Cells. Viruses 2021; 13:v13102007. [PMID: 34696437 PMCID: PMC8540262 DOI: 10.3390/v13102007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 01/10/2023] Open
Abstract
The 2016 Zika virus (ZIKV) epidemic illustrates the impact of flaviviruses as emerging human pathogens. For unknown reasons, ZIKV replicates more efficiently in neural progenitor cells (NPCs) than in postmitotic neurons. Here, we identified host factors used by ZIKV using the NCI-60 library of cell lines and COMPARE analysis, and cross-analyzed this library with two other libraries of host factors with importance for ZIKV infection. We identified BAF45b, a subunit of the BAF (Brg1/Brm-associated factors) protein complexes that regulate differentiation of NPCs to post-mitotic neurons. ZIKV (and other flaviviruses) infected HAP1 cells deficient in expression of BAF45b and other BAF subunits less efficiently than wildtype (WT) HAP1 cells. We concluded that subunits of the BAF complex are important for infection of ZIKV and other flavivirus. Given their function in cell and tissue differentiation, such regulators may be important determinants of tropism and pathogenesis of arthropod-borne flaviviruses.
Collapse
|
24
|
Polonio CM, Peron JPS. ZIKV Infection and miRNA Network in Pathogenesis and Immune Response. Viruses 2021; 13:v13101992. [PMID: 34696422 PMCID: PMC8541119 DOI: 10.3390/v13101992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 01/01/2023] Open
Abstract
Over the years, viral infections have caused severe illness in humans. Zika Virus (ZIKV) is a flavivirus transmitted by mosquito vectors that leads to notable neurological impairment, whose most dramatic impact is the Congenital ZIKV Syndrome (CZS). ZIKV targets neuronal precursor cells leading to apoptosis and further impairment of neuronal development, causing microcephaly, lissencephaly, ventriculomegaly, and calcifications. Several regulators of biological processes are involved in CZS development, and in this context, microRNAs (miRNAs) seem to have a fundamental role. miRNAs are important regulators of protein translation, as they form the RISC silencing complex and interact with complementary mRNA target sequences to further post-transcriptional repression. In this context, little is known about their participation in the pathogenesis of viral infections. In this review, we discuss how miRNAs could relate to ZIKV and other flavivirus infections.
Collapse
Affiliation(s)
- Carolina Manganeli Polonio
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil;
- Laboratory of Neuroimmunology of Arboviruses, Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-020, Brazil
| | - Jean Pierre Schatzmann Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo, São Paulo 05508-000, Brazil;
- Laboratory of Neuroimmunology of Arboviruses, Scientific Platform Pasteur-USP (SPPU), University of São Paulo, São Paulo 05508-020, Brazil
- Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo, São Paulo 01246-000, Brazil
- Correspondence:
| |
Collapse
|
25
|
Bhagat R, Rajpara P, Kaur G, Gupta K, Seth P. Zika virus E protein dysregulate mir-204/WNT2 signalling in human fetal neural stem cells. Brain Res Bull 2021; 176:93-102. [PMID: 34425198 DOI: 10.1016/j.brainresbull.2021.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022]
Abstract
Zika Virus (ZIKV) belongs to the family of flaviviruses, and is neurotrophic. It has been known to cause severe congenital disabilities including microcephaly in neonates. The virus has a specific preference towards neural stem cells (NSCs). ZIKV impairs proliferation and differentiation of NSCs during in-utero brain development of the fetus. However, molecular pathways involved in ZIKV induced alteration in NSCs are yet to be explored. In our previous study, we have described that ZIKV E protein dysregulates microRNA circuitry in NSCs and also impairs their proliferative and differentiation abilities. WNT signalling was found to be the target of differentially expressed miRNAs as suggested by PANTHER PATHWAY analysis of differentially expressed miRNA targets. In our current follow-up study, we investigate that WNT2 is downregulated in response to ZIKV E protein in human fetal NSCs and WNT2 is the molecular target of microRNA miR-204-5p. We provide pieces of evidences that miR-204-5p/WNT2 axis is involved in ZIKV induced impairment in the proliferation and immature differentiation of neural stem cells.
Collapse
Affiliation(s)
- Reshma Bhagat
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Prateek Rajpara
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Guneet Kaur
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Karnika Gupta
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| | - Pankaj Seth
- Department of Cellular and Molecular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Gurgaon, Haryana, India.
| |
Collapse
|
26
|
Zhang L, Jiao C, Liu L, Wang A, Tang L, Ren Y, Huang P, Xu J, Mao D, Liu L. NLRC5: A Potential Target for Central Nervous System Disorders. Front Immunol 2021; 12:704989. [PMID: 34220868 PMCID: PMC8250149 DOI: 10.3389/fimmu.2021.704989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/07/2021] [Indexed: 12/22/2022] Open
Abstract
Nucleotide oligomerization domain-like receptors (NLRs), a class of pattern recognition receptors, participate in the host’s first line of defense against invading pathogenic microorganisms. NLR family caspase recruitment domain containing 5 (NLRC5) is the largest member of the NLR family and has been shown to play an important role in inflammatory processes, angiogenesis, immunity, and apoptosis by regulating the nuclear factor-κB, type I interferon, and inflammasome signaling pathways, as well as the expression of major histocompatibility complex I genes. Recent studies have found that NLRC5 is also associated with neuronal development and central nervous system (CNS) diseases, such as CNS infection, cerebral ischemia/reperfusion injury, glioma, multiple sclerosis, and epilepsy. This review summarizes the research progress in the structure, expression, and biological characteristics of NLRC5 and its relationship with the CNS.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cui Jiao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Aiping Wang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Tang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Xu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dingan Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
Alpuche-Lazcano SP, Saliba J, Costa VV, Campolina-Silva GH, Marim FM, Ribeiro LS, Blank V, Mouland AJ, Teixeira MM, Gatignol A. Profound downregulation of neural transcription factor Npas4 and Nr4a family in fetal mice neurons infected with Zika virus. PLoS Negl Trop Dis 2021; 15:e0009425. [PMID: 34048439 PMCID: PMC8191876 DOI: 10.1371/journal.pntd.0009425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 06/10/2021] [Accepted: 04/30/2021] [Indexed: 01/01/2023] Open
Abstract
Zika virus (ZIKV) infection of neurons leads to neurological complications and congenital malformations of the brain of neonates. To date, ZIKV mechanism of infection and pathogenesis is not entirely understood and different studies on gene regulation of ZIKV-infected cells have identified a dysregulation of inflammatory and stem cell maintenance pathways. MicroRNAs (miRNAs) are post-transcriptional regulators of cellular genes and they contribute to cell development in normal function and disease. Previous reports with integrative analyses of messenger RNAs (mRNAs) and miRNAs during ZIKV infection have not identified neurological pathway defects. We hypothesized that dysregulation of pathways involved in neurological functions will be identified by RNA profiling of ZIKV-infected fetal neurons. We therefore used microarrays to analyze gene expression levels following ZIKV infection of fetal murine neurons. We observed that the expression levels of transcription factors such as neural PAS domain protein 4 (Npas4) and of three members of the orphan nuclear receptor 4 (Nr4a) were severely decreased after viral infection. We confirmed that their downregulation was at both the mRNA level and at the protein level. The dysregulation of these transcription factors has been previously linked to aberrant neural functions and development. We next examined the miRNA expression profile in infected primary murine neurons by microarray and found that various miRNAs were dysregulated upon ZIKV infection. An integrative analysis of the differentially expressed miRNAs and mRNAs indicated that miR-7013-5p targets Nr4a3 gene. Using miRmimics, we corroborated that miR-7013-5p downregulates Nr4a3 mRNA and protein levels. Our data identify a profound dysregulation of neural transcription factors with an overexpression of miR-7013-5p that results in decreased Nr4a3 expression, likely a main contributor to ZIKV-induced neuronal dysfunction. Zika virus (ZIKV) is an emerging virus transmitted horizontally between humans through mosquito bites, and sexual intercourse generally inducing a mild disease. ZIKV is also transmitted vertically from mother-to-child producing congenital ZIKV syndrome (CZVS) in neonates. CZVS leads to severe microcephaly associated with neurological, ocular, musculoskeletal, genitourinary disorders and other disabilities. Although numerous studies have been performed on ZIKV infection of brain cells, we are still far from understanding how ZIKV infection leads to dysregulation of host genes, virus-induced cytopathicity and consequent pathology. Micro (mi)RNAs are small noncoding RNAs encoded and processed by the host cell. They regulate gene expression at the post-transcriptional level in a process called RNA interference (RNAi). Here, we evaluated the relationship between ZIKV infection and the level of mRNAs and miRNAs expressed in the cell. ZIKV infection of mouse embryo neurons downregulated several neural immediate-early genes (IEG). Moreover, we revealed that ZIKV infection led to aberrant regulation of several miRNAs, and identified one whose cognate target was a neural IEG. Our work identifies novel genes and miRNAs that are modulated upon ZIKV infection of fetal murine neurons, therefore linking neuronal dysfunction to transcription and the RNA interference pathway.
Collapse
Affiliation(s)
- Sergio P. Alpuche-Lazcano
- Virus-Cell Interactions Laboratory, Lady Davis Institute for Medical Research, Montréal, Canada
- RNA Trafficking Laboratory, Lady Davis Institute for Medical Research, Montréal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada
| | - James Saliba
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Canada
- Lady Davis Institute for Medical Research, Montréal, Canada
| | - Vivian V. Costa
- Departamento de Bioquimica e Imunologia do Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departamento de Morfologia do Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gabriel H. Campolina-Silva
- Departamento de Bioquimica e Imunologia do Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda M. Marim
- Departamento de Bioquimica e Imunologia do Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas S. Ribeiro
- Departamento de Bioquimica e Imunologia do Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Volker Blank
- Lady Davis Institute for Medical Research, Montréal, Canada
- Department of Medicine, Montréal, Canada
- Department of Physiology, McGill University, Montréal, Canada
| | - Andrew J. Mouland
- RNA Trafficking Laboratory, Lady Davis Institute for Medical Research, Montréal, Canada
- Department of Medicine, Montréal, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
| | - Mauro M. Teixeira
- Departamento de Bioquimica e Imunologia do Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Anne Gatignol
- Virus-Cell Interactions Laboratory, Lady Davis Institute for Medical Research, Montréal, Canada
- Department of Medicine, Montréal, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
- * E-mail:
| |
Collapse
|
28
|
Zika Virus Pathogenesis: A Battle for Immune Evasion. Vaccines (Basel) 2021; 9:vaccines9030294. [PMID: 33810028 PMCID: PMC8005041 DOI: 10.3390/vaccines9030294] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infection and its associated congenital and other neurological disorders, particularly microcephaly and other fetal developmental abnormalities, constitute a World Health Organization (WHO) Zika Virus Research Agenda within the WHO’s R&D Blueprint for Action to Prevent Epidemics, and continue to be a Public Health Emergency of International Concern (PHEIC) today. ZIKV pathogenicity is initiated by viral infection and propagation across multiple placental and fetal tissue barriers, and is critically strengthened by subverting host immunity. ZIKV immune evasion involves viral non-structural proteins, genomic and non-coding RNA and microRNA (miRNA) to modulate interferon (IFN) signaling and production, interfering with intracellular signal pathways and autophagy, and promoting cellular environment changes together with secretion of cellular components to escape innate and adaptive immunity and further infect privileged immune organs/tissues such as the placenta and eyes. This review includes a description of recent advances in the understanding of the mechanisms underlying ZIKV immune modulation and evasion that strongly condition viral pathogenesis, which would certainly contribute to the development of anti-ZIKV strategies, drugs, and vaccines.
Collapse
|
29
|
Tabari D, Scholl C, Steffens M, Weickhardt S, Elgner F, Bender D, Herrlein ML, Sabino C, Semkova V, Peitz M, Till A, Brüstle O, Hildt E, Stingl J. Impact of Zika Virus Infection on Human Neural Stem Cell MicroRNA Signatures. Viruses 2020; 12:E1219. [PMID: 33121145 PMCID: PMC7693339 DOI: 10.3390/v12111219] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus, which can cause brain abnormalities in newborns, including microcephaly. MicroRNAs (miRNAs) are small non-coding RNAs, which post- transcriptionally regulate gene expression. They are involved in various processes including neurological development and host responses to viral infection, but their potential role in ZIKV pathogenesis remains poorly understood. MiRNAs can be incorporated into extracellular vesicles (EVs) and mediate cell-to-cell communication. While it is well known that in viral infections EVs carrying miRNAs can play a crucial role in disease pathogenesis, ZIKV effects on EV-delivered miRNAs and their contribution to ZIKV pathogenesis have not been elucidated. In the present study, we profiled intracellular and EV-derived miRNAs by next generation sequencing and analyzed the host mRNA transcriptome of neural stem cells during infection with ZIKV Uganda and French Polynesia strains. We identified numerous miRNAs, including miR-4792, which were dysregulated at the intracellular level and had altered levels in EVs during ZIKV infection. Integrated analyses of differentially expressed genes and miRNAs showed that ZIKV infection had an impact on processes associated with neurodevelopment and oxidative stress. Our results provide insights into the roles of intracellular and EV-associated host miRNAs in ZIKV pathogenesis.
Collapse
Affiliation(s)
- Denna Tabari
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Catharina Scholl
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Michael Steffens
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Sandra Weickhardt
- Research Division, Federal Institute for Drugs and Medical Devices, 53175 Bonn, Germany; (D.T.); (M.S.); (S.W.)
| | - Fabian Elgner
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Daniela Bender
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Marie-Luise Herrlein
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Catarina Sabino
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Vesselina Semkova
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
- Cell Programming Core Facility, Medical Faculty, University of Bonn, 53172 Bonn, Germany
| | - Andreas Till
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany; (V.S.); (M.P.); (A.T.); (O.B.)
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany; (F.E.); (D.B.); (M.-L.H.); (C.S.); (E.H.)
| | - Julia Stingl
- Department of Clinical Pharmacology, University Hospital, RWTH Aachen University, 52074 Aachen, Germany;
| |
Collapse
|
30
|
The FDA-approved gold drug auranofin inhibits novel coronavirus (SARS-COV-2) replication and attenuates inflammation in human cells. Virology 2020; 547:7-11. [PMID: 32442105 PMCID: PMC7236683 DOI: 10.1016/j.virol.2020.05.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 11/25/2022]
Abstract
SARS-COV-2 has recently emerged as a new public health threat. Herein, we report that the FDA-approved drug, auranofin, inhibits SARS-COV-2 replication in human cells at low micro molar concentration. Treatment of cells with auranofin resulted in a 95% reduction in the viral RNA at 48 h after infection. Auranofin treatment dramatically reduced the expression of SARS-COV-2-induced cytokines in human cells. These data indicate that auranofin could be a useful drug to limit SARS-CoV-2 infection and associated lung injury due to its antiviral, anti-inflammatory and anti-reactive oxygen species (ROS) properties. Further animal studies are warranted to evaluate the safety and efficacy of auranofin for the management of SARS-COV-2 associated disease. Auranofin inhibits replication of SARS-COV-2 in human cells at low micro molar concentration. Auranofin treatment resulted in significant reduction in SARS-COV-2-induced cytokines in human cells. Auranofin could mitigate SARS-COV-2 infection and lung damage due to its anti-viral and anti-inflammatory properties. Auranofin is a gold-containing FDA-approved drug.
Collapse
|
31
|
Abstract
Previous work showed that the thymus can be infected by RNA viruses as HIV and HTLV-1. We thus hypothesized that the thymus might also be infected by the Zika virus (ZIKV). Herein we provide compelling evidence that ZIKV targets human thymic epithelial cells (TEC) in vivo and in vitro. ZIKV-infection enhances keratinization of TEC, with a decrease in proliferation and increase in cell death. Moreover, ZIKV modulates a high amount of coding RNAs with upregulation of genes related to cell adhesion and migration, as well as non-coding genes including miRNAs, circRNAs and lncRNAs. Moreover, we observed enhanced attachment of lymphoblastic T-cells to infected TEC, as well as virus transfer to those cells. Lastly, alterations in thymuses from babies congenitally infected were seen, with the presence of viral envelope protein in TEC. Taken together, our data reveals that the thymus, particularly the thymic epithelium, is a target for the ZIKV with changes in the expression of molecules that are relevant for interactions with developing thymocytes.
Collapse
|
32
|
Cellular microRNA-155 Regulates Virus-Induced Inflammatory Response and Protects against Lethal West Nile Virus Infection. Viruses 2019; 12:v12010009. [PMID: 31861621 PMCID: PMC7019255 DOI: 10.3390/v12010009] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022] Open
Abstract
West Nile virus (WNV) is a flavivirus that has disseminated globally as a significant cause of viral encephalitis in humans. MircoRNA-155 (miR-155) regulates various aspects of innate and adaptive immune responses. We previously reported that WNV infection induces upregulation of miR-155 in mice brains. In the current study, we demonstrate the critical role of miR-155 in restricting the pathogenesis of WNV infection in mice. Compared to wild-type (WT) mice, miR-155 knockout mice exhibited significantly higher morbidity and mortality after infection with either a lethal strain, WNV NY99, or a non-lethal strain, WNV Eg101. Increased mortality in miR-155−/− mice was associated with significantly high WNV burden in the serum and brains. Protein levels of interferon (IFN)-α in the serum and brains were higher in miR-155−/− mice. However, miR-155−/− mice exhibited significantly lower protein levels of anti-viral interleukin (IL)-1β, IL-12, IL-6, IL-15, and GM-CSF despite the high viral load. Primary mouse cells lacking miR-155 were more susceptible to infection with WNV compared to cells derived from WT mice. Besides, overexpression of miR-155 in human neuronal cells modulated anti-viral cytokine response and resulted in significantly lower WNV replication. These data collectively indicate that miR-155 restricts WNV production in mouse and human cells and protects against lethal WNV infection in mice.
Collapse
|
33
|
Rothan HA, Arora K, Natekar JP, Strate PG, Brinton MA, Kumar M. Z-DNA-Binding Protein 1 Is Critical for Controlling Virus Replication and Survival in West Nile Virus Encephalitis. Front Microbiol 2019; 10:2089. [PMID: 31572318 PMCID: PMC6749019 DOI: 10.3389/fmicb.2019.02089] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/26/2019] [Indexed: 01/31/2023] Open
Abstract
West Nile virus (WNV), a neurotropic flavivirus, is the leading cause of viral encephalitis in the United States. Recently, Zika virus (ZIKV) infections have caused serious neurological diseases and birth defects, specifically Guillain-Barrè syndrome and microcephaly. Z-DNA binding protein 1 (ZBP1) is a cytoplasmic sensor that that has been shown to play a significant role in initiating a robust immune response. We previously reported that WNV and ZIKV infections induce dramatic up-regulation of ZBP1 in mouse brains as well as in infected primary mouse cells. Herein, we show the critical role of ZBP1 in restricting the pathogenesis of WNV and ZIKV infections. Deletion of ZBP1 resulted in significantly higher morbidity and mortality after infection with a pathogenic WNV NY99 strain in mice. No mortality was observed in wild-type (WT) mice infected with the non-pathogenic WNV strain, Eg101. Interestingly, infection of ZBP1-/- mice with WNV Eg101 was lethal resulting in 100% mortality, suggesting that ZBP1 is required for survival after WNV infection. Viremia and brain viral load were significantly higher in ZBP1-/- mice compared to WT mice. In addition, protein levels of interferon (IFN)-α, and inflammatory cytokines and chemokines were significantly higher in the serum and brains of infected ZBP1-/- mice compared to the WT mice. Primary mouse cortical neurons and mouse embryonic fibroblasts (MEFs) derived from ZBP1-/- mice produced higher virus titers compared to WT cells after infection with WNV NY99 and WNV Eg101. Similarly, neurons and MEFs lacking ZBP1 exhibited significantly enhanced replication of PRVABC59 (Asian) and MR766 (African) ZIKV compared to WT cells. The knockout of ZBP1 function in MEFs inhibited ZBP1-dependent virus-induced cell death. In conclusion, these data reveal that ZBP1 restricts WNV and ZIKV production in mouse cells and is required for survival of a peripheral WNV infection in mice.
Collapse
Affiliation(s)
- Hussin A Rothan
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| | - Komal Arora
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| | - Janhavi P Natekar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| | - Philip G Strate
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| | - Margo A Brinton
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
34
|
New Advances on Zika Virus Research. Viruses 2019; 11:v11030258. [PMID: 30875715 PMCID: PMC6466272 DOI: 10.3390/v11030258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
|
35
|
Castro FL, Geddes VEV, Monteiro FLL, Gonçalves RMDT, Campanati L, Pezzuto P, Paquin-Proulx D, Schamber-Reis BL, Azevedo GS, Gonçalves AL, Cunha DP, Moreira MEL, Vasconcelos ZFM, Chimeli L, Melo A, Tanuri A, Nixon DF, Ribeiro-Alves M, Aguiar RS. MicroRNAs 145 and 148a Are Upregulated During Congenital Zika Virus Infection. ASN Neuro 2019; 11:1759091419850983. [PMID: 31213064 PMCID: PMC6585135 DOI: 10.1177/1759091419850983] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/04/2019] [Accepted: 04/13/2019] [Indexed: 12/24/2022] Open
Abstract
Zika virus (ZIKV) is an arthropod-borne virus (arbovirus) member of the Flaviviridae family, which has been associated with the development of the congenital Zika syndrome (CZS). RNA viruses, such as flaviviruses, have been reported to exert a profound impact on host microRNAs (miRNAs). Cellular miRNAs modulated by ZIKV may help identify cellular pathways of relevance to pathogenesis. Here, we screened 754 human cellular miRNAs modulated by ZIKV infection (Brazilian PE strain) in a neuroblastoma cell line. Seven miRNAs (miR-99a*, miR-126*, miR-190b, miR-361-3p, miR-522-3p, miR-299-5p, and miR-1267) were downregulated during ZIKV infection, while miR-145 was upregulated. Furthermore, 11 miRNAs were exclusively expressed in ZIKV-infected (miR-148a, miR-342-5p, miR-598, and miR-708-3p) or mock cells (miR-208, miR-329, miR-432-5p, miR-488, miR-518b, miR-520g, and miR-767-5p). Furthermore, in silico analysis indicated that some central nervous system, cellular migration, and adhesion function-related biological processes were overrepresented in the list of target genes of the miRNAs regulated in ZIKV-infected cells, especially for miR-145 and miR-148a. The induction of miR-145 and miR-148a was confirmed in postmortem brain samples from stillborn with severe CZS. Finally, we determined the expression regulation of microcephaly related genes through RNA interference pathway caused by ZIKV directly on neuron cells.
Collapse
Affiliation(s)
- Fernanda L. Castro
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor E. V. Geddes
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio L. L. Monteiro
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raphael M. D. T. Gonçalves
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Loraine Campanati
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Pezzuto
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dominic Paquin-Proulx
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Bruno L. Schamber-Reis
- Faculdade de Ciências Médicas de Campina Grande, Núcleo de Genética Médica, Centro Universitário UniFacisa, Campina Grande, Brazil
| | | | | | | | | | | | - Leila Chimeli
- Laboratório de Neuropatologia, Instituto Estadual do Cérebro, Rio de Janeiro, Brazil
| | - Adriana Melo
- Faculdade de Ciências Médicas de Campina Grande, Núcleo de Genética Médica, Centro Universitário UniFacisa, Campina Grande, Brazil
- Instituto de Pesquisa Professor Amorim Neto, Campina Grande, Brazil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas F. Nixon
- Division of Infectious Diseases, Weill Cornell Medicine, New York City, NY, USA
| | | | - Renato S. Aguiar
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|