1
|
Gomes-de-Pontes L, Barreiros LA, Gomes LN, Salgado RC, da Silva Napoleão SM, Soeiro-Pereira PV, Passos SD, Condino-Neto A. Congenital Zika Syndrome: Insights from Integrated Proteomic and Metabolomic Analysis. Biomolecules 2024; 15:32. [PMID: 39858427 PMCID: PMC11762526 DOI: 10.3390/biom15010032] [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: 08/07/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 01/27/2025] Open
Abstract
Background: In this study, we investigated the role of extracellular vesicles (EVs) in the pathogenesis of Congenital Zika Syndrome (CZS). Previous studies have highlighted the role of EVs in intercellular communication and the modulation of biological processes during viral infections, motivating our in-depth analysis. Our objective was to identify specific molecular signatures in the EVs of patients with CZS, focusing on their potential as biomarkers and on cellular pathways affected by the infection. Methods: We conducted advanced proteomic and metabolomic analyses using mass spectrometry for protein and metabolite identification. EVs were isolated from CZS patient samples and control groups using Izon qEV size-exclusion chromatography columns. Results: The analyzed EVs presented distinct molecular profiles in patients with CZS. Proteomic analysis revealed significant alterations in specific proteins, suggesting involvement in the PI3K-AKT-mTOR pathway, while metabolomics highlighted metabolites related to critical processes in Zika virus pathogenesis. These findings suggest a key role for the PI3K-AKT-mTOR pathway in regulating cellular processes during infection and indicate the involvement of EVs in intercellular communication. Additionally, the results identified potential biomarkers capable of aiding early diagnosis and assessing disease progression. Conclusions: This study demonstrates that EVs play a crucial role in intercellular communication during Zika virus infection. The identification of specific alterations in the PI3K-AKT-mTOR pathway highlights a possible therapeutic target, providing new opportunities for the development of more effective treatment strategies for CZS. Our findings significantly advance the understanding of CZS and underscore the need for further investigations using advanced techniques to validate and explore these potential molecular targets.
Collapse
Affiliation(s)
- Leticia Gomes-de-Pontes
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Lucila Akune Barreiros
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Lillian Nunes Gomes
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Ranieri Coelho Salgado
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | - Sarah Maria da Silva Napoleão
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| | | | - Saulo Duarte Passos
- Infectious Pediatric Laboratory, Medicine School of Jundiaí, Jundiaí 13202-550, SP, Brazil;
| | - Antonio Condino-Neto
- Department of Immunology (LIH), Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 2415-Butantã, São Paulo 05508-000, SP, Brazil; (L.A.B.); (L.N.G.); (R.C.S.); (S.M.d.S.N.)
| |
Collapse
|
2
|
Garcia G, Chakravarty N, Paiola S, Urena E, Gyani P, Tse C, French SW, Danielpour M, Breunig JJ, Nathanson DA, Arumugaswami V. Differential Susceptibility of Ex Vivo Primary Glioblastoma Tumors to Oncolytic Effect of Modified Zika Virus. Cells 2023; 12:2384. [PMID: 37830597 PMCID: PMC10572118 DOI: 10.3390/cells12192384] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023] Open
Abstract
Glioblastoma (GBM), the most common primary malignant brain tumor, is a highly lethal form of cancer with a very limited set of treatment options. High heterogeneity in the tumor cell population and the invasive nature of these cells decrease the likely efficacy of traditional cancer treatments, thus requiring research into novel treatment options. The use of oncolytic viruses as potential therapeutics has been researched for some time. Zika virus (ZIKV) has demonstrated oncotropism and oncolytic effects on GBM stem cells (GSCs). To address the need for safe and effective GBM treatments, we designed an attenuated ZIKV strain (ZOL-1) that does not cause paralytic or neurological diseases in mouse models compared with unmodified ZIKV. Importantly, we found that patient-derived GBM tumors exhibited susceptibility (responders) and non-susceptibility (non-responders) to ZOL-1-mediated tumor cell killing, as evidenced by differential apoptotic cell death and cell viability upon ZOL-1 treatment. The oncolytic effect observed in responder cells was seen both in vitro in neurosphere models and in vivo upon xenograft. Finally, we observed that the use of ZOL-1 as combination therapy with multiple PI3K-AKT inhibitors in non-responder GBM resulted in enhanced chemotherapeutic efficacy. Altogether, this study establishes ZOL-1 as a safe and effective treatment against GBM and provides a foundation to conduct further studies evaluating its potential as an effective adjuvant with other chemotherapies and kinase inhibitors.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Nikhil Chakravarty
- Department of Epidemiology, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Sophia Paiola
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Estrella Urena
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Priya Gyani
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Christopher Tse
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Samuel W. French
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Moise Danielpour
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.D.); (J.J.B.)
| | - Joshua J. Breunig
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.D.); (J.J.B.)
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - David A. Nathanson
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Bacon A, Teixeira M, Costa V, Bone P, Simmons J, Drew J. Generation of a thermostable, oral Zika vaccine that protects against virus challenge in non-human primates. Vaccine 2023; 41:2524-2533. [PMID: 36894395 DOI: 10.1016/j.vaccine.2023.02.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/03/2023] [Accepted: 02/17/2023] [Indexed: 03/09/2023]
Abstract
Here we report the development of a thermally stable, orally administered, candidate Zika vaccine using human serotype 5 adenovirus (AdHu5). We engineered AdHu5 to express the genes for the envelope and NS1 proteins of Zika virus. AdHu5 was formulated using a proprietary platform, OraPro, comprising a mix of sugars and modified amino acids that can overcome elevated temperatures (37 C), and an enteric coated capsule that protects the integrity of the AdHu5 from the acid in the stomach. This enables the delivery AdHu5 to the immune system of the small intestine. We show that oral delivery of AdHu5 elicited antigen-specific serum IgG immune responses in a mouse model and in a non-human primate model. Importantly, these immune responses were able reduce viral counts in mice and to prevent detectable viraemia in the non-human primates on challenge with live Zika virus. This candidate vaccine has significant advantages over many current vaccines that are maintained in a cold or ultra-cold chain and require parenteral administration.
Collapse
Affiliation(s)
- Andrew Bacon
- iosBio Ltd, Sovereign Business Park, Albert Dr, Burgess Hill RH15 9TY, United Kingdom
| | - Mauro Teixeira
- Centro de Pesquisa e Desenvolvimento de Fármacos (CPDF), Laboratórios Temáticos - Bloco G3, Instituto de Ciências Biológicas - UFMG, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, Brazil
| | - Vivian Costa
- Centro de Pesquisa e Desenvolvimento de Fármacos (CPDF), Laboratórios Temáticos - Bloco G3, Instituto de Ciências Biológicas - UFMG, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, Brazil
| | - Peter Bone
- iosBio Ltd, Sovereign Business Park, Albert Dr, Burgess Hill RH15 9TY, United Kingdom
| | - Jennifer Simmons
- iosBio Ltd, Sovereign Business Park, Albert Dr, Burgess Hill RH15 9TY, United Kingdom
| | - Jeffrey Drew
- iosBio Ltd, Sovereign Business Park, Albert Dr, Burgess Hill RH15 9TY, United Kingdom.
| |
Collapse
|
4
|
Quincozes-Santos A, Bobermin LD, Costa NLF, Thomaz NK, Almeida RRDS, Beys-da-Silva WO, Santi L, Rosa RL, Capra D, Coelho-Aguiar JM, DosSantos MF, Heringer M, Cirne-Lima EO, Guimarães JA, Schuler-Faccini L, Gonçalves CA, Moura-Neto V, Souza DO. The role of glial cells in Zika virus-induced neurodegeneration. Glia 2023. [PMID: 36866453 DOI: 10.1002/glia.24353] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Zika virus (ZIKV) is a strongly neurotropic flavivirus whose infection has been associated with microcephaly in neonates. However, clinical and experimental evidence indicate that ZIKV also affects the adult nervous system. In this regard, in vitro and in vivo studies have shown the ability of ZIKV to infect glial cells. In the central nervous system (CNS), glial cells are represented by astrocytes, microglia, and oligodendrocytes. In contrast, the peripheral nervous system (PNS) constitutes a highly heterogeneous group of cells (Schwann cells, satellite glial cells, and enteric glial cells) spread through the body. These cells are critical in both physiological and pathological conditions; as such, ZIKV-induced glial dysfunctions can be associated with the development and progression of neurological complications, including those related to the adult and aging brain. This review will address the effects of ZIKV infection on CNS and PNS glial cells, focusing on cellular and molecular mechanisms, including changes in the inflammatory response, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate homeostasis, neural metabolism, and neuron-glia communication. Of note, preventive and therapeutic strategies that focus on glial cells may emerge to delay and/or prevent the development of ZIKV-induced neurodegeneration and its consequences.
Collapse
Affiliation(s)
- André Quincozes-Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Naithan Ludian Fernandes Costa
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Natalie K Thomaz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rômulo Rodrigo de Souza Almeida
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Lucélia Santi
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Rafael L Rosa
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Daniela Capra
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana M Coelho-Aguiar
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcos Fabio DosSantos
- Laboratório de Propriedades Mecânicas e Biologia Celular, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Manoela Heringer
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
5
|
Tatara JM, Rosa RL, Varela APM, Teixeira TF, Sesterheim P, Gris A, Driemeier D, Moraes ANS, Berger M, Peña RD, Roehe PM, Souza DOG, Guimarães JA, Campos AR, Santi L, Beys-da-Silva WO. Differential proteomics of Zika virus (ZIKV) infection reveals molecular changes potentially involved in immune system evasion by a Brazilian strain of ZIKV. Arch Virol 2023; 168:70. [PMID: 36658439 DOI: 10.1007/s00705-022-05629-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/01/2022] [Indexed: 01/21/2023]
Abstract
Zika virus (ZIKV) is an arbovirus that was responsible for multiple outbreaks from 2007 to 2015. It has been linked to cases of microcephaly in Brazil in 2015, among other neurological disorders. Differences among strains might be the reason for different clinical outcomes of infection. To evaluate this hypothesis, we performed a comparative proteomic analysis of Vero cells infected with the African strain MR766 (ZIKVAFR) and the Brazilian strain 17 SM (ZIKVBR). A total of 550 proteins were identified as differentially expressed in ZIKVAFR- or ZIKVBR-infected cells compared to the control. The main findings included upregulation of immune system pathways (neutrophil degranulation and adaptive/innate immune system) and potential activation of immune-system-related pathways by ZIKVAFR (mTOR, JAK-STAT, NF-κB, and others) compared with the ZIKVBR/control. In addition, phagocytosis by macrophages and engulfment of leukocytes were activated in ZIKVAFR infection. An in vivo analysis using an immunocompetent C57BL/6N mouse model identified interstitial pneumonia with neutrophil infiltration in the lungs only in mice infected with ZIKVBR at 48 hours postinfection, with a significant amount of virus detected. Likewise, only animals infected with ZIKVBR had viral material in the cytoplasm of lung macrophages. These results suggest that activation of the immune system by ZIKVAFR infection may lead to faster viral clearance by immune cells.
Collapse
Affiliation(s)
- Juliana M Tatara
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil
| | - Rafael L Rosa
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil
| | - Ana Paula M Varela
- Department of Microbiology, Immunology and Parasitology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Tais F Teixeira
- Center for Experimental Cardiology, Institute of Cardiology, Porto Alegre, Brazil
| | - Patrícia Sesterheim
- Center for Experimental Cardiology, Institute of Cardiology, Porto Alegre, Brazil
| | - Anderson Gris
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - David Driemeier
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Amanda N S Moraes
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil
| | - Markus Berger
- Experimental Research Center, Clinical Hospital of Porto Alegre, Porto Alegre, Brazil
| | - Ramon D Peña
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Paulo M Roehe
- Department of Microbiology, Immunology and Parasitology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Diogo O G Souza
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jorge A Guimarães
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil.,Center for Experimental Cardiology, Institute of Cardiology, Porto Alegre, Brazil
| | | | - Lucélia Santi
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil.,Experimental Research Center, Clinical Hospital of Porto Alegre, Porto Alegre, Brazil.,Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Walter O Beys-da-Silva
- Post-Graduation Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul, Av Ipiranga, 2752 suit 709, Porto Alegre, RS, Brazil. .,Experimental Research Center, Clinical Hospital of Porto Alegre, Porto Alegre, Brazil. .,Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
6
|
Contreras D, Garcia G, Jones MK, Martinez LE, Jayakarunakaran A, Gangalapudi V, Tang J, Wu Y, Zhao JJ, Chen Z, Ramaiah A, Tsui I, Kumar A, Nielsen-Saines K, Wang S, Arumugaswami V. Differential Susceptibility of Fetal Retinal Pigment Epithelial Cells, hiPSC- Retinal Stem Cells, and Retinal Organoids to Zika Virus Infection. Viruses 2023; 15:142. [PMID: 36680182 PMCID: PMC9864143 DOI: 10.3390/v15010142] [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: 11/15/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Zika virus (ZIKV) causes microcephaly and congenital eye disease. The cellular and molecular basis of congenital ZIKV infection are not well understood. Here, we utilized a biologically relevant cell-based system of human fetal retinal pigment epithelial cells (FRPEs), hiPSC-derived retinal stem cells (iRSCs), and retinal organoids to investigate ZIKV-mediated ocular cell injury processes. Our data show that FRPEs were highly susceptible to ZIKV infection exhibiting increased apoptosis, whereas iRSCs showed reduced susceptibility. Detailed transcriptomics and proteomics analyses of infected FRPEs were performed. Nucleoside analogue drug treatment inhibited ZIKV replication. Retinal organoids were susceptible to ZIKV infection. The Asian genotype ZIKV exhibited higher infectivity, induced profound inflammatory response, and dysregulated transcription factors involved in retinal organoid differentiation. Collectively, our study shows that ZIKV affects ocular cells at different developmental stages resulting in cellular injury and death, further providing molecular insight into the pathogenesis of congenital eye disease.
Collapse
Affiliation(s)
- Deisy Contreras
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Melissa Kaye Jones
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Laura E. Martinez
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Akshaya Jayakarunakaran
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | | | - Jie Tang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Ying Wu
- Alpine BioTherapeutics Corporation, 11107 Roselle Street, Suite 210, San Diego, CA 92121, USA
| | - Jiagang J. Zhao
- Alpine BioTherapeutics Corporation, 11107 Roselle Street, Suite 210, San Diego, CA 92121, USA
| | - Zhaohui Chen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Arunachalam Ramaiah
- Tata Institute for Genetics and Society, Center at inStem, Bangalore 560065, India
| | - Irena Tsui
- Retina Division, Department of Ophthalmology, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI 48201, USA
| | | | - Shaomei Wang
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vaithilingaraja Arumugaswami
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Shofa M, Okamura T, Urano E, Matsuura Y, Yasutomi Y, Saito A. Repeated Intravaginal Inoculation of Zika Virus Protects Cynomolgus Monkeys from Subcutaneous Superchallenge. Int J Mol Sci 2022; 23:ijms232214002. [PMID: 36430481 PMCID: PMC9696507 DOI: 10.3390/ijms232214002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV) outbreaks in Central and South America caused severe public health problems in 2015 and 2016. These outbreaks were finally contained through several methods, including mosquito control using insecticides and repellents. Additionally, the development of herd immunity in these countries might have contributed to containing the epidemic. While ZIKV is mainly transmitted by mosquito bites and mucosal transmission via bodily fluids, including the semen of infected individuals, has also been reported. We evaluated the effect of mucosal ZIKV infection on continuous subcutaneous challenges in a cynomolgus monkey model. Repeated intravaginal inoculations of ZIKV did not induce detectable viremia or clinical symptoms, and all animals developed a potent neutralizing antibody, protecting animals from the subsequent subcutaneous superchallenge. These results suggest that viral replication at mucosal sites can induce protective immunity without causing systemic viremia or symptoms.
Collapse
Affiliation(s)
- Maya Shofa
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Tomotaka Okamura
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 305-0843, Japan
| | - Emiko Urano
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 305-0843, Japan
| | - Yoshiharu Matsuura
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 305-0843, Japan
- Department of Molecular and Experimental Medicine, Mie University Graduate School of Medicine, Mie 514-8507, Japan
- Correspondence: (Y.Y.); (A.S.)
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Center for Animal Disease Control, University of Miyazaki, Miyazaki 889-2192, Japan
- Correspondence: (Y.Y.); (A.S.)
| |
Collapse
|
8
|
Garcia G, Chakravarty N, Abu AE, Jeyachandran AV, Takano KA, Brown R, Morizono K, Arumugaswami V. Replication-Deficient Zika Vector-Based Vaccine Provides Maternal and Fetal Protection in Mouse Model. Microbiol Spectr 2022; 10:e0113722. [PMID: 36169338 PMCID: PMC9602260 DOI: 10.1128/spectrum.01137-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/28/2022] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne human pathogen, causes dire congenital brain developmental abnormalities in children of infected mothers. The global health crisis precipitated by this virus has led to a concerted effort to develop effective therapies and prophylactic measures although, unfortunately, not very successfully. The error-prone nature of RNA viral genome replication tends to promote evolution of novel viral strains, which could cause epidemics and pandemics. As such, our objective was to develop a safe and effective replication-deficient ZIKV vector-based vaccine candidate. We approached this by generating a ZIKV vector containing only the nonstructural (NS) 5'-untranslated (UTR)-NS-3' UTR sequences, with the structural proteins capsid (C), precursor membrane (prM), and envelope (E) (CprME) used as a packaging system. We efficiently packaged replication-deficient Zika vaccine particles in human producer cells and verified antigen expression in vitro. In vivo studies showed that, after inoculation in neonatal mice, the Zika vaccine candidate (ZVAX) was safe and did not produce any replication-competent revertant viruses. Immunization of adult, nonpregnant mice showed that ZVAX protected mice from lethal challenge by limiting viral replication. We then evaluated the safety and efficacy of ZVAX in pregnant mice, where it was shown to provide efficient maternal and fetal protection against Zika disease. Mass cytometry analysis showed that vaccinated pregnant animals had high levels of splenic CD8+ T cells and effector memory T cell responses with reduced proinflammatory cell responses, suggesting that endogenous expression of NS proteins by ZVAX induced cellular immunity against ZIKV NS proteins. We also investigated humoral immunity against ZIKV, which is potentially induced by viral proteins present in ZVAX virions. We found no significant difference in neutralizing antibody titer in vaccinated or unvaccinated challenged animals; therefore, it is likely that cellular immunity plays a major role in ZVAX-mediated protection against ZIKV infection. In conclusion, we demonstrated ZVAX as an effective inducer of protective immunity against ZIKV, which can be further evaluated for potential prophylactic application in humans. IMPORTANCE This research is important as it strives to address the critical need for effective prophylactic measures against the outbreak of Zika virus (ZIKV) and outlines an important vaccine technology that could potentially be used to induce immune responses against other pandemic-potential viruses.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Nikhil Chakravarty
- Department of Epidemiology, University of California, Los Angeles, Los Angeles, California, USA
| | - Angel Elma Abu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Kari-Ann Takano
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Rebecca Brown
- Departments of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Kouki Morizono
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
9
|
Mungin JW, Chen X, Liu B. Interferon Epsilon Signaling Confers Attenuated Zika Replication in Human Vaginal Epithelial Cells. Pathogens 2022; 11:853. [PMID: 36014974 PMCID: PMC9415962 DOI: 10.3390/pathogens11080853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 02/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus that causes congenital birth defects and neurological compilations in the human host. Although ZIKV is primarily transmitted through infected mosquitos, recent studies reveal sexual contact as a potential transmission route. In vagina-bearing individuals, the vaginal epithelium constitutes the first line of defense against viruses. However, it is unclear how ZIKV interacts with the vaginal epithelium to initiate ZIKV transmission. In this study, we demonstrate that exposing ZIKV to human vaginal epithelial cells (hVECs) resulted in de novo viral RNA replication, increased envelope viral protein production, and a steady, extracellular release of infectious viral particles. Interestingly, our data show that, despite an increase in viral load, the hVECs did not exhibit significant cytopathology in culture as other cell types typically do. Furthermore, our data reveal that the innate antiviral state of hVECs plays a crucial role in preventing viral cytopathology. For the first time, our data show that interferon epsilon inhibits ZIKV replication. Collectively, our results in this study provide a novel perspective on the viral susceptibility and replication dynamics during ZIKV infection in the human vaginal epithelium. These findings will be instrumental towards developing therapeutic agents aimed at eliminating the pathology caused by the virus.
Collapse
Affiliation(s)
| | | | - Bindong Liu
- Centers for AIDS Health Disparity Research, Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (J.W.M.J.); (X.C.)
| |
Collapse
|
10
|
Luria-Pérez R, Sánchez-Vargas LA, Muñoz-López P, Mellado-Sánchez G. Mucosal Vaccination: A Promising Alternative Against Flaviviruses. Front Cell Infect Microbiol 2022; 12:887729. [PMID: 35782117 PMCID: PMC9241634 DOI: 10.3389/fcimb.2022.887729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae are a family of positive-sense, single-stranded RNA enveloped viruses, and their members belong to a single genus, Flavivirus. Flaviviruses are found in mosquitoes and ticks; they are etiological agents of: dengue fever, Japanese encephalitis, West Nile virus infection, Zika virus infection, tick-borne encephalitis, and yellow fever, among others. Only a few flavivirus vaccines have been licensed for use in humans: yellow fever, dengue fever, Japanese encephalitis, tick-borne encephalitis, and Kyasanur forest disease. However, improvement is necessary in vaccination strategies and in understanding of the immunological mechanisms involved either in the infection or after vaccination. This is especially important in dengue, due to the immunological complexity of its four serotypes, cross-reactive responses, antibody-dependent enhancement, and immunological interference. In this context, mucosal vaccines represent a promising alternative against flaviviruses. Mucosal vaccination has several advantages, as inducing long-term protective immunity in both mucosal and parenteral tissues. It constitutes a friendly route of antigen administration because it is needle-free and allows for a variety of antigen delivery systems. This has promoted the development of several ways to stimulate immunity through the direct administration of antigens (e.g., inactivated virus, attenuated virus, subunits, and DNA), non-replicating vectors (e.g., nanoparticles, liposomes, bacterial ghosts, and defective-replication viral vectors), and replicating vectors (e.g., Salmonella enterica, Lactococcus lactis, Saccharomyces cerevisiae, and viral vectors). Because of these characteristics, mucosal vaccination has been explored for immunoprophylaxis against pathogens that enter the host through mucosae or parenteral areas. It is suitable against flaviviruses because this type of immunization can stimulate the parenteral responses required after bites from flavivirus-infected insects. This review focuses on the advantages of mucosal vaccine candidates against the most relevant flaviviruses in either humans or animals, providing supporting data on the feasibility of this administration route for future clinical trials.
Collapse
Affiliation(s)
- Rosendo Luria-Pérez
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
| | - Luis A. Sánchez-Vargas
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Paola Muñoz-López
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
- Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gabriela Mellado-Sánchez
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Ciudad de México, Mexico
| |
Collapse
|
11
|
Feng G, Zhang J, Zhang Y, Li C, Zhang D, Li Y, Zhou H, Li N, Xiao P. Metagenomic Analysis of Togaviridae in Mosquito Viromes Isolated From Yunnan Province in China Reveals Genes from Chikungunya and Ross River Viruses. Front Cell Infect Microbiol 2022; 12:849662. [PMID: 35223559 PMCID: PMC8878809 DOI: 10.3389/fcimb.2022.849662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
We collected 5,500 mosquitoes belonging to six species in three locations in China. Their viromes were tested using metagenomic sequencing and bioinformatic analysis. The affluent viral sequences that were detected and annotated belong to 22 viral taxonomic families. Then, PCR was performed to confirm the results, followed by phylogenetic analysis. Herein, part of mosquito virome was identified, including chikungunya virus (CHIKV), Getah virus (GETV), and Ross river virus (RRV). After metagenomic analysis, seven CHIKV sequences were verified by PCR amplification, among which CHIKV-China/YN2018-1 had the highest homology with the CHIKV isolated in Senegal, 1983, with a nucleotide (nt) identity of at least 81%, belonging to genotype West Africa viral genes. Five GETV sequences were identified, which had a high homology with the GETV sequences isolated from Equus caballus in Japan, 1978, with a (nt) identity of at least 97%. The newly isolated virus CHIKV-China/YN2018-1 became more infectious after passage of the BHK-21 cell line to the Vero cell line. The newly identified RRV gene had the highest homology with the 2006 RRV isolate from Australia, with a (nt) identity of at least 94%. In addition, numerous known and unknown viruses have also been detected in mosquitoes from Yunnan province, China, and propagation tests will be carried out.
Collapse
Affiliation(s)
- Guanrong Feng
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Jinyong Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Ying Zhang
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China
| | - Chenghui Li
- College of Agriculture, Yanbian University, Yanji, China
| | - Duo Zhang
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, China
| | | | - Nan Li
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
- *Correspondence: Nan Li, ; Pengpeng Xiao,
| | - Pengpeng Xiao
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
- *Correspondence: Nan Li, ; Pengpeng Xiao,
| |
Collapse
|
12
|
Zhang T, Warden AR, Li Y, Ding X. Progress and applications of mass cytometry in sketching immune landscapes. Clin Transl Med 2020; 10:e206. [PMID: 33135337 PMCID: PMC7556381 DOI: 10.1002/ctm2.206] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Recently emerged mass cytometry (cytometry by time-of-flight [CyTOF]) technology permits the identification and quantification of inherently diverse cellular systems, and the simultaneous measurement of functional attributes at the single-cell resolution. By virtue of its multiplex ability with limited need for compensation, CyTOF has led a critical role in immunological research fields. Here, we present an overview of CyTOF, including the introduction of CyTOF principle and advantages that make it a standalone tool in deciphering immune mysteries. We then discuss the functional assays, introduce the bioinformatics to interpret the data yield via CyTOF, and depict the emerging clinical and research applications of CyTOF technology in sketching immune landscape in a wide variety of diseases.
Collapse
Affiliation(s)
- Ting Zhang
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Antony R. Warden
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yiyang Li
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Xianting Ding
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
13
|
Blitvich BJ, Magalhaes T, Laredo-Tiscareño SV, Foy BD. Sexual Transmission of Arboviruses: A Systematic Review. Viruses 2020; 12:v12090933. [PMID: 32854298 PMCID: PMC7552039 DOI: 10.3390/v12090933] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/15/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) are primarily maintained in nature in transmission cycles between hematophagous arthropods and vertebrate hosts, but an increasing number of arboviruses have been isolated from or indirectly detected in the urogenital tract and sexual secretions of their vertebrate hosts, indicating that further investigation on the possibility of sexual transmission of these viruses is warranted. The most widely recognized sexually-transmitted arbovirus is Zika virus but other arboviruses, including Crimean-Congo hemorrhagic fever virus and dengue virus, might also be transmitted, albeit occasionally, by this route. This review summarizes our current understanding on the ability of arboviruses to be sexually transmitted. We discuss the sexual transmission of arboviruses between humans and between vertebrate animals, but not arthropod vectors. Every taxonomic group known to contain arboviruses (Asfarviridae, Bunyavirales, Flaviviridae, Orthomyxoviridae, Reoviridae, Rhabdoviridae and Togaviridae) is covered.
Collapse
Affiliation(s)
- Bradley J. Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
- Correspondence: ; Tel.: +1-515-294-9861; Fax: +1-515-294-8500
| | - Tereza Magalhaes
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| | - S. Viridiana Laredo-Tiscareño
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Brian D. Foy
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| |
Collapse
|