1
|
Côrtes N, Lira A, Silva JDQ, Carvalho E, Prates-Syed WA, Hamaguchi B, Durães-Carvalho R, Balan A, Câmara NOS, Cabral-Marques O, Pardi N, Sabino EC, Krieger JE, Cabral-Miranda G. A VLPs based vaccine protects against Zika virus infection and prevents cerebral and testicular damage. NPJ Vaccines 2025; 10:107. [PMID: 40425591 PMCID: PMC12116995 DOI: 10.1038/s41541-025-01163-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Still, Zika virus (ZIKV) infection poses a substantial public health risk, especially for pregnant women and their fetuses, as it can result in congenital abnormalities and fetal mortality during pregnancy. Despite significant advances in understanding and combating ZIKV, considerable challenges remain in the fight against this flavivirus. A crucial component of this effort is the development of vaccines, none of which have yet been licensed for human use. Here, we present a comprehensive study of a novel ZIKV vaccine candidate based on virus-like particles (VLPs), designed to provide broad immunological protection against viral infection combined with safety, without the need for additional adjuvants. A self-adjuvanted VLPs-based vaccine displaying the envelope protein domain III (EDIII) of ZIKV was built. The EDIII protein was expressed in E. coli and chemically conjugated to QβVLPs. Immunization of C57BL/6 mice with two doses of the EDIII-QβVLPs vaccine elicited strong EDIII-specific Th1-based immune response. Notably, the vaccine induced neutralizing antibodies and conferred protection in type I IFN receptor-deficient (G129) mice against ZIKV challenge. Furthermore, vaccinated male mice were protected from ZIKV-induced cerebral and testicular damage, critical concerns for ZIKV pathogenesis. These findings suggest that the EDIII-QβVLP vaccine is a promising candidate for preventing ZIKV infection, with potential applications in combatting this and other emerging flaviviruses.
Collapse
Affiliation(s)
- Nelson Côrtes
- Department of Infectious Diseases and Tropical Medicine, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline Lira
- Department of Infectious Diseases and Tropical Medicine, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jaqueline D Q Silva
- Department of Infectious Diseases and Tropical Medicine, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Evelyn Carvalho
- Department of Infectious Diseases and Tropical Medicine, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Wasim A Prates-Syed
- Department of Infectious Diseases and Tropical Medicine, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Barbara Hamaguchi
- Department of Infectious Diseases and Tropical Medicine, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Biophysics, São Paulo School of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Ricardo Durães-Carvalho
- Department of Microbiology, Immunology and Parasitology, São Paulo School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, Brazil
- Interunit Bioinformatics Graduate Program, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| | - Andrea Balan
- Applied Structural Biology Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Niels O S Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- DO'R Institute for Research, São Paulo, Brazil
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ester C Sabino
- Department of Pathology, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - José E Krieger
- Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Heart Institute, São Paulo, Brazil
| | - Gustavo Cabral-Miranda
- Department of Infectious Diseases and Tropical Medicine, Laboratory of Medical Investigation 46, Faculty of Medicine, University of São Paulo, São Paulo, Brazil.
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
2
|
Bi G, Yu Q, Guo J, Zhang S, Wang P, Jiang X, Wu C, Hu S, Yang X, Fang JH, Long Y, Bi H. Development and Validation of a HPLC-MS/MS Method for the Determination of Compound 13b in Rat Plasma and Its Application to a Pharmacokinetic Study. Biomed Chromatogr 2025; 39:e70044. [PMID: 40071502 DOI: 10.1002/bmc.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 05/13/2025]
Abstract
Compound 13b, a newly identified anthraquinone derivative, has demonstrated potent efficacy against the Zika virus. To explore the bioavailability and pharmacokinetic properties of compound 13b, a robust and sensitive HPLC-MS/MS method was established and verified to determine its plasma concentration in rats. Sample preparation involved protein precipitation using acetonitrile with testosterone as an internal standard. A Phenomenex Kinetex XB-C18 column (2.1 × 100 mm, 2.6 μm) was utilized for sample separation with gradient elution. The mobile phase, consisting of water and acetonitrile, was dispensed at a flow rate of 1 mL/min. The multiple reaction monitoring mode (MRM) approach was used to detect compound 13b and testosterone, characterized by their respective ionization transitions: m/z 410.2 → 91.1 and m/z 289.2 → 109.2. The method demonstrated superior linearity within rat plasma samples, ranging from 2 to 400 ng/mL, and met all FDA guidelines for bioanalytical method validation. The pharmacokinetic properties were calculated by non-compartmental approach following administration of compound 13b at 14 mg/kg in rats, and the absolute oral bioavailability was found to be 15.45%. Hence, a highly sensitive and rapid HPLC-MS/MS method for measuring plasma concentration of compound 13b in rats has been successfully developed, rigorously validated, and subsequently utilized in a bioavailability and pharmacokinetic study.
Collapse
Affiliation(s)
- Guofang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Qingqing Yu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiayin Guo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuqing Zhang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| | - Peng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaowen Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Chenghua Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuang Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, China
| | - Jian-Hong Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yuhua Long
- School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, China
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, China
| |
Collapse
|
3
|
Hueso L, Martorell S, Sena-Torralba A, Ferrando M, Ferri M, Maquieira A, Ntoumi F, Morais S. Recombinase polymerase amplification technology for point-of-care diagnosis of neglected tropical diseases. Int J Infect Dis 2025; 153:107831. [PMID: 39900222 DOI: 10.1016/j.ijid.2025.107831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025] Open
Abstract
Recombinase Polymerase Amplification (RPA) technology significantly advances the diagnostics of neglected tropical diseases (NTDs), providing rapid, isothermal, and minimally preparative testing ideally suited for under-resourced countries. This review critically assesses the current applications, limitations, and potential of RPA for detecting a broad spectrum of NTD pathogens, including viruses, bacteria, helminths, and fungi. The ability of RPA to operate under constant temperature conditions without the need for complex thermal cycling facilitates rapid pathogen detection within minutes, enhancing its utility for decentralized point-of-care testing in remote and underserved regions. RPA, however, faces limitations, including the labor-intensive and costly validation of primer design, especially for multiplex assays, and a susceptibility to nonspecific amplification. These challenges highlight the need for continuous refinement to ensure reliable and consistent performance across diverse environmental conditions. Despite these constraints, the scalability of RPA assays and their compatibility with portable detection platforms make them well-suited for deployment in field settings without access to traditional laboratory infrastructure. This review emphasizes the transformative potential of RPA in NTD diagnostics, enhancing accessibility, precision, and timeliness of interventions, ultimately contributing to improved global public health outcomes.
Collapse
Affiliation(s)
- Luisa Hueso
- Kveloce (Senior Europa S.L.), Plaza de la Reina 19, Valencia, Spain
| | - Sara Martorell
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
| | - Amadeo Sena-Torralba
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
| | - Maite Ferrando
- Kveloce (Senior Europa S.L.), Plaza de la Reina 19, Valencia, Spain
| | - Mireia Ferri
- Kveloce (Senior Europa S.L.), Plaza de la Reina 19, Valencia, Spain
| | - Angel Maquieira
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain; Departamento de Química, Universitat Politècnica de València, Valencia, Spain
| | - Francine Ntoumi
- Institute for Tropical Medicine, University of Tübingen, Germany; Congolese Foundation for Medical Research (FCRM), Brazzaville, Republic of Congo
| | - Sergi Morais
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain; Departamento de Química, Universitat Politècnica de València, Valencia, Spain.
| |
Collapse
|
4
|
Mattar S, López Y, Arrieta G, Mattar AS, Bandeira AC, Paniz-Mondolfi A, Drexler JF, Rodriguez-Morales AJ. The next arbovirus epidemic in Latin America and beyond: A question of when, not if - Mayaro, Oropouche, Usutu or Disease X? Travel Med Infect Dis 2025; 64:102818. [PMID: 39954795 DOI: 10.1016/j.tmaid.2025.102818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Affiliation(s)
- Salim Mattar
- Instituto de Investigaciones Biologicas del Tropico, Universidad de Córdoba, Monteria, Colombia.
| | - Yesica López
- Instituto de Investigaciones Biologicas del Tropico, Universidad de Córdoba, Monteria, Colombia.
| | - German Arrieta
- Instituto de Investigaciones Biologicas del Tropico, Universidad de Córdoba, Monteria, Colombia; Corporación Universitaria del Caribe -CECAR, Grupo Salud Pública y Auditoría en Salud, Sincelejo, Sucre, Colombia.
| | - Ameth Salim Mattar
- Faculty of Medicine, Universidad del Norte, Atlantico, Barranquilla, Colombia.
| | | | | | - Jan Felix Drexler
- Institute of Virology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Centre for Infection Research (DZIF), associated partner Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Alfonso J Rodriguez-Morales
- Faculty of Health Sciences, Universidad Científica del Sur, Lima, Peru; Grupo de Investigación Biomedicina, Facultad de Medicina, Fundación Universitaria Autónoma de las Américas-Institución Universitaria Visión de las Américas, Risaralda, Pereira, 660003, Colombia.
| |
Collapse
|
5
|
Nicacio JM, de Souza CDF, Khouri R, Pereira VC, do Carmo RF, Patriota PVADM, Nunes SLP, de Morais Júnior JC, Barral-Netto M, Lima JAC, Armstrong ADC. Arbovirus exposure and subclinical myocardial dysfunction in an Indigenous population in Northeast Brazil: a cross-sectional study. BMC Infect Dis 2025; 25:89. [PMID: 39833670 PMCID: PMC11748565 DOI: 10.1186/s12879-024-10320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Human activities, such as urbanization and climate change, have facilitated the spread of arbovirus-carrying vectors, disproportionately affecting vulnerable traditional Indigenous communities. OBJECTIVE To explore the relationships between subclinical myocardial dysfunction, assessed by global longitudinal strain (GLS), and comprehensive arbovirus serology in an Indigenous population, while also describing the serological and epidemiological profile of dengue, chikungunya, and Zika viruses. METHODS This ancillary study is part of the first phase (2016-2017) of the Project of Atherosclerosis among Indigenous Populations (PAI), a cross-sectional study involving participants from two Indigenous communities with different degrees of urbanization and a highly urbanized city in Northeast Brazil. We assessed the seroprevalence of dengue, chikungunya, and Zika viruses in the Fulni-ô Indigenous community, the less urbanized and most traditional group. Additionally, we explored the relationship between these viruses and subclinical heart disease, assessed by speckle-tracking echocardiography-derived GLS. RESULTS One hundred seventy-four participants were included, with a median age of 45.0 years (interquartile range 38.0-55.0). The majority were female (58.6%; n = 102). The prevalence of anti-ZIKV IgG was 95.3%; anti-DENV IgG was 85.8%, and anti-CHIKV IgG was 70.9%. GLS abnormalities were detected in nearly half (48.3%) of the cohort. However, no significant association was found between arbovirus serology and GLS. CONCLUSIONS The findings reveal a high prevalence of positive serology for arboviruses and a significant rate of subclinical cardiac dysfunction. There was no significant association between reduced left ventricular longitudinal strain and positive arbovirus serology, likely due to the limited number of participants with indications of acute contact with the viruses studied. However, the unprecedented and relevant results of this study are noteworthy, as they address critical public health issues, particularly in vulnerable populations. Further research is needed to explore these findings in more depth.
Collapse
Affiliation(s)
- Jandir Mendonça Nicacio
- College of Medicine, Federal University of Vale do São Francisco-UNIVASF, Petrolina, Pernambuco, 56304-917, Brazil.
- Postgraduate Program in Human Ecology and Socio-Environmental Management, Bahia State University- UNEB, Juazeiro, Bahia, 48900-000, Brazil.
- Department of Medicine, Federal University of Vale do São Francisco- UNIVASF, Petrolina, Pernambuco, Brazil.
| | - Carlos Dornels Freire de Souza
- College of Medicine, Federal University of Vale do São Francisco-UNIVASF, Petrolina, Pernambuco, 56304-917, Brazil
- Postgraduate Program in Epidemiology and Health Problems Control - Oswaldo Cruz Foundation/Fiocruz, Recife, Pernambuco, 50670-420, Brazil
| | - Ricardo Khouri
- Oswaldo Cruz Foundation/Fiocruz, Institute Gonçalo Moniz, Salvador, Bahia, 40296-710, Brazil
- School of Medicine, Federal University of Bahia - UFBA, Salvador, Bahia, 40110-909, Brazil
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Vanessa Cardoso Pereira
- Postgraduate Program in Human Ecology and Socio-Environmental Management, Bahia State University- UNEB, Juazeiro, Bahia, 48900-000, Brazil
| | - Rodrigo Feliciano do Carmo
- College of Medicine, Federal University of Vale do São Francisco-UNIVASF, Petrolina, Pernambuco, 56304-917, Brazil
| | | | - Sávio Luiz Pereira Nunes
- Postgraduate Program in Applied Cellular and Molecular Biology, University of Pernambuco- UPE, Recife, Pernambuco, 50100-010, Brazil
| | | | - Manoel Barral-Netto
- Oswaldo Cruz Foundation/Fiocruz, Institute Gonçalo Moniz, Salvador, Bahia, 40296-710, Brazil
- School of Medicine, Federal University of Bahia - UFBA, Salvador, Bahia, 40110-909, Brazil
- National Institute of Science and Technology for Immunology Research, University of São Paulo- USP, São Paulo, 05468-901, Brazil
| | | | - Anderson da Costa Armstrong
- College of Medicine, Federal University of Vale do São Francisco-UNIVASF, Petrolina, Pernambuco, 56304-917, Brazil
- Postgraduate Program in Human Ecology and Socio-Environmental Management, Bahia State University- UNEB, Juazeiro, Bahia, 48900-000, Brazil
| |
Collapse
|
6
|
Martínez-Rojas PP, Monroy-Martínez V, Ruiz-Ordaz BH. Role of extracellular vesicles in the pathogenesis of mosquito-borne flaviviruses that impact public health. J Biomed Sci 2025; 32:4. [PMID: 39754219 PMCID: PMC11699717 DOI: 10.1186/s12929-024-01096-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/08/2024] [Indexed: 01/06/2025] Open
Abstract
Mosquito-borne flaviviruses represent a public health challenge due to the high-rate endemic infections, severe clinical outcomes, and the potential risk of emerging global outbreaks. Flavivirus disease pathogenesis converges on cellular factors from vectors and hosts, and their interactions are still unclear. Exosomes and microparticles are extracellular vesicles released from cells that mediate the intercellular communication necessary for maintaining homeostasis; however, they have been shown to be involved in disease establishment and progression. This review focuses on the roles of extracellular vesicles in the pathogenesis of mosquito-borne flavivirus diseases: how they contribute to viral cycle completion, cell-to-cell transmission, and cellular responses such as inflammation, immune suppression, and evasion, as well as their potential use as biomarkers or therapeutics (antiviral or vaccines). We highlight the current findings concerning the functionality of extracellular vesicles in different models of dengue virus, Zika virus, yellow fever virus, Japanese encephalitis virus, and West Nile virus infections and diseases. The available evidence suggests that extracellular vesicles mediate diverse functions between hosts, constituting novel effectors for understanding the pathogenic mechanisms of flaviviral diseases.
Collapse
Affiliation(s)
- Pedro Pablo Martínez-Rojas
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), 04510, Mexico City, Mexico
| | - Verónica Monroy-Martínez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), 04510, Mexico City, Mexico
| | - Blanca H Ruiz-Ordaz
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), 04510, Mexico City, Mexico.
| |
Collapse
|
7
|
Yuan M, Tian X, Ma W, Zhang R, Zou X, Jin Y, Zheng N, Wu Z, Wang Y. miRNA-431-5p enriched in EVs derived from IFN-β stimulated MSCs potently inhibited ZIKV through CD95 downregulation. Stem Cell Res Ther 2024; 15:435. [PMID: 39563434 PMCID: PMC11575116 DOI: 10.1186/s13287-024-04040-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/03/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Zika virus (ZIKV) primarily spreads through mosquito bites and can lead to microcephaly in infants and Guillain-Barre syndrome in adults. It is noteworthy that ZIKV can persist in the semen of infected males for extended periods and can be sexually transmitted. Infection with ZIKV has severe pathological manifestations on the testicular tissues of male mice, resulting in reduced sperm motility and fertility. However, there are no approved prophylactic vaccines or therapeutics available to treat Zika virus infection. METHODS Using a male type I and II interferon receptor-deficient (ifnar1(-/-) ifngr1(-/-)) C57BL/6 (AG6) mouse model infected with ZIKV as a representative model, we evaluated the degree of testicular damage and viral replication in various organs in mice treated with EVs derived from MSC-stimulated with IFN-β (IFNβ-EVs) and treated with controls. We measured testicle size, detected viral load in various organs, and analyzed gene expression to assess treatment efficacy. RESULTS Our findings demonstrated that intravenous administration of IFNβ-EVs effectively suppressed ZIKV replication in the testes. Investigation with in-depth RNA sequencing analysis found that IFN-β treatment changed the cargo miRNA of EVs. Notably, miR-431-5p was identified to be significantly enriched in IFNβ-EVs and exhibited potent antiviral activity in vitro. We showed that CD95 was a direct downstream target for miR-431-5p and played a role in facilitating ZIKV replication. miR-431-5p effectively downregulated the expression of CD95 protein, consequently promoted the phosphorylation and nuclear localization of NF-kB, which resulted in the activation of anti-viral status, leading to the suppression of viral replication. CONCLUSIONS Our study demonstrated that the EVs produced by IFNβ-treated MSCs could effectively convey antiviral activity.
Collapse
Affiliation(s)
- Meng Yuan
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xiaoyan Tian
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, 210093, China
| | - Wenyuan Ma
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, 210093, China
| | - Rui Zhang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, PR China
| | - Xue Zou
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Yu Jin
- Department of Clinical Medicine, Medical School of Nanjing University , Nanjing, 210093, China.
- Nanjing Children's Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| | - Nan Zheng
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, 210093, China.
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, People's Republic of China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, People's Republic of China.
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School,Nanjing University, Yangzhou, China.
| |
Collapse
|
8
|
Wang YT, Hsieh YC, Wu TY. In silico validation of allosteric inhibitors targeting Zika virus NS2B-NS3 protease. Phys Chem Chem Phys 2024; 26:27684-27693. [PMID: 39469836 DOI: 10.1039/d4cp02867h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The Zika virus (ZIKV), a member of the Flaviviridae family, poses a major threat to human health because of the lack of effective antiviral drugs. Although the NS2B-NS3 protease of ZIKV (NS2B-NS3pro) is regarded as a major target for antiviral inhibitors, viral mutations can lead to ineffective competitive inhibitors. Allosteric inhibitors bind to highly conserved nonprotease active sites, induce conformational changes in the protease active site, and prevent substrate binding. Currently, no molecular simulation techniques are available for accurately predicting and analysing conformational changes in the protease catalytic domain. In this study, we developed a combined approach that involves blind docking, Gaussian accelerated molecular dynamics, two-dimensional potential of mean force profiling, density functional theory (DFT) calculations, and interaction region indicator (IRI) analysis and employed it to examine the allosteric inhibitor-01 molecule and its interaction with ZIKV NS2B-NS3pro. Our results indicated that the binding of inhibitor-01 to NS2B-NS3pro resulted in two major conformational states, state I and state II, which in turn changed the volume of the protease active site from 1014 Å3 to 710 and 820 Å3, respectively. These two states had an inactive catalytic domain (residues His116, Asp140, and Ser200). DFT and IRI analyses revealed that, in state I, Lys138 and Gln139 formed hydrogen bonds with inhibitor-01, whereas Lys138, Leu214, Asn217, Val220, and Ile221 engaged in van der Waals interactions with inhibitor-01. Advancements in computational techniques and power are expected to facilitate further progress in overcoming challenges associated with designing allosteric inhibitors for viral proteases.
Collapse
Affiliation(s)
- Yeng-Tseng Wang
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Taiwan, ROC.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
| | - Yuan-Chin Hsieh
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan, ROC
| | - Tin-Yu Wu
- Department of Management Information Systems, National Pingtung University of Science and Technology, Taiwan, ROC
| |
Collapse
|
9
|
Khairallah M, Abroug N, Smit D, Chee SP, Nabi W, Yeh S, Smith JR, Ksiaa I, Cunningham E. Systemic and Ocular Manifestations of Arboviral Infections: A Review. Ocul Immunol Inflamm 2024; 32:2190-2208. [PMID: 38441549 DOI: 10.1080/09273948.2024.2320724] [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: 11/05/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 10/26/2024]
Abstract
PURPOSE To provide an overview of pre-selected emerging arboviruses (arthropod-borne viruses) that cause ocular inflammation in humans. METHODS A comprehensive review of the literature published between 1997 and 2023 was conducted in PubMed database. We describe current insights into epidemiology, systemic and ocular manifestations, diagnosis, treatment, and prognosis of arboviral diseases including West Nile fever, Dengue fever, Chikungunya, Rift Valley fever, Zika, and Yellow fever. RESULTS Arboviruses refer to a group of ribonucleic acid viruses transmitted to humans by the bite of hematophagous arthropods, mainly mosquitoes. They mostly circulate in tropical and subtropical zones and pose important public health challenges worldwide because of rising incidence, expanding geographic range, and occurrence of prominent outbreaks as a result of climate change, travel, and globalization. The clinical signs associated with infection from these arboviruses are often inapparent, mild, or non-specific, but they may include serious, potentially disabling or life-threatening complications. A wide spectrum of ophthalmic manifestations has been described including conjunctival involvement, anterior uveitis, intermediate uveitis, various forms of posterior uveitis, maculopathy, optic neuropathy, and other neuro-ophthalmic manifestations. Diagnosis of arboviral diseases is confirmed with either real time polymerase chain reaction or serology. Management involves supportive care as there are currently no specific antiviral drug options. Corticosteroids are often used for the treatment of associated ocular inflammation. Most patients have a good visual prognosis, but there may be permanent visual impairment due to ocular structural complications in some. Community-based integrated mosquito management programs and personal protection measures against mosquito bites are the best ways to prevent human infection and disease. CONCLUSION Emerging arboviral diseases should be considered in the differential diagnosis of ocular inflammatory conditions in patients living in or returning from endemic regions. Early clinical consideration followed by confirmatory testing can limit or prevent unnecessary treatments for non-arboviral causes of ocular inflammation. Prevention of these infections is crucial.
Collapse
Affiliation(s)
- Moncef Khairallah
- Department of Ophthalmology, Fattouma Bourguiba University Hospital, Faculty of Medicine, University of Monastir, Monastir, Tunisia
| | - Nesrine Abroug
- Department of Ophthalmology, Fattouma Bourguiba University Hospital, Faculty of Medicine, University of Monastir, Monastir, Tunisia
| | - Derrick Smit
- Division of Ophthalmology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Soon-Phaik Chee
- Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Eye Research Institute, Singapore, Singapore
- Department of Ophthalmology & Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- Eye & Retina Surgeons, Singapore, Singapore
| | - Wijden Nabi
- Department of Ophthalmology, Fattouma Bourguiba University Hospital, Faculty of Medicine, University of Monastir, Monastir, Tunisia
| | - Steven Yeh
- Department of Ophthalmology, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Justine R Smith
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Imen Ksiaa
- Department of Ophthalmology, Fattouma Bourguiba University Hospital, Faculty of Medicine, University of Monastir, Monastir, Tunisia
| | - Emmett Cunningham
- The Department of Ophthalmology, California Pacific Medical Center, San Francisco, California, USA
- The Department of Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
- The Francis I. Proctor Foundation, UCSF School of Medicine, San Francisco, California, USA
| |
Collapse
|
10
|
Marques EM, Rocha RL, Brandão CM, Xavier JKAM, Camara MBP, Mendonça CDJS, de Lima RB, Souza MP, Costa EV, Gonçalves RS. Development of an Eco-Friendly Nanogel Incorporating Pectis brevipedunculata Essential Oil as a Larvicidal Agent Against Aedes aegypti. Pharmaceutics 2024; 16:1337. [PMID: 39458666 PMCID: PMC11510620 DOI: 10.3390/pharmaceutics16101337] [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: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Arboviruses, transmitted by mosquitoes like Aedes aegypti, pose significant public health challenges globally, particularly in tropical regions. The rapid spread and adaptation of viruses such as Dengue, Zika, and Chikungunya have emphasized the need for innovative control methods. Essential oils from plants, such as Pectis brevipedunculata (Gardner) Sch.Bip. (Pb), have emerged as potential alternatives to conventional insecticides. METHODS In this work, we developed an eco-friendly nanogel using a low-energy, solvent-free method, incorporating the copolymer F127 and Carbopol 974p, enriched with a high concentration of essential oil from Pb (EOPb). The resulting nanogel displayed excellent physical stability, maintained under varying temperature conditions. Characterization techniques, including FTIR and DLS, confirmed the stable incorporation of EOPb within the nanogel matrix. RESULTS The in vitro assays against Aedes aegypti larvae revealed that at 500 μg/mL, the mortality rates were 96.0% ± 7.0 after 24 h and 100.0% ± 0.0 after 48 h. The positive control group treated with temefos, achieved 100% mortality at both time points, validating the experimental conditions and providing a benchmark for assessing the efficacy of the nGF2002Pb nanogel. CONCLUSIONS These results indicate that nGF2002Pb demonstrates a pronounced concentration-dependent larvicidal effect against Aedes aegypti, offering an innovative and sustainable approach to arbovirus vector control.
Collapse
Affiliation(s)
- Estela Mesquita Marques
- Laboratory of Chemistry of Natural Products, Department of Chemistry, Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil; (E.M.M.); (R.L.R.); (J.K.A.M.X.); (M.B.P.C.)
| | - Raiene Lisboa Rocha
- Laboratory of Chemistry of Natural Products, Department of Chemistry, Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil; (E.M.M.); (R.L.R.); (J.K.A.M.X.); (M.B.P.C.)
| | | | - Júlia Karla Albuquerque Melo Xavier
- Laboratory of Chemistry of Natural Products, Department of Chemistry, Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil; (E.M.M.); (R.L.R.); (J.K.A.M.X.); (M.B.P.C.)
| | - Marcos Bispo Pinheiro Camara
- Laboratory of Chemistry of Natural Products, Department of Chemistry, Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil; (E.M.M.); (R.L.R.); (J.K.A.M.X.); (M.B.P.C.)
| | - Caritas de Jesus Silva Mendonça
- Center for Fuels, Catalysis, and Environment (NCCA), Department of Chemistry, Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil;
| | | | - Melissa Pires Souza
- Postgraduate Program in Chemistry, Federal University of Amazonas (UFAM), Manaus 69080-900, Brazil; (M.P.S.); (E.V.C.)
| | - Emmanoel Vilaça Costa
- Postgraduate Program in Chemistry, Federal University of Amazonas (UFAM), Manaus 69080-900, Brazil; (M.P.S.); (E.V.C.)
- Department of Chemistry, Federal University of Amazonas (UFAM), Manaus 69080-900, Brazil
| | - Renato Sonchini Gonçalves
- Laboratory of Chemistry of Natural Products, Department of Chemistry, Federal University of Maranhão (UFMA), São Luís 65080-805, Brazil; (E.M.M.); (R.L.R.); (J.K.A.M.X.); (M.B.P.C.)
| |
Collapse
|
11
|
Palmero Casanova B, Albentosa González L, Maringer K, Sabariegos R, Mas A. A conserved role for AKT in the replication of emerging flaviviruses in vertebrates and vectors. Virus Res 2024; 348:199447. [PMID: 39117146 PMCID: PMC11364138 DOI: 10.1016/j.virusres.2024.199447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/11/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
One third of all emerging infectious diseases are vector-borne, with no licensed antiviral therapies available against any vector-borne viruses. Zika virus and Usutu virus are two emerging flaviviruses transmitted primarily by mosquitoes. These viruses modulate different host pathways, including the PI3K/AKT/mTOR pathway. Here, we report the effect on ZIKV and USUV replication of two AKT inhibitors, Miransertib (ARQ-092, allosteric inhibitor) and Capivasertib (AZD5363, competitive inhibitor) in different mammalian and mosquito cell lines. Miransertib showed a stronger inhibitory effect against ZIKV and USUV than Capivasertib in mammalian cells, while Capivasertib showed a stronger effect in mosquito cells. These findings indicate that AKT plays a conserved role in flavivirus infection, in both the vertebrate host and invertebrate vector. Nevertheless, the specific function of AKT may vary depending on the host species. These findings indicate that AKT may be playing a conserved role in flavivirus infection in both, the vertebrate host and the invertebrate vector. However, the specific function of AKT may vary depending on the host species. A better understanding of virus-host interactions is therefore required to develop new treatments to prevent human disease and new approaches to control transmission by insect vectors.
Collapse
Affiliation(s)
- Blanca Palmero Casanova
- Instituto de Investigación Biomédica de la UCLM (IB-UCLM), C/Almansa 14, 02008 Albacete, Spain
| | - Laura Albentosa González
- Instituto de Investigación Biomédica de la UCLM (IB-UCLM), C/Almansa 14, 02008 Albacete, Spain; Facultad de farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02008 Albacete, Spain
| | - Kevin Maringer
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, UK
| | - Rosario Sabariegos
- Instituto de Investigación Biomédica de la UCLM (IB-UCLM), C/Almansa 14, 02008 Albacete, Spain; Unidad asociada de Biomedicina UCLM-CSIC. Universidad de Castilla-La Mancha. C/Altagracia 50, 13071 Ciudad Real, Spain; Facultad de Medicina, Universidad de Castilla-La Mancha. C/Almansa 14, 02008 Albacete, Spain
| | - Antonio Mas
- Instituto de Investigación Biomédica de la UCLM (IB-UCLM), C/Almansa 14, 02008 Albacete, Spain; Facultad de farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02008 Albacete, Spain; Unidad asociada de Biomedicina UCLM-CSIC. Universidad de Castilla-La Mancha. C/Altagracia 50, 13071 Ciudad Real, Spain.
| |
Collapse
|
12
|
Aguiar GRF, da Silva GB, Ramalho JDAM, Srisawat N, Daher EDF. Common arboviruses and the kidney: a review. J Bras Nefrol 2024; 46:e20230168. [PMID: 39074252 PMCID: PMC11287847 DOI: 10.1590/2175-8239-jbn-2023-0168en] [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: 11/16/2023] [Accepted: 05/17/2024] [Indexed: 07/31/2024] Open
Abstract
Arboviruses are endemic in several countries and represent a worrying public health problem. The most important of these diseases is dengue fever, whose numbers continue to rise and have reached millions of annual cases in Brazil since the last decade. Other arboviruses of public health concern are chikungunya and Zika, both of which have caused recent epidemics, and yellow fever, which has also caused epidemic outbreaks in our country. Like most infectious diseases, arboviruses have the potential to affect the kidneys through several mechanisms. These include the direct action of the viruses, systemic inflammation, hemorrhagic phenomena and other complications, in addition to the toxicity of the drugs used in treatment. In this review article, the epidemiological aspects of the main arboviruses in Brazil and other countries where these diseases are endemic, clinical aspects and the main laboratory changes found, including changes in renal function, are addressed. It also describes how arboviruses behave in kidney transplant patients. The pathophysiological mechanisms of kidney injury associated with arboviruses are described and finally the recommended treatment for each disease and recommendations for kidney support in this context are given.
Collapse
Affiliation(s)
- Gabriel Rotsen Fortes Aguiar
- Universidade Federal do Ceará, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Departamento de Medicina Interna, Fortaleza, CE, Brazil
| | - Geraldo Bezerra da Silva
- Universidade de Fortaleza, Centro de Ciências da Saúde, Curso de Medicina, Fortaleza, CE, Brazil
| | - Janaína de Almeida Mota Ramalho
- Universidade Federal do Ceará, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Departamento de Medicina Interna, Fortaleza, CE, Brazil
- Universidade de Fortaleza, Centro de Ciências da Saúde, Curso de Medicina, Fortaleza, CE, Brazil
| | - Nattachai Srisawat
- Chulalongkorn University, Faculty of Medicine, Department of Medicine, Division of Nephrology, Center of Excellence for Critical Care Nephrology, and Tropical Medicine Cluster, Bangkok, Tailândia
| | - Elizabeth de Francesco Daher
- Universidade Federal do Ceará, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Departamento de Medicina Interna, Fortaleza, CE, Brazil
| |
Collapse
|
13
|
Duan ZL, Zou WW, Chen D, Zhu JY, Wen JS. Japanese encephalitis virus E protein domain III immunization mediates cross-protection against Zika virus in mice via antibodies and CD8 +T cells. Virus Res 2024; 345:199376. [PMID: 38643856 PMCID: PMC11046216 DOI: 10.1016/j.virusres.2024.199376] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/10/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Zika virus (ZIKV) and Japanese encephalitis virus (JEV) are antigenically related flaviviruses that co-circulate in many countries/territories. The interaction between the two viruses needs to be determined. Recent findings by ourselves and other labs showed that JEV-elicited antibodies (Abs) and CD8+T cells exacerbate and protect against subsequent ZIKV infection, respectively. However, the impact of JEV envelope (E) protein domain III (EDIII)-induced immune responses on ZIKV infection is unclear. We show here that sera from JEV-EDIII-vaccinated mice cross-react with ZIKV-EDIII in vitro, and transfer of the same sera to mice significantly decreases death upon lethal ZIKV infection at a dose-dependent manner. Maternally acquired anti-JEV-EDIII Abs also significantly reduce the mortality of neonatal mice born to JEV-EDIII-immune mothers post ZIKV challenge. Similarly, transfer of ZIKV-EDIII-reactive IgG purified from JEV-vaccinated humans increases the survival of ZIKV-infected mice. Notably, transfer of an extremely low volume of JEV-EDIII-immune sera or ZIKV-EDIII-reactive IgG does not mediate the Ab-mediated enhancement (ADE) of ZIKV infection. Similarly, transfer of JEV-EDIII-elicited CD8+T cells protects recipient mice against ZIKV challenge. These results demonstrate that JEV-EDIII-induced immune components including Abs and T cells have protective roles in ZIKV infection, suggesting EDIII is a promising immunogen for developing effective and safety JEV vaccine.
Collapse
Affiliation(s)
- Zhi-Liang Duan
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Department of Clinical Laboratory, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Wei-Wei Zou
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Dong Chen
- Wenzhou Central Blood Station, Wenzhou, China; Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
| | - Jia-Yang Zhu
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Jin-Sheng Wen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
14
|
Xie F, Zhu Q. The regulation of cGAS-STING signaling by RNA virus-derived components. Virol J 2024; 21:101. [PMID: 38693578 PMCID: PMC11064393 DOI: 10.1186/s12985-024-02359-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024] Open
Abstract
The Cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) serves as a key innate immune signaling axis involved in the regulation of various human diseases. It has been found that cGAS-STING pathway can recognize a variety of cytosolic double-stranded DNA (dsDNA), contributing to cause a robust type I interferon response thereby affecting the occurrence and progression of viral infection. Accumulating evidence indicates RNA virus-derived components play an important role in regulating cGAS-STING signaling, either as protective or pathogenic factors in the pathogenesis of diseases. Thus, a comprehensive understanding of the function of RNA virus-derived components in regulating cGAS-STING signaling will provide insights into developing novel therapies. Here, we review the existing literature on cGAS-STING pathway regulated by RNA virus-derived components to propose insights into pharmacologic strategies targeting the cGAS-STING pathway.
Collapse
Affiliation(s)
- Feiting Xie
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China.
| | - Qiugang Zhu
- Department of Laboratory Medicine, Shangyu People's Hospital of Shaoxing, Shaoxing, China
| |
Collapse
|
15
|
Pereira SH, Sá Magalhães Serafim M, Moraes TDFS, Zini N, Abrahão JS, Nogueira ML, Coelho dos Reis JGA, Bagno FF, da Fonseca FG. Design, development, and validation of multi-epitope proteins for serological diagnosis of Zika virus infections and discrimination from dengue virus seropositivity. PLoS Negl Trop Dis 2024; 18:e0012100. [PMID: 38635656 PMCID: PMC11025737 DOI: 10.1371/journal.pntd.0012100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Zika virus (ZIKV), an arbovirus from the Flaviviridae family, is the causative agent of Zika fever, a mild and frequent oligosymptomatic disease in humans. Nonetheless, on rare occasions, ZIKV infection can be associated with Guillain-Barré Syndrome (GBS), and severe congenital complications, such as microcephaly. The oligosymptomatic disease, however, presents symptoms that are quite similar to those observed in infections caused by other frequent co-circulating arboviruses, including dengue virus (DENV). Moreover, the antigenic similarity between ZIKV and DENV, and even with other members of the Flaviviridae family, complicates serological testing due to the high cross-reactivity of antibodies. Here, we designed, produced in a prokaryotic expression system, and purified three multiepitope proteins (ZIKV-1, ZIKV-2, and ZIKV-3) for differential diagnosis of Zika. The proteins were evaluated as antigens in ELISA tests for the detection of anti-ZIKV IgG using ZIKV- and DENV-positive human sera. The recombinant proteins were able to bind and detect anti-ZIKV antibodies without cross-reactivity with DENV-positive sera and showed no reactivity with Chikungunya virus (CHIKV)- positive sera. ZIKV-1, ZIKV-2, and ZIKV-3 proteins presented 81.6%, 95%, and 66% sensitivity and 97%, 96%, and 84% specificity, respectively. Our results demonstrate the potential of the designed and expressed antigens in the development of specific diagnostic tests for the detection of IgG antibodies against ZIKV, especially in regions with the circulation of multiple arboviruses.
Collapse
Affiliation(s)
- Samille Henriques Pereira
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mateus Sá Magalhães Serafim
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thaís de Fátima Silva Moraes
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nathalia Zini
- Laboratório de Pesquisa em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- Centro de Tecnologia em Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jônatas Santos Abrahão
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maurício Lacerda Nogueira
- Laboratório de Pesquisa em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | | | - Flávia Fonseca Bagno
- Centro de Tecnologia em Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Centro de Tecnologia em Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
16
|
Huang GG, Wang HY, Wang XH, Yang T, Zhang XM, Feng CL, Zhao WM, Tang W. Atranorin inhibits Zika virus infection in human glioblastoma cell line SNB-19 via targeting Zika virus envelope protein. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155343. [PMID: 38290230 DOI: 10.1016/j.phymed.2024.155343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/26/2023] [Accepted: 01/07/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Zika virus (ZIKV) is a single-stranded RNA flavivirus transmitted by mosquitoes. Its infection is associated with neurological complications such as neonatal microcephaly and adult Guillain-Barré syndrome, posing a serious threat to the health of people worldwide. Therefore, there is an urgent need to develop effective anti-ZIKV drugs. Atranorin is a lichen secondary metabolite with a wide range of biological activities, including anti-inflammatory, antibacterial and antioxidant, etc. However, the antiviral activity of atranorin and underlying mechanism has not been fully elucidated. PURPOSE We aimed to determine the anti-ZIKV activity of atranorin in human glioma cell line SNB-19 and investigate the potential mechanism from the perspective of viral life cycle and the host cell functions. METHODS We first established ZIKV-infected human glioma cells (SNB-19) model and used Western Blot, RT-qPCR, immunofluorescence, fluorescence-activated cell sorting (FACS) and plaque assay to evaluate the anti-ZIKV activity of atranorin. Then we assessed the regulation effect of atranorin on ZIKV induced IFN signal pathway activation by RT-qPCR. Afterward, we introduced time-of-addition assay, viral adsorption assay, viral internalization assay and transferrin uptake assay to define which step of ZIKV lifecycle is influenced by atranorin. Finally, we performed virus infectivity assay, molecular docking and thermal shift assay to uncover the target protein of atranorin on ZIKV. RESULTS Our study showed that atranorin could protect SNB-19 cells from ZIKV infection, as evidenced by inhibited viral protein expression and progeny virus yield. Meanwhile, atranorin attenuated the activation of IFN signal pathway and downstream inflammatory response that induced by ZIKV infection. The results of time-of-addition assay indicated that atranorin acted primarily by disturbing the viral entry process. After ruling out the effect of atranorin on AXL receptor tyrosine kinase (AXL) dependent virus adsorption and clathrin-mediated endocytosis, we confirmed that atranorin directly targeted the viral envelope protein and lowered ZIKV infectivity by thermal shift assay and virus infectivity assay respectively. CONCLUSION We found atranorin inhibits ZIKV infection in SNB-19 cells via targeting ZIKV envelope protein. Our study provided an experimental basis for the further development of atranorin and a reference for antiviral drug discovery from natural resources.
Collapse
Affiliation(s)
- Guan-Gen Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Hao-Yu Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiao-Han Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Tao Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiao-Meng Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Chun-Lan Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Wei-Min Zhao
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Wei Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
17
|
Malik S, Muhammad K, Ahsan O, Khan MT, Sah R, Waheed Y. Advances in Zika virus vaccines and therapeutics: A systematic review. ASIAN PAC J TROP MED 2024; 17:97-109. [DOI: 10.4103/apjtm.apjtm_680_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 03/15/2024] [Indexed: 12/06/2024] Open
Abstract
Zika virus (ZIKV) is the causative agent of a viral infection that causes neurological complications in newborns and adults worldwide. Its wide transmission route and alarming spread rates are of great concern to the scientific community. Numerous trials have been conducted to develop treatment options for ZIKV infection. This review highlights the latest developments in the fields of vaccinology and pharmaceuticals developments for ZIKV infection. A systematic and comprehensive approach was used to gather relevant and up-to-date data so that inferences could be made about the gaps in therapeutic development. The results indicate that several therapeutic interventions are being tested against ZIKV infection, such as DNA vaccines, subunit vaccines, live-attenuated vaccines, virus-vector-based vaccines, inactivated vaccines, virus-like particles, and mRNA-based vaccines. In addition, approved anti-ZIKV drugs that can reduce the global burden are discussed. Although many vaccine candidates for ZIKV are at different stages of development, none of them have received Food and Drug Authority approval for use up to now. The issue of side effects associated with these drugs in vulnerable newborns and pregnant women is a major obstacle in the therapeutic pathway.
Collapse
Affiliation(s)
- Shiza Malik
- Bridging Health Foundation, Rawalpindi 46000, Pakistan
| | - Khalid Muhammad
- Department of Biology, College of Sciences, UAE University, 15551, Al Ain, United Arab Emirates
| | - Omar Ahsan
- Department of Medicine, Foundation University Medical College, Foundation University Islamabad, Islamabad 44000, Pakistan
| | - Muhammad Tahir Khan
- INTI International University, Persiaran Perdana BBN Putra Nilai, 71800 Nilai, Negeri Sembilan, Malaysia
- Institute of Molecular Biology and Biotechnology, the University of Lahore, KM Defence Road, Lahore 58810, Pakistan
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Nanyang 473006, China
| | - Ranjit Sah
- Department of Microbiology, Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu 44600, Nepal
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
| | - Yasir Waheed
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos 1401, Lebanon
| |
Collapse
|
18
|
Serafim MSM, Kronenberger T, Rocha REO, Rosa ADRA, Mello TLG, Poso A, Ferreira RS, Abrahão JS, Kroon EG, Mota BEF, Maltarollo VG. Aminopyrimidine Derivatives as Multiflavivirus Antiviral Compounds Identified from a Consensus Virtual Screening Approach. J Chem Inf Model 2024; 64:393-411. [PMID: 38194508 DOI: 10.1021/acs.jcim.3c01505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Around three billion people are at risk of infection by the dengue virus (DENV) and potentially other flaviviruses. Worldwide outbreaks of DENV, Zika virus (ZIKV), and yellow fever virus (YFV), the lack of antiviral drugs, and limitations on vaccine usage emphasize the need for novel antiviral research. Here, we propose a consensus virtual screening approach to discover potential protease inhibitors (NS3pro) against different flavivirus. We employed an in silico combination of a hologram quantitative structure-activity relationship (HQSAR) model and molecular docking on characterized binding sites followed by molecular dynamics (MD) simulations, which filtered a data set of 7.6 million compounds to 2,775 hits. Lastly, docking and MD simulations selected six final potential NS3pro inhibitors with stable interactions along the simulations. Five compounds had their antiviral activity confirmed against ZIKV, YFV, DENV-2, and DENV-3 (ranging from 4.21 ± 0.14 to 37.51 ± 0.8 μM), displaying aggregator characteristics for enzymatic inhibition against ZIKV NS3pro (ranging from 28 ± 7 to 70 ± 7 μM). Taken together, the compounds identified in this approach may contribute to the design of promising candidates to treat different flavivirus infections.
Collapse
Affiliation(s)
- Mateus Sá Magalhães Serafim
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Tübingen 72076, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Rafael Eduardo Oliveira Rocha
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Amanda Del Rio Abreu Rosa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Thaysa Lara Gonçalves Mello
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Antti Poso
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- Department of Medical Oncology and Pneumology, University Hospital of Tübingen, Tübingen 70211, Germany
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Jonatas Santos Abrahão
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Erna Geessien Kroon
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Bruno Eduardo Fernandes Mota
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Vinícius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| |
Collapse
|
19
|
Ma Z, Guo J, Jiang L, Zhao S. Lateral flow immunoassay (LFIA) for dengue diagnosis: Recent progress and prospect. Talanta 2024; 267:125268. [PMID: 37813013 DOI: 10.1016/j.talanta.2023.125268] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/22/2023] [Accepted: 10/01/2023] [Indexed: 10/11/2023]
Abstract
Dengue is one of the most widespread and fatal arboviral infections in the world. Early detection of dengue virus (DENV) is essential to prevent the spread of the disease and provide an immediate response. The lateral flow immunoassay (LFIA) systems are low-cost, rapid, sensitive, targeted, and straightforward detection, which is an ideal early detection candidate for point-of-care testing (POCT) in dengue-affected areas. However, current commercial LFIA kits cannot fully satisfy the sensitivity, specificity, serotype differentiation, and multiplex detection requirements. Therefore, various strategies have been applied to optimize the LFIA for DENV detection, including label material improvement, optical enhancement and novel structure design. In this review, we comprehensively presented the snapshot of dengue, the principle of LFIA, and recent progress in the LFIA optimization for dengue diagnoses. Furthermore, this review also discusses insights into the prospect of LFIA dengue diagnostic methods, such as microfluidics, multiplex design, nucleic acid-typed probes and smartphone-assisted result analysis.
Collapse
Affiliation(s)
- Ziting Ma
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China
| | - Jinnian Guo
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China
| | - Lu Jiang
- Department of Biomedical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Suqing Zhao
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
20
|
Dobhal K, Garg R, Singh A, Semwal A. Insight into the Natural Biomolecules (BMs): Promising Candidates as Zika Virus Inhibitors. Infect Disord Drug Targets 2024; 24:e020224226681. [PMID: 38318833 DOI: 10.2174/0118715265272414231226092146] [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: 08/03/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 02/07/2024]
Abstract
Zika virus (ZIKV) is among the relatively new infectious disease threats that include SARS-CoV-2, coronavirus, monkeypox (Mpox) virus, etc. ZIKV has been reported to cause severe health risks to the fetus. To date, satisfactory treatment is still not available for the treatment of ZIKV infection. This review examines the last five years of work using natural biomolecules (BMs) to counteract the ZIKV through virtual screening and in vitro investigations. Virtual screening has identified doramectin, pinocembrin, hesperidins, epigallocatechin gallate, pedalitin, and quercetin as potentially active versus ZIKV infection. In vitro, testing has shown that nordihydroguaiaretic acid, mefloquine, isoquercitrin, glycyrrhetinic acid, patentiflorin-A, rottlerin, and harringtonine can reduce ZIKV infections in cell lines. However, in vivo, testing is limited, fortunately, emetine, rottlerin, patentiflorin-A, and lycorine have shown in vivo anti- ZIKV potential. This review focuses on natural biomolecules that show a particularly high selective index (>10). There is limited in vivo and clinical trial data for natural BMs, which needs to be an active area of investigation. This review aims to compile the known reference data and discuss the barriers associated with discovering and using natural BM agents to control ZIKV infection.
Collapse
Affiliation(s)
- Kiran Dobhal
- College of Pharmacy, Shivalik College, Dehradun, Uttarakhand, India
| | - Ruchika Garg
- School of Pharmacy, Maharaja Agrasen Universities, Baddi, Solan, Himachal Pradesh, 174103, India
| | - Alka Singh
- School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University Balawala, Dehradun, Uttarakhand, India
| | - Amit Semwal
- College of Pharmacy, Shivalik College, Dehradun, Uttarakhand, India
| |
Collapse
|
21
|
Ferraz AC, Gomes PWP, Menegatto MBDS, Lima RLS, Guimarães PH, Reis JDE, Carvalho ARV, Pamplona SDGSR, Muribeca ADJB, de Magalhães JC, Yoshioka E Silva CY, da Silva MN, Magalhães CLDB. Exploring the antiviral potential of justicidin B and four glycosylated lignans from Phyllanthus brasiliensis against Zika virus: A promising pharmacological approach. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155197. [PMID: 37952409 DOI: 10.1016/j.phymed.2023.155197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Zika virus (ZIKV) is an emerging arbovirus that in recent years has been associated with cases of severe neurological disorders, such as microcephaly in newborns and Guillain-Barré syndrome in adults. As there is no vaccine or treatment, the search for new therapeutic targets is of great relevance. In this sense, plants are extremely rich sources for the discovery of new bioactive compounds and the species Phyllanthus brasiliensis (native to the Amazon region) remains unexplored. PURPOSE To investigate the potential antiviral activity of compounds isolated from P. brasiliensis leaves against ZIKV infection. METHODS In vitro antiviral assays were performed with justicidin B (a lignan) and four glycosylated lignans (tuberculatin, phyllanthostatin A, 5-O-β-d-glucopyranosyljusticidin B, and cleistanthin B) against ZIKV in Vero cells. MTT colorimetric assay was used to assess cell viability and plaque forming unit assay to quantify viral load. In addition, for justicidin B, tests were performed to investigate the mechanism of action (virucidal, adsorption, internalization, post-infection). RESULTS The isolated compounds showed potent anti-ZIKV activities and high selectivity indexes. Moreover, justicidin B, tuberculatin, and phyllanthostatin A completely reduced the viral load in at least one of the concentrations evaluated. Among them, justicidin B stood out as the main active, and further investigation revealed that justicidin B exerts its antiviral effect during post-infection stages, resulting in a remarkable 99.9 % reduction in viral load when treatment was initiated 24 h after infection. CONCLUSION Our findings suggest that justicidin B inhibits endosomal internalization and acidification, effectively interrupting the viral multiplication cycle. Therefore, the findings shed light on the promising potential of isolated compounds isolated from P. brasiliensis, especially justicidin B, which could contribute to the drug development and treatments for Zika virus infections.
Collapse
Affiliation(s)
- Ariane Coelho Ferraz
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Paulo Wender P Gomes
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, San Diego, California, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, San Diego, California, USA
| | - Marília Bueno da Silva Menegatto
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Rafaela Lameira Souza Lima
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Pedro Henrique Guimarães
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - José Diogo Evangelista Reis
- Instituto de Ciências Exatas e Naturais, Programa de Pós-Graduação em Química, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Alice Rhelly Veloso Carvalho
- Instituto de Ciências da Saúde, Faculdade de Ciências Farmacêuticas, Universidade Federal do Pará, Belém, Pará, Brazil
| | | | - Abraão de Jesus Barbosa Muribeca
- Instituto de Ciências Exatas e Naturais, Programa de Pós-Graduação em Química, Universidade Federal do Pará, Belém, Pará, Brazil
| | - José Carlos de Magalhães
- Departamento de Química, Biotecnologia e Engenharia de Bioprocessos, Universidade Federal de São João del-Rei, Ouro Branco, Minas Gerais, Brazil; Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Consuelo Yumiko Yoshioka E Silva
- Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Milton Nascimento da Silva
- Instituto de Ciências Exatas e Naturais, Programa de Pós-Graduação em Química, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Cintia Lopes de Brito Magalhães
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
22
|
Gonçalves SMC, Galdino LV, Lima MC, da Silva Moura JA, Viana DCF, da Rosa MM, Ferreira LFGR, Hernandes MZ, Pereira MC, de Melo Rêgo MJB, da Rocha Pitta I, de Oliveira França R, da Rocha Pitta MG, da Rocha Pitta MG. Evaluation of Thiazolidine Derivatives with Potential Anti-ZIKV Activity. Curr Top Med Chem 2024; 24:2224-2237. [PMID: 39136505 DOI: 10.2174/0115680266315388240801053401] [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: 03/08/2024] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE In this study, we have synthesized 19 Thiazolidine (TZD) derivatives to investigate their potential anti-ZIKV effects. METHODS Nineteen thiazolidine derivatives were synthesized and evaluated for their cytotoxicity and antiviral activity against the ZIKA virus. RESULTS Among them, six demonstrated remarkable selectivity against the ZIKV virus, exhibiting IC50 values of <5μM, and the other compounds did not demonstrate selectivity for the virus. Interestingly, several derivatives effectively suppressed the replication of ZIKV RNA copies, with derivatives significantly reducing ZIKV mRNA levels at 24 hours post-infection (hpi). Notably, two derivatives (ZKC-4 and -9) stood out by demonstrating a protective effect against ZIKV cell entry. Informed by computational analysis of binding affinity and intermolecular interactions within the NS5 domain's N-7 and O'2 positions, ZKC-4 and FT-39 displayed the highest predicted affinities. Intriguingly, ZKC-4 and ZKC-9 derivatives exhibited the most favorable predicted binding affinities for the ZIKV-E binding site. CONCLUSION The significance of TZDs as potent antiviral agents is underscored by these findings, suggesting that exploring TZD derivatives holds promise for advancing antiviral therapeutic strategies.
Collapse
Affiliation(s)
| | - Lília Vieira Galdino
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Morganna Costa Lima
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation / FIOCRUZ, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - José Arion da Silva Moura
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Douglas Carvalho Francisco Viana
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Michelle Melgarejo da Rosa
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | | | - Marcelo Zaldini Hernandes
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Artur de Sá, Recife-PE, 50740-521, Brazil
| | - Michelly Cristiny Pereira
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | | | - Ivan da Rocha Pitta
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Rafael de Oliveira França
- Department of Virology and Experimental Therapy, Aggeu Magalhães Institute, Oswaldo Cruz Foundation / FIOCRUZ, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Marina Galdino da Rocha Pitta
- Pharmaceutical Planning and Synthesis Laboratory, Federal University of Pernambuco Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| | - Maira Galdino da Rocha Pitta
- Therapeutic Innovation Research Center, Federal University of Pernambuco, Av. Moraes Rego, 1235, Recife, 50670-901, Brazil
| |
Collapse
|
23
|
Brango-Vanegas J, Leite ML, de Oliveira KBS, da Cunha NB, Franco OL. From exploring cancer and virus targets to discovering active peptides through mRNA display. Pharmacol Ther 2023; 252:108559. [PMID: 37952905 DOI: 10.1016/j.pharmthera.2023.108559] [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: 08/09/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
During carcinogenesis, neoplastic cells accumulate mutations in genes important for cellular homeostasis, producing defective proteins. Viral infections occur when viral capsid proteins bind to the host cell receptor, allowing the virus to enter the cells. In both cases, proteins play important roles in cancer development and viral infection, so these targets can be exploited to develop alternative treatments. mRNA display technology is a very powerful tool for the development of peptides capable of acting on specific targets in neoplastic cells or on viral capsid proteins. mRNA display technology allows the selection and evolution of peptides with desired functional properties from libraries of many nucleic acid variants. Among other advantages of this technology, the use of flexizymes allows the production of peptides with unnatural amino acid residues, which can enhance the activity of these molecules. From target immobilization, peptides with greater specificity for the targets of interest are generated during the selection rounds. Herein, we will explore the use of mRNA display technology for the development of active peptides after successive rounds of selection, using proteins present in neoplastic cells and viral particles as targets.
Collapse
Affiliation(s)
- José Brango-Vanegas
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Michel Lopes Leite
- Departamento de Biologia Molecular, Instituto de Ciências Biológicas, Universidade de Brasília, Campus Darcy Ribeiro, Brasília, DF, Brazil
| | - Kamila Botelho Sampaio de Oliveira
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Nicolau Brito da Cunha
- Universidade de Brasília, Faculdade de Agronomia e Medicina Veterinária, Campus Darcy Ribeiro, Brasília, DF, Brazil
| | - Octávio Luiz Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil.
| |
Collapse
|
24
|
Bhandari V, Taksande AB, Sapkale B. Disease Transmission and Diagnosis of Zika Virus. Cureus 2023; 15:e49263. [PMID: 38465265 PMCID: PMC10923262 DOI: 10.7759/cureus.49263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/22/2023] [Indexed: 03/12/2024] Open
Abstract
An arbovirus belonging to the Flaviviridae family and the Flavivirus genus, the Zika virus (ZIKV), has profoundly transformed global health perception. Historically, ZIKV infections were considered infrequent, with generally mild manifestations. However, this perception changed dramatically when the virus quickly spread from Asia to the Americas, impacting many nations. It was alarming that there was a connection between ZIKV infection in pregnant women and the beginning of microcephaly in their offspring. ZIKV control and treatment are further complicated because Aedes mosquitoes, which primarily bite during the day, are the primary vectors of the virus. ZIKV diagnostic processes are complex since the virus shares symptoms with other illnesses like dengue and chikungunya. Despite the effectiveness of current diagnostic methods like real-time reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA), there is a clear need for more accurate antibody tests. This is especially true given that many people undergo testing while asymptomatic or after the ideal detection window. The capacity of ZIKV to infect human-derived neural progenitor cells raises worrying possibilities for severe neurological effects. With all these characteristics and their connection to birth abnormalities, research efforts into the virus's efficient treatment and prevention have increased. Overall, the emergence of ZIKV has demonstrated the necessity of a comprehensive and team-based strategy to address its myriad problems. This entails comprehending its transmission dynamics, enhancing diagnostic accuracy, and creating efficient therapies and preventive measures, all crucial to lessening the threat that ZIKV poses to the world's health.
Collapse
Affiliation(s)
- Vinaya Bhandari
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Avinash B Taksande
- Physiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Bhagyesh Sapkale
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
25
|
Aquino VH, Fumagalli MJ, Silva A, de Moura Negrini BV, Rojas A, Guillen Y, Bernal C, Figueiredo LTM. Linear epitope mapping in the E and NS1 proteins of dengue and Zika viruses: Prospection of peptides for vaccines and diagnostics. PLoS One 2023; 18:e0292451. [PMID: 37788262 PMCID: PMC10547212 DOI: 10.1371/journal.pone.0292451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
The arrival of the Zika virus (ZIKV) in dengue virus (DENV)-endemic areas has posed challenges for both differential diagnosis and vaccine development. Peptides have shown promise in addressing these issues. The aim of this study was to identify the linear epitope profile recognized by serum samples from dengue and Zika patients in the E and NS1 proteins of DENV and ZIKV. This cross-sectional study included individuals of all ages with laboratory-confirmed DENV and ZIKV infections, who were selected through convenience sampling. The serum samples from dengue and Zika patients detected epitopes evenly distributed across the viral proteins in a peptide microarray platform. However, several epitopes were located within "epitope hotspots", characterized by clusters of peptides recognized in more than 30% of the sub-arrays analyzed using individual or pooled serum samples. The serum samples from dengue and Zika patients showed a high level of cross-reactivity with peptides in the DENV and ZIKV proteins. Analysis using an additional peptide microarray platform, which contained peptides selected based on the results of the initial screening, revealed that two DENV and one ZIKV peptide, highly specific to their related viruses, were located within the epitope hotspots; however, they presented low detection rates (32.5, 35.0, and 28.6%, respectively). In addition, two DENV peptides detected at similarly high rates by both dengue and Zika patients were also found within the epitope hotspots. These hotspots contain several immunodominant epitopes that are recognized by a larger number of individuals when compared to 15-amino acid (aa) sequence peptides. Thus, epitope hotspots may have greater potential to serve as antigens in diagnostic tests and vaccine development than peptides composed of only 15 amino acids.
Collapse
Affiliation(s)
- Victor Hugo Aquino
- Immunology Department, Research Institute for Health Sciences, National University of Asuncion, San Lorenzo, Central, Paraguay
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Marcilio J. Fumagalli
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Angélica Silva
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | | | - Alejandra Rojas
- Production Department, Research Institute for Health Sciences, National University of Asuncion, San Lorenzo, Central, Paraguay
| | - Yvalena Guillen
- Production Department, Research Institute for Health Sciences, National University of Asuncion, San Lorenzo, Central, Paraguay
| | - Cynthia Bernal
- Production Department, Research Institute for Health Sciences, National University of Asuncion, San Lorenzo, Central, Paraguay
| | - Luiz Tadeu Moraes Figueiredo
- Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
26
|
da Costa HHM, Bielavsky M, Orts DJB, Araujo S, Adriani PP, Nogueira JS, Astray RM, Pandey RP, Lancellotti M, Cunha-Junior JP, Prudencio CR. Production of Recombinant Zika Virus Envelope Protein by Airlift Bioreactor as a New Subunit Vaccine Platform. Int J Mol Sci 2023; 24:13955. [PMID: 37762254 PMCID: PMC10531330 DOI: 10.3390/ijms241813955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 09/29/2023] Open
Abstract
The Zika Virus (ZIKV) is an emerging arbovirus of great public health concern, particularly in the Americas after its last outbreak in 2015. There are still major challenges regarding disease control, and there is no ZIKV vaccine currently approved for human use. Among many different vaccine platforms currently under study, the recombinant envelope protein from Zika Virus (rEZIKV) constitutes an alternative option for vaccine development and has great potential for monitoring ZIKV infection and antibody response. This study describes a method to obtain a bioactive and functional rEZIKV using an E. coli expression system, with the aid of a 5-L airlift bioreactor and following an automated fast protein liquid chromatography (FPLC) protocol, capable of obtaining high yields of approximately 20 mg of recombinant protein per liter of bacterium cultures. The purified rEZIKV presented preserved antigenicity and immunogenicity. Our results show that the use of an airlift bioreactor for the production of rEZIKV is ideal for establishing protocols and further research on ZIKV vaccines bioprocess, representing a promising system for the production of a ZIKV envelope recombinant protein-based vaccine candidate.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Monica Bielavsky
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Diego J. B. Orts
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Laboratory of Cardiobiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, São Paulo 04023-062, Brazil
| | - Sergio Araujo
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Patrícia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Renato M. Astray
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
- Multi-Purpose Laboratory Butantan Institute, São Paulo 05503-900, Brazil
| | - Ramendra P. Pandey
- School of Health Sciences and Technology, UPES University, Dehradun 248007, Uttarakhand, India
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences—FCF, University of Campinas—UNICAMP, Campinas 13083-871, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
27
|
Pereira RS, Santos FCP, Campana PRV, Costa VV, de Pádua RM, Souza DG, Teixeira MM, Braga FC. Natural Products and Derivatives as Potential Zika virus Inhibitors: A Comprehensive Review. Viruses 2023; 15:v15051211. [PMID: 37243296 DOI: 10.3390/v15051211] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/30/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Zika virus (ZIKV) is an arbovirus whose infection in humans can lead to severe outcomes. This article reviews studies reporting the anti-ZIKV activity of natural products (NPs) and derivatives published from 1997 to 2022, which were carried out with NPs obtained from plants (82.4%) or semisynthetic/synthetic derivatives, fungi (3.1%), bacteria (7.6%), animals (1.2%) and marine organisms (1.9%) along with miscellaneous compounds (3.8%). Classes of NPs reported to present anti-ZIKV activity include polyphenols, triterpenes, alkaloids, and steroids, among others. The highest values of the selectivity index, the ratio between cytotoxicity and antiviral activity (SI = CC50/EC50), were reported for epigallocatechin gallate (SI ≥ 25,000) and anisomycin (SI ≥ 11,900) obtained from Streptomyces bacteria, dolastane (SI = 1246) isolated from the marine seaweed Canistrocarpus cervicorni, and the flavonol myricetin (SI ≥ 862). NPs mostly act at the stages of viral adsorption and internalization in addition to presenting virucidal effect. The data demonstrate the potential of NPs for developing new anti-ZIKV agents and highlight the lack of studies addressing their molecular mechanisms of action and pre-clinical studies of efficacy and safety in animal models. To the best of our knowledge, none of the active compounds has been submitted to clinical studies.
Collapse
Affiliation(s)
- Rosângela Santos Pereira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Françoise Camila Pereira Santos
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | | | - Vivian Vasconcelos Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Rodrigo Maia de Pádua
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Daniele G Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Fernão Castro Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| |
Collapse
|
28
|
Shukla R, Chandra A, Kumar A, Kandpal P, Avashthi H, Goel VK, Qamar I, Singh N, Kelvin DJ, Singh TR. Repurposing of drugs against methyltransferase as potential Zika virus therapies. Sci Rep 2023; 13:7870. [PMID: 37188743 PMCID: PMC10184974 DOI: 10.1038/s41598-023-33341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
In recent years, the outbreak of infectious disease caused by Zika Virus (ZIKV) has posed a major threat to global public health, calling for the development of therapeutics to treat ZIKV disease. Several possible druggable targets involved in virus replication have been identified. In search of additional potential inhibitors, we screened 2895 FDA-approved compounds using Non-Structural Protein 5 (NS5) as a target utilizing virtual screening of in-silco methods. The top 28 compounds with the threshold of binding energy -7.2 kcal/mol value were selected and were cross-docked on the three-dimensional structure of NS5 using AutoDock Tools. Of the 2895 compounds screened, five compounds (Ceforanide, Squanavir, Amcinonide, Cefpiramide, and Olmesartan_Medoxomil) ranked highest based on filtering of having the least negative interactions with the NS5 and were selected for Molecular Dynamic Simulations (MDS) studies. Various parameters such as RMSD, RMSF, Rg, SASA, PCA and binding free energy were calculated to validate the binding of compounds to the target, ZIKV-NS5. The binding free energy was found to be -114.53, -182.01, -168.19, -91.16, -122.56, and -150.65 kJ mol-1 for NS5-SFG, NS5-Ceforanide, NS5-Squanavir, NS5-Amcinonide, NS5-Cefpiramide, and NS5-Ol_Me complexes respectively. The binding energy calculations suggested Cefpiramide and Olmesartan_Medoxomil (Ol_Me) as the most stable compounds for binding to NS5, indicating a strong rationale for their use as lead compounds for development of ZIKV inhibitors. As these drugs have been evaluated on pharmacokinetics and pharmacodynamics parameters only, in vitro and in vivo testing and their impact on Zika viral cell culture may suggest their clinical trials on ZIKV patients.
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India
- Centre for Excellence in Healthcare Technologies and Informatics (CEHTI), Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Anshuman Chandra
- School of Biotechnology, Gautam Buddha University, Gautam Buddh Nagar, Greater Noida, Uttar Pradesh, 201312, India
- School of Physical Science, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Anuj Kumar
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Department of Pediatrics, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada
- European Virus Bioinformatics Center, Leutragraben 1, Jena, Germany
| | | | - Himanshu Avashthi
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, Pusa, New Delhi, India
| | - Vijay Kumar Goel
- School of Physical Science, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Imteyaz Qamar
- School of Biotechnology, Gautam Buddha University, Gautam Buddh Nagar, Greater Noida, Uttar Pradesh, 201312, India
| | - Nagendra Singh
- School of Biotechnology, Gautam Buddha University, Gautam Buddh Nagar, Greater Noida, Uttar Pradesh, 201312, India.
| | - David J Kelvin
- Laboratory of Immunity, Shantou University Medical College, Shantou, China.
- Department of Microbiology and Immunology, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada.
- Department of Pediatrics, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada.
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India.
- Centre for Excellence in Healthcare Technologies and Informatics (CEHTI), Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India.
| |
Collapse
|
29
|
Franco EJ, Hanrahan KC, Brown AN. Favipiravir Inhibits Zika Virus (ZIKV) Replication in HeLa Cells by Altering Viral Infectivity. Microorganisms 2023; 11:1097. [PMID: 37317071 PMCID: PMC10223361 DOI: 10.3390/microorganisms11051097] [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: 03/30/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 06/16/2023] Open
Abstract
This study aims to evaluate the antiviral potential of the nucleoside analogue favipiravir (FAV) against ZIKV, an arbovirus for which there are no approved antiviral therapies, in three human-derived cell lines. HeLa (cervical), SK-N-MC (neuronal), and HUH-7 (liver) cells were infected with ZIKV and exposed to different concentrations of FAV. Viral supernatant was sampled daily, and infectious viral burden was quantified by plaque assay. Changes in ZIKV infectivity were quantified by calculating specific infectivity. FAV-related toxicities were also assessed for each cell line in both infected and uninfected cells. Our results demonstrate that FAV activity was most pronounced in HeLa cells, as substantial declines in infectious titers and viral infectivity were observed in this cell type. The decline in infectious virus occurred in an exposure-dependent manner and was more pronounced as FAV exposure times increased. Additionally, toxicity studies showed that FAV was not toxic to any of the three cell lines and, surprisingly, caused substantial improvements in the viability of infected HeLa cells. Although SK-N-MC and HUH-7 cells were susceptible to FAV's anti-ZIKV activity, similar effects on viral infectivity and improvements in cell viability with therapy were not observed. These results indicate that FAV's ability to substantially alter viral infectivity is host cell specific and suggest that the robust antiviral effect observed in HeLa cells is mediated through drug-induced losses of viral infectivity.
Collapse
Affiliation(s)
- Evelyn J. Franco
- Institute for Therapeutic Innovation, Department of Medicine, University of Florida College of Medicine, Orlando, FL 32827, USA; (E.J.F.); (K.C.H.)
- Department of Pharmaceutics, University of Florida College of Pharmacy, Orlando, FL 32827, USA
| | - Kaley C. Hanrahan
- Institute for Therapeutic Innovation, Department of Medicine, University of Florida College of Medicine, Orlando, FL 32827, USA; (E.J.F.); (K.C.H.)
| | - Ashley N. Brown
- Institute for Therapeutic Innovation, Department of Medicine, University of Florida College of Medicine, Orlando, FL 32827, USA; (E.J.F.); (K.C.H.)
- Department of Pharmaceutics, University of Florida College of Pharmacy, Orlando, FL 32827, USA
| |
Collapse
|
30
|
Kumar S, Sreelekshmi PR, Godke YS, Sudeep AB. Vector competence of three species of mosquitoes to Ingwavuma virus ( Manzanilla orthobunyavirus), a new bunyavirus found circulating in India. Virusdisease 2023; 34:15-20. [PMID: 37009260 PMCID: PMC10050543 DOI: 10.1007/s13337-023-00808-z] [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: 09/13/2022] [Accepted: 01/02/2023] [Indexed: 02/05/2023] Open
Abstract
Ingwavuma virus (INGV), a mosquito-borne arbovirus reported from Africa and Southeast Asia has been found circulating in India as evidenced by virus isolation and antibody prevalence. INGV is now classified as Manzanilla orthobunyavirus belonging to family Peribunyaviridae. The virus is maintained in nature in a pig-mosquito-bird cycle. Human infection has been confirmed by virus isolation and detection of neutralizing antibodies. A study was initiated to determine the vector competence of Aedes aegypti, Culex quinquefasciatus, and Cx tritaeniorhynchus mosquitoes to INGV due to their high prevalence in India. Mosquitoes were oral fed on viraemic mice and INGV dissemination to legs, wings, salivary glands (saliva) was studied alongwith virus growth kinetics. The three mosquitoes replicated INGV with maximum titers of 3.7, 3.7 and 4.7log10TCID50/ml respectively and maintained the virus till 16th day post infection. However, vector competence and horizontal transmission to infant mouse was demonstrated only by Cx quinquefasciatus mosquitoes. Vertical or trans-ovarial transmission of INGV could not be demonstrated in the mosquito during the study. Though no major outbreak involving humans has been reported yet, the potential of the virus to replicate in different species of mosquitoes and vertebrates including humans pose a threat to public health should there be a change in its genome.
Collapse
Affiliation(s)
- Surendra Kumar
- Division of Medical Entomology, ICMR-National Institute of Virology, (Indian Council of Medical Research), Microbial Containment Complex, 130/1, Sus Road, Pashan, Pune, 411021 India
| | - P. R. Sreelekshmi
- Division of Medical Entomology, ICMR-National Institute of Virology, (Indian Council of Medical Research), Microbial Containment Complex, 130/1, Sus Road, Pashan, Pune, 411021 India
| | - Y. S. Godke
- Division of Medical Entomology, ICMR-National Institute of Virology, (Indian Council of Medical Research), Microbial Containment Complex, 130/1, Sus Road, Pashan, Pune, 411021 India
| | - A. B. Sudeep
- Division of Medical Entomology, ICMR-National Institute of Virology, (Indian Council of Medical Research), Microbial Containment Complex, 130/1, Sus Road, Pashan, Pune, 411021 India
| |
Collapse
|
31
|
Yang W, Zhang C, Wu YH, Liu LB, Zhen ZD, Fan DY, Song ZR, Chang JT, Wang PG, An J. Mice 3D testicular organoid system as a novel tool to study Zika virus pathogenesis. Virol Sin 2023; 38:66-74. [PMID: 36241087 PMCID: PMC10006202 DOI: 10.1016/j.virs.2022.10.001] [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: 05/27/2022] [Accepted: 10/08/2022] [Indexed: 11/05/2022] Open
Abstract
Zika virus (ZIKV) poses a serious threat to global public health due to its close relationship with neurological and male reproductive damage. However, deficiency of human testicular samples hinders the in-depth research on ZIKV-induced male reproductive system injury. Organoids are relatively simple in vitro models, which could mimic the pathological changes of corresponding organs. In this study, we constructed a 3D testicular organoid model using primary testicular cells from adult BALB/c mice. Similar to the testis, this organoid system has a blood-testis barrier (BTB)-like structure and could synthesize testosterone. ZIKV tropism of testicular cells and ZIKV-induced pathological changes in testicular organoid was also similar to that in mammalian testis. Therefore, our results provide a simple and reproducible in vitro testicular model for the investigations of ZIKV-induced testicular injury.
Collapse
Affiliation(s)
- Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Department of Neurosurgery, Capital Medical University Sanbo Brain Hospital, Beijing, 100093, China
| | - Chen Zhang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan-Hua Wu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Li-Bo Liu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zi-Da Zhen
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dong-Ying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zheng-Ran Song
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jia-Tong Chang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100093, China.
| |
Collapse
|
32
|
Fernandez GJ, Ramírez-Mejía JM, Urcuqui-Inchima S. Transcriptional and post-transcriptional mechanisms that regulate the genetic program in Zika virus-infected macrophages. Int J Biochem Cell Biol 2022; 153:106312. [DOI: 10.1016/j.biocel.2022.106312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
|
33
|
Ramiharimanana FD, Haddad JG, Andrianavalonirina MA, Apel C, Olivon F, Diotel N, Desprès P, Ramanandraibe VV, El Kalamouni C. Antiviral Effect of Stenocline ericoides DC. and Stenocline inuloides DC., Two Flavonoid-Rich Endemic Plants from Madagascar, against Dengue and Zika Viruses. Pharmaceuticals (Basel) 2022; 15:ph15121500. [PMID: 36558951 PMCID: PMC9787939 DOI: 10.3390/ph15121500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Dengue and Zika viruses are identified as the most medically important arthropod-borne viral pathogens. Over the past 20 years, the global dengue incidence has dramatically increased with epidemics of severe dengue where the case fatality rate can reach up to 20% in untreated patients. The association between Zika virus infection and severe congenital anomalies was first reported in 2015. Today no specific antiviral therapies are available for dengue and Zika virus infections, accentuating the need of adapted antiviral strategies based on medicinal plant drug discovery. Plants are a potential source of antiviral phytocompounds which act primarily by blocking virus entry in the host-cell. In the present study, we evaluated whether crude extracts from Stenocline ericoides DC. and Stenocline inuloides DC., two endemic plants from Madagascar, may have antiviral effects against dengue and Zika viruses. We showed that S. ericoides has virucidal action whereas S. inuloides inhibits the early steps of virus infection with a non-cytotoxic effect in human cells. The administration of S. ericoides and S. inuloides extracts in zebrafish had no effect on the behavior of animals at the active doses against dengue and Zika viruses, suggesting the absence of adverse effects at these doses. LC-HRMS2 and molecular networking analyses revealed the richness of these two plants in polyphenols and flavonoid with the presence of clusters of phytocompounds specific to each Stenocline species. Consequently, S. ericoides and S. inuloides represent potential sources for natural and safe antiviral phytocompounds against flaviviruses of medical concern.
Collapse
Affiliation(s)
- Fenia D. Ramiharimanana
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
- International Associated Laboratory, University of Antananarivo-Lyon 1, Antananarivo P.O. Box 906, Madagascar
| | - Juliano G. Haddad
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
| | | | - Cécile Apel
- Institut de Chimie des Substances Naturelles, CNRS, University of Paris-Saclay, UPR 2301, 91198 Gif-sur-Yvette, France
| | | | - Nicolas Diotel
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, UMR 1188, 97491 Sainte Clotilde, France
| | - Philippe Desprès
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
| | | | - Chaker El Kalamouni
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Université de la Réunion, 94791 Sainte Clotilde, France
- Correspondence:
| |
Collapse
|
34
|
Qin ZL, Yao QF, Ren H, Zhao P, Qi ZT. Lipid Droplets and Their Participation in Zika Virus Infection. Int J Mol Sci 2022; 23:ijms232012584. [PMID: 36293437 PMCID: PMC9604050 DOI: 10.3390/ijms232012584] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
Lipid droplets (LDs) are highly conserved and dynamic intracellular organelles. Their functions are not limited to serving as neutral lipid reservoirs; they also participate in non-energy storage functions, such as cell lipid metabolism, protection from cell stresses, maintaining protein homeostasis, and regulating nuclear function. During a Zika virus (ZIKV) infection, the viruses hijack the LDs to provide energy and lipid sources for viral replication. The co-localization of ZIKV capsid (C) protein with LDs supports its role as a virus replication platform and a key compartment for promoting the generation of progeny virus particles. However, in view of the multiple functions of LDs, their role in ZIKV infection needs further elucidation. Here, we review the basic mechanism of LD biogenesis and biological functions and discuss how ZIKV infection utilizes these effects of LDs to facilitate virus replication, along with the future application strategy of developing new antiviral drugs based on the interaction of ZIKV with LDs.
Collapse
|
35
|
Guo Z, Jing W, Liu J, Liu M. The global trends and regional differences in incidence of Zika virus infection and implications for Zika virus infection prevention. PLoS Negl Trop Dis 2022; 16:e0010812. [PMID: 36269778 PMCID: PMC9586358 DOI: 10.1371/journal.pntd.0010812] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 09/14/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Zika virus (ZIKV) infection has potential result in severe birth effects. An improved understanding of global trend and regional differences is needed. METHODS Annual ZIKV infection episodes and incidence rates were collected from Global Burden of Disease Study 2019. Episodes changes and estimated annual percentage changes (EAPCs) of age-standardized incidence rate (ASR) were calculated. Top passenger airport-pairs were obtained from the International Air Transport Association to understand places susceptible to imported ZIKV cases. RESULTS Globally, the ASR increased by an average of 72.85% (95%CI: 16.47% to 156.53%) per year from 2011 to 2015 and subsequently decreased from 20.25 per 100,000 in 2015 to 3.44 per 100,000 in 2019. Most of ZIKV infections clustered in Latin America. The proportion of episodes in Central and Tropical Latin America decreased in 2019 with sporadic episodes elsewhere. High Socio-Demographic Index (SDI) regions had more episodes in 2019 than in 2015. Additionally, 15-49 years group had the largest proportion of episodes, females had a higher number of episodes, and a higher incidence rate of 70 plus group was observed in males than females. Certain cities in Europe, North America and Latin America/Caribbean had a high population mobility in ZIKV outbreak areas considered a high risk of imported cases. CONCLUSIONS ZIKV infection is still a public health threat in Latin America and Caribbean and high SDI regions suffered an increasing trend of ZIKV infection. Interventions such as development of surveillance networks and vector-control should be attached to ZIKV control in these key regions. Reproductive suggestions should be taken to reduce ZIKV-related birth defects for the people of reproductive age who are facing a higher threat of ZIKV infection, especially females. Moreover, surveillance of travellers is needed to reverse the uptrends of travel-related imported ZIKV infection. More studies focusing on ZIKV should be performed to make targeted and effective prevention strategies in the future.
Collapse
Affiliation(s)
- Zirui Guo
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Wenzhan Jing
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jue Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Min Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
36
|
Gomez SA, Rojas-Valencia N, Gomez S, Lans I, Restrepo A. Initial recognition and attachment of the Zika virus to host cells: A molecular dynamics and quantum interaction approach. Chembiochem 2022; 23:e202200351. [PMID: 35951472 DOI: 10.1002/cbic.202200351] [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: 06/19/2022] [Revised: 08/02/2022] [Indexed: 11/08/2022]
Abstract
The zika virus (ZIKV), transmitted to humans from the bites of Aedes Aegypti and Aedes Albopictus mosquitoes produces Zika fever and neurodegenerative disorders that despite affecting millions of people, most recently in Africa and the Americas, has been declared a neglected tropical disease by the World Health Organization. In this work, atomistic molecular dynamics simulations followed by rigorous analysis of the intermolecular interactions reveal crucial aspects of the initial virus···cell molecular recognition and attachment, events that trigger the infectious cycle. Previous experimental studies have shown that Dermatan Sulphate (DS) and Chondroitin Sulphate A (CSA), two glycosaminoglycans which are actually epimers to each other and that are structural constituents of receptors expressed in cell membranes, are the preferred anchorage sites, with a marked preference for DS. Our calculations rationalize this preference from a molecular perspective as follows: when free of the virus, DS has one sulfate group that does not participate in intramolecular strong hydrogen bonds, thus, it is readily available to interact with the envelope protein of the virus (Zika-E), then, after formation of the complexes, Zika-E···DS exhibits ten strong salt brides connecting the two fragments against only six salt bridges and two hydrogen bonds in Zika-E···CSA.
Collapse
Affiliation(s)
- Santiago A Gomez
- University of Antioquia: Universidad de Antioquia, Chemistry, COLOMBIA
| | | | - Sara Gomez
- Scuola Normale Superiore Classe di Scienze, Chemistry, COLOMBIA
| | - Isaias Lans
- University of Antioquia: Universidad de Antioquia, Chemistry, COLOMBIA
| | - Albeiro Restrepo
- Universidad de Antioquia, Chemistry, AA 1226, 00000, Medellin, COLOMBIA
| |
Collapse
|
37
|
Rahman MR, Majumder TR, Apu MAI, Paul AK, Afrose A, Dash BK. CRISPR-Based Programmable Nucleic Acid-Binding Protein Technology Can Specifically Detect Fatal Tropical Disease-Causing Pathogens. J Trop Med 2022; 2022:5390685. [PMID: 36199433 PMCID: PMC9529443 DOI: 10.1155/2022/5390685] [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: 03/18/2022] [Revised: 06/13/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
Diagnostic approaches capable of ultrasensitive pathogen detection from low-volume clinical samples, running without any sophisticated instrument and laboratory setup, are easily field-deployable, inexpensive, and rapid, and are considered ideal for monitoring disease progression and surveillance. However, standard pathogen detection methods, including culture and microscopic observation, antibody-based serologic tests, and primarily polymerase chain reaction (PCR)-oriented nucleic acid screening techniques, have shortcomings that limit their widespread use in responding to outbreaks and regular diagnosis, especially in remote resource-limited settings (RLSs). Recently, clustered regularly interspaced short palindromic repeats (CRISPR)-based programmable technology has emerged to challenge the unmet criteria of conventional methods. It consists of CRISPR-associated proteins (Cas) capable of targeting virtually any specific RNA or DNA genome based on the guide RNA (gRNA) sequence. Furthermore, the discovery of programmable trans-cleavage Cas proteins like Cas12a and Cas13 that can collaterally damage reporter-containing single-stranded DNA or RNA upon formation of target Cas-gRNA complex has strengthened this technology with enhanced sensitivity. Current advances, including automated multiplexing, ultrasensitive single nucleotide polymorphism (SNP)-based screening, inexpensive paper-based lateral flow readouts, and ease of use in remote global settings, have attracted the scientific community to introduce this technology in nucleic acid-based precise detection of bacterial and viral pathogens at the point of care (POC). This review highlights CRISPR-Cas-based molecular technologies in diagnosing several tropical diseases, namely malaria, zika, chikungunya, human immunodeficiency virus and acquired immunodeficiency syndrome (HIV-AIDS), tuberculosis (TB), and rabies.
Collapse
Affiliation(s)
- Md. Rashidur Rahman
- Department of Pharmacy, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Toma Rani Majumder
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| | - Md. Aminul Islam Apu
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
| | - Alok K. Paul
- Department of Biotechnology and Genetic Engineering, University of Development Alternative, Dhaka 1207, Bangladesh
| | - Afrina Afrose
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh
| | - Biplab Kumar Dash
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore 7408, Bangladesh
| |
Collapse
|
38
|
Alcalá AC, Contreras MA, Cuevas-Juárez E, Ramírez OT, Palomares LA. Effect of sericin, a silk derived protein, on the amplification of Zika virus in insect and mammalian cell cultures. J Biotechnol 2022; 353:28-35. [PMID: 35623476 DOI: 10.1016/j.jbiotec.2022.05.005] [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: 12/30/2021] [Revised: 04/15/2022] [Accepted: 05/13/2022] [Indexed: 02/07/2023]
Abstract
Sericin, a silk-derived non-immunogenic protein, has been used to improve cell culture performance by increasing viability, cell concentration, and promoting adherence of several cell lines. Here, we hypothesized that the properties of sericin can enhance the amplification of flaviviruses in cell cultures. The propagation of flavivirus is inefficient and limits scientific research. Zika virus (ZIKV) is an important human pathogen that has been widely studied because of its high impact on public health. There is a need to amplify Zika virus both for research and vaccine development. In this work, we show that sericin improves ZIKV amplification in insect (C6/36) and mammalian (Vero) cell cultures, and that it has a cryoprotectant capacity. Supplementation of cell culture media with sericin at 80 µg/mL resulted in a significant increase of 1 log in the concentration of ZIKV infectious particles produced from both cell lines. Furthermore, final virus yields increased between 5 and 10-fold in Vero cells and between 7 and 23-fold in C6/36 cells when sericin was supplemented, compared to control conditions. These results show that sericin is an effective supplement to increase ZIKV production by Vero and C6/36 cells. Additionally, sericin was a suitable cryoprotective agent, and hence an alternative to FBS and DMSO, for the cryopreservation of C6/36 cells but not for Vero cells.
Collapse
Affiliation(s)
- Ana C Alcalá
- Departamento de Medicina Molecular y Bioprocesosō, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Martha A Contreras
- Departamento de Medicina Molecular y Bioprocesosō, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Esmeralda Cuevas-Juárez
- Departamento de Medicina Molecular y Bioprocesosō, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Octavio T Ramírez
- Departamento de Medicina Molecular y Bioprocesosō, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México.
| | - Laura A Palomares
- Departamento de Medicina Molecular y Bioprocesosō, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México.
| |
Collapse
|
39
|
Tamkutė L, Haddad JG, Diotel N, Desprès P, Venskutonis PR, El Kalamouni C. Cranberry Pomace Extract Exerts Antiviral Activity against Zika and Dengue Virus at Safe Doses for Adult Zebrafish. Viruses 2022; 14:1101. [PMID: 35632841 PMCID: PMC9147401 DOI: 10.3390/v14051101] [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: 03/31/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
Mosquito-borne dengue virus (DENV) and zika virus (ZIKV) infections constitute a global health emergency. Antivirals directly targeting the virus infectious cycle are still needed to prevent dengue hemorrhagic fever and congenital zika syndrome. In the present study, we demonstrated that Cranberry Pomace (CP) extract, a polyphenol-rich agrifood byproduct recovered following cranberry juice extraction, blocks DENV and ZIKV infection in human Huh7.5 and A549 cell lines, respectively, in non-cytotoxic concentrations. Our virological assays identified CP extract as a potential inhibitor of virus entry into the host-cell by acting directly on viral particles, thus preventing their attachment to the cell surface. At effective antiviral doses, CP extract proved safe and tolerable in a zebrafish model. In conclusion, polyphenol-rich agrifood byproducts such as berry extracts are a promising source of safe and naturally derived nutraceutical antivirals that target medically important pathogens.
Collapse
Affiliation(s)
- Laura Tamkutė
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
- Department of Food Science and Technology, Kaunas University of Technology, Radvilenu, pl. 19, LT-50254 Kaunas, Lithuania;
| | - Juliano G. Haddad
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
| | - Nicolas Diotel
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, UMR 1188, 97490 Saint-Denis de La Réunion, France;
| | - Philippe Desprès
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
| | - Petras Rimantas Venskutonis
- Department of Food Science and Technology, Kaunas University of Technology, Radvilenu, pl. 19, LT-50254 Kaunas, Lithuania;
| | - Chaker El Kalamouni
- Unité Mixte Processus Infectieux En Milieu Insulaire Tropical, Université De La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (L.T.); (J.G.H.); (P.D.)
| |
Collapse
|
40
|
Curry K, Chandler R, Kostas-Polston EA, Alexander I, Orsega S, Johnson-Mallard V. Recommendations for managing sexually transmitted infections: Incorporating the 2021 guidelines. Nurse Pract 2022; 47:10-18. [PMID: 35349512 DOI: 10.1097/01.npr.0000822528.27483.b2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Sexually transmitted infections (STIs) are common and costly, with about 26 million STIs occurring each year in the US. Guidelines for the prevention and management of STIs are updated periodically. In 2021, the CDC updated its guidelines for the treatment of STIs. This article provides information on the most recent updates on managing STIs to help advanced practice nurses in their practice.
Collapse
|
41
|
Morbilliform Eruptions in the Hospitalized Child. Dermatol Clin 2022; 40:191-202. [PMID: 35366972 PMCID: PMC8896762 DOI: 10.1016/j.det.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
42
|
Rashid MU, Lao Y, Spicer V, Coombs KM. Zika Virus Infection of Sertoli Cells Alters Protein Expression Involved in Activated Immune and Antiviral Response Pathways, Carbohydrate Metabolism and Cardiovascular Disease. Viruses 2022; 14:v14020377. [PMID: 35215967 PMCID: PMC8878972 DOI: 10.3390/v14020377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 11/29/2022] Open
Abstract
Zika virus (ZIKV), a re-emerging virus, causes congenital brain abnormalities and Guillain–Barré syndrome. It is mainly transmitted by Aedes mosquitoes, but infections are also linked to sexual transmissions. Infectious ZIKV has been isolated, and viral RNA has been detected in semen over a year after the onset of initial symptoms, but the mode of long-term persistence is not yet understood. ZIKV can proliferate in human Sertoli cells (HSerC) for several weeks in vitro, suggesting that it might be a reservoir for persistent ZIKV infection. This study determined proteomic changes in HSerC during ZIKV infections by TMT-mass spectrometry analysis. Levels of 4416 unique Sertoli cell proteins were significantly altered at 3, 5, and 7 days after ZIKV infection. The significantly altered proteins include enzymes, transcription regulators, transporters, kinases, peptidases, transmembrane receptors, cytokines, ion channels, and growth factors. Many of these proteins are involved in pathways associated with antiviral response, antigen presentation, and immune cell activation. Several immune response pathway proteins were significantly activated during infection, e.g., interferon signaling, T cell receptor signaling, IL-8 signaling, and Th1 signaling. The altered protein levels were linked to predicted activation of immune response in HSerC, which was predicted to suppress ZIKV infection. ZIKV infection also affected the levels of critical regulators of gluconeogenesis and glycolysis pathways such as phosphoglycerate mutase, phosphoglycerate kinase, and enolase. Interestingly, many significantly altered proteins were associated with cardiac hypertrophy, which may induce heart failure in infected patients. In summary, our research contributes to a better understanding of ZIKV replication dynamics and infection in Sertoli cells.
Collapse
Affiliation(s)
- Mahamud-ur Rashid
- Department of Medical Microbiology and Infectious Diseases, The University of Manitoba, Room 543 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada;
- Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada; (Y.L.); (V.S.)
- Correspondence:
| | - Ying Lao
- Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada; (Y.L.); (V.S.)
| | - Victor Spicer
- Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada; (Y.L.); (V.S.)
| | - Kevin M. Coombs
- Department of Medical Microbiology and Infectious Diseases, The University of Manitoba, Room 543 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9, Canada;
- Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada; (Y.L.); (V.S.)
- Children’s Hospital Research Institute of Manitoba, Room 513, John Buhler Research Centre, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
43
|
Transcriptome Analysis of an Aedes albopictus Cell Line Single- and Dual-Infected with Lammi Virus and WNV. Int J Mol Sci 2022; 23:ijms23020875. [PMID: 35055061 PMCID: PMC8777793 DOI: 10.3390/ijms23020875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 11/27/2022] Open
Abstract
Understanding the flavivirus infection process in mosquito hosts is important and fundamental in the search for novel control strategies that target the mosquitoes’ ability to carry and transmit pathogenic arboviruses. A group of viruses known as insect-specific viruses (ISVs) has been shown to interfere with the infection and replication of a secondary arbovirus infection in mosquitoes and mosquito-derived cell lines. However, the molecular mechanisms behind this interference are unknown. Therefore, in the present study, we infected the Aedes albopictus cell line U4.4 with either the West Nile virus (WNV), the insect-specific Lammi virus (LamV) or an infection scheme whereby cells were pre-infected with LamV 24 h prior to WNV challenge. The qPCR analysis showed that the dual-infected U4.4 cells had a reduced number of WNV RNA copies compared to WNV-only infected cells. The transcriptome profiles of the different infection groups showed a variety of genes with altered expression. WNV-infected cells had an up-regulation of a broad range of immune-related genes, while in LamV-infected cells, many genes related to stress, such as different heat-shock proteins, were up-regulated. The transcriptome profile of the dual-infected cells was a mix of up- and down-regulated genes triggered by both viruses. Furthermore, we observed an up-regulation of signal peptidase complex (SPC) proteins in all infection groups. These SPC proteins have shown importance for flavivirus assembly and secretion and could be potential targets for gene modification in strategies for the interruption of flavivirus transmission by mosquitoes.
Collapse
|
44
|
Pereira SH, Almeida LT, Ferraz AC, Ladeira MDS, Ladeira LO, Magalhães CLDB, Silva BDM. Antioxidant and antiviral activity of fullerol against Zika virus. Acta Trop 2021; 224:106135. [PMID: 34536367 DOI: 10.1016/j.actatropica.2021.106135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 08/05/2021] [Accepted: 09/06/2021] [Indexed: 11/17/2022]
Abstract
Neglected for years, Zika virus (ZIKV) has become one of the most relevant arboviruses in current public health. The recent Zika fever epidemic in the Americas generated a worldwide alert due to the association with diseases such as Guillain-Barré syndrome and congenital syndromes. Among the pathogenesis of ZIKV, recent studies suggest that oxidative stress plays an important role during infection and that compounds capable of modulating oxidative stress are promising as therapeutics. Furthermore, so far there are no specific and efficient antiviral drug or vaccine available against ZIKV. Thus, fullerol was evaluated in the context of infection by ZIKV, since it is a carbon nanomaterial known for its potent antioxidant action. In this study, fullerol did not alter cell viability at the concentrations tested, proving to be inert, beyond to presenting high antioxidant power at low concentrations. ZIKV infection of human glioblastoma increased the production of reactive oxygen species by 60% and modulated the Nrf-2 pathway activity negatively. After treatment with fullerol, both conditions were restored to baseline levels. Additionally, fullerol was able to reduce viral production by up to 90%. Therefore, our results suggest that fullerol as a promising candidate in the control of ZIKV infections, presenting both antioxidant and antiviral action.
Collapse
Affiliation(s)
- Samille Henriques Pereira
- Laboratório de Biologia e Tecnologia de Micro-organismos, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil; Programa de Pós Graduação em Biotecnologia, Universidade Federal de Ouro Preto, Brazil
| | - Letícia Trindade Almeida
- Laboratório de Biologia e Tecnologia de Micro-organismos, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil; Programa de Pós Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil
| | - Ariane Coelho Ferraz
- Laboratório de Biologia e Tecnologia de Micro-organismos, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil; Programa de Pós Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil
| | | | | | - Cíntia Lopes de Brito Magalhães
- Laboratório de Biologia e Tecnologia de Micro-organismos, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil; Programa de Pós Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil
| | - Breno de Mello Silva
- Laboratório de Biologia e Tecnologia de Micro-organismos, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil; Programa de Pós Graduação em Biotecnologia, Universidade Federal de Ouro Preto, Brazil; Programa de Pós Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto, Brazil.
| |
Collapse
|
45
|
Qin Z, Chen Y, Yu J, He X, Liu X, Zhu L, Wu Q, Wan C, Zhang B, Zhao W. Rapid Construction of an Infectious Clone of the Zika Virus, Strain ZKC2. Front Microbiol 2021; 12:752578. [PMID: 34745057 PMCID: PMC8567171 DOI: 10.3389/fmicb.2021.752578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022] Open
Abstract
Zika virus (ZIKV) has had detrimental effects on global public health in recent years. This is because the management of the disease has been limited, in part because its pathogenic mechanisms are not yet completely understood. Infectious clones are an important tool that utilize reverse genetics; these can be used to modify the ZIKV genomic RNA at the DNA level. A homologous recombination clone was used to construct pWSK29, a low copy plasmid that contained sequences for a T7 promoter, the whole genome of ZIKV ZKC2 strain, and a hepatitis delta virus ribozyme. High fidelity PCR was then used to amplify the T7 transcription template. The transcript was then transfected into susceptible cells via lipofection to recover the ZIKV ZKC2 strain. Finally, the virulence of rZKC2 was evaluated both in vitro and in vivo. The rZKC2 was successfully obtained and it showed the same virulence as its parent, the ZIKV ZKC2 strain (pZKC2), both in vitro and in vivo. The 3730 (NS2A-D62G) mutation site was identified as being important, since it had significant impacts on rZKC2 recovery. The 4015 (NS2A, A157V) mutation may reduce virus production by increasing the interferon type I response. In this study, one of the earliest strains of ZIKV that was imported into China was used for infectious clone construction and one possible site for antiviral medication development was discovered. The use of homologous recombination clones, of PCR products as templates for T7 transcription, and of lipofection for large RNA transfection could increase the efficiency of infectious clone construction. Our infectious clone provides an effective tool which can be used to explore the life cycle and medical treatment of ZIKV.
Collapse
Affiliation(s)
- Zhiran Qin
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yangyang Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jianhai Yu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoen He
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xuling Liu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Li Zhu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qinghua Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Chengsong Wan
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Bao Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
46
|
Zou M, Li JY, Zhang MJ, Li JH, Huang JT, You PD, Liu SW, Zhou CQ. G-quadruplex binder pyridostatin as an effective multi-target ZIKV inhibitor. Int J Biol Macromol 2021; 190:178-188. [PMID: 34461156 DOI: 10.1016/j.ijbiomac.2021.08.121] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/13/2022]
Abstract
At present, there are still no anti-Zika virus (ZIKV) drugs or vaccines approved by FDA with accurate targets and antiviral mechanisms. Considering the RNA G-quadruplex sequences in ZIKV genome, it is very meaningful to develop G-quadruplex binders as potential anti-ZIKV drugs with novel and accurate targets. In this paper, five classical G-quadruplex binders including Ber, Braco 19, NiL, 360A and PDS have been chosen to discuss their interaction with ZIKV RNA G-quadruplexes. PDS shows higher binding affinity and thermal stability than the other G-quadruplex binders. This property is further evidenced in cells by immunofluorescence microscopy. And PDS shows higher anti-ZIKV activity (EC50 = 4.2 ± 0.4 μM) than the other G-quadruplex binders as well as the positive control ribavirin, with a low cytotoxicity. By time-of-addition assay, PDS exerts antiviral activity at the post-entry process of ZIKV replication cycle, thus inhibiting ZIKV mRNA replication and protein expression. Furthermore, PDS combines with ZIKV NS2B-NS3 protease and reduces its catalytic activity. This study suggests that G-quadruplex binder PDS is an effective multi-target ZIKV inhibitor, which provides more guidance to design some novel anti-ZIKV drugs targeting ZIKV RNA G-quadruplexes.
Collapse
Affiliation(s)
- Min Zou
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jing-Yan Li
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Meng-Jia Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jun-Hui Li
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jun-Tao Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Pei-Dan You
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Shu-Wen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| | - Chun-Qiong Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
47
|
Ferraris P, Wichit S, Cordel N, Missé D. Human host genetics and susceptibility to ZIKV infection. INFECTION GENETICS AND EVOLUTION 2021; 95:105066. [PMID: 34487865 DOI: 10.1016/j.meegid.2021.105066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 12/26/2022]
Abstract
Managing emerging infectious diseases is a current challenge in the fields of microbiology and epidemiology. Indeed, among other environmental and human-related factors, climate change and global warming favor the emergence of new pathogens. The recent Zika virus (ZIKV) epidemic, of which the large and rapid spread surprised the scientific community, is a reminder of the importance to study viruses currently responsible for sporadic infections. Increasing our knowledge of key factors involved in emerging infections is essential to implement specific monitoring that can be oriented according to the pathogen, targeted population, or at-risk environment. Recent technological developments, such as high-throughput sequencing, genome-wide association studies and CRISPR screenings have allowed the identification of human single nucleotide polymorphisms (SNPs) involved in infectious disease outcome. This review focuses on the human genetic host factors that have been identified and shown to be associated with the pathogenesis of ZIKV infection and candidate SNP targets.
Collapse
Affiliation(s)
- Pauline Ferraris
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France.
| | - Sineewanlaya Wichit
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Nadège Cordel
- Guadeloupe University Hospital, Department of Dermatology and Clinical Immunology, Pointe-à-Pitre, Guadeloupe and Normandie University, UNIROUEN, IRIB, Inserm, U1234, Rouen, France
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34394 Montpellier, France
| |
Collapse
|
48
|
Insights on Dengue and Zika NS5 RNA-dependent RNA polymerase (RdRp) inhibitors. Eur J Med Chem 2021; 224:113698. [PMID: 34274831 DOI: 10.1016/j.ejmech.2021.113698] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/20/2022]
Abstract
Over recent years, many outbreaks caused by (re)emerging RNA viruses have been reported worldwide, including life-threatening Flaviviruses, such as Dengue (DENV) and Zika (ZIKV). Currently, there is only one licensed vaccine against Dengue, Dengvaxia®. However, its administration is not recommended for children under nine years. Still, there are no specific inhibitors available to treat these infectious diseases. Among the flaviviral proteins, NS5 RNA-dependent RNA polymerase (RdRp) is a metalloenzyme essential for viral replication, suggesting that it is a promising macromolecular target since it has no human homolog. Nowadays, several NS5 RdRp inhibitors have been reported, while none inhibitors are currently in clinical development. In this context, this review constitutes a comprehensive work focused on RdRp inhibitors from natural, synthetic, and even repurposing sources. Furthermore, their main aspects associated with the structure-activity relationship (SAR), proposed mechanisms of action, computational studies, and other topics will be discussed in detail.
Collapse
|
49
|
Fernandes RS, de Godoy AS, Santos IA, Noske GD, de Oliveira KIZ, Gawriljuk VO, Gomes Jardim AC, Oliva G. Discovery of an imidazonaphthyridine and a riminophenazine as potent anti-Zika virus agents through a replicon-based high-throughput screening. Virus Res 2021; 299:198388. [PMID: 33887282 DOI: 10.1016/j.virusres.2021.198388] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/19/2021] [Accepted: 03/06/2021] [Indexed: 12/17/2022]
Abstract
The 2015/16 Zika virus (ZIKV) epidemic led to almost 1 million confirmed cases in 84 countries and was associated to the development of congenital microcephaly and Guillain-Barré syndrome. More recently, a ZIKV African lineage was identified in Brazil raising concerns about a future outbreak. The long-term consequences of viral infection emphasizes the need for the development of effective anti-ZIKV drugs. In this study, we developed and characterized a ZIKV replicon cell line for the screening of viral replication inhibitors. The replicon system was developed by engineering the IRES-Neo cassette into the 3' UTR terminus of the ZIKV Rluc DNA construct. After in vitro transcription, replicon RNA was used to transfect BHK-21 cells, that were selected with G418, thus generating the BHK-21-RepZIKV_IRES-Neo cell line. Through this replicon-based cell system, we identified two molecules with potent anti-ZIKV activities, an imidazonaphthyridine and a riminophenazine, both from the MMV/DNDi Pandemic Response Box library of 400 drug-like compounds. The imidazonaphthyridine, known as RO8191, showed remarkable selectivity against ZIKV, while the riminophenazine, the antibiotic Clofazimine, could act as a non-nucleoside analog inhibitor of viral RNA-dependent RNA polymerase (RdRp), as evidenced both in vitro and in silico. The data showed herein supports the use of replicon-based assays in high-throughput screening format as a biosafe and reliable tool for antiviral drug discovery.
Collapse
Affiliation(s)
- Rafaela Sachetto Fernandes
- Physics Institute of Sao Carlos, University of Sao Paulo, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, 13563-120, Brazil.
| | - Andre Schutzer de Godoy
- Physics Institute of Sao Carlos, University of Sao Paulo, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, 13563-120, Brazil
| | - Igor Andrade Santos
- Institute of Biomedical Sciences, Federal University of Uberlândia, Av. Amazonas, 1700, bloco 4C sala 216, Umuarama, Uberlândia, 38405-317, Brazil
| | - Gabriela Dias Noske
- Physics Institute of Sao Carlos, University of Sao Paulo, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, 13563-120, Brazil
| | - Ketllyn Irene Zagato de Oliveira
- Physics Institute of Sao Carlos, University of Sao Paulo, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, 13563-120, Brazil
| | - Victor Oliveira Gawriljuk
- Physics Institute of Sao Carlos, University of Sao Paulo, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, 13563-120, Brazil
| | - Ana Carolina Gomes Jardim
- Institute of Biomedical Sciences, Federal University of Uberlândia, Av. Amazonas, 1700, bloco 4C sala 216, Umuarama, Uberlândia, 38405-317, Brazil
| | - Glaucius Oliva
- Physics Institute of Sao Carlos, University of Sao Paulo, Av. Joao Dagnone, 1100, Jardim Santa Angelina, São Carlos, 13563-120, Brazil
| |
Collapse
|
50
|
Wichit S, Gumpangseth N, Hamel R, Yainoy S, Arikit S, Punsawad C, Missé D. Chikungunya and Zika Viruses: Co-Circulation and the Interplay between Viral Proteins and Host Factors. Pathogens 2021; 10:448. [PMID: 33918691 PMCID: PMC8068860 DOI: 10.3390/pathogens10040448] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Chikungunya and Zika viruses, both transmitted by mosquito vectors, have globally re-emerged over for the last 60 years and resulted in crucial social and economic concerns. Presently, there is no specific antiviral agent or vaccine against these debilitating viruses. Understanding viral-host interactions is needed to develop targeted therapeutics. However, there is presently limited information in this area. In this review, we start with the updated virology and replication cycle of each virus. Transmission by similar mosquito vectors, frequent co-circulation, and occurrence of co-infection are summarized. Finally, the targeted host proteins/factors used by the viruses are discussed. There is an urgent need to better understand the virus-host interactions that will facilitate antiviral drug development and thus reduce the global burden of infections caused by arboviruses.
Collapse
Affiliation(s)
- Sineewanlaya Wichit
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand; (N.G.); (S.Y.)
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand;
| | - Nuttamonpat Gumpangseth
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand; (N.G.); (S.Y.)
| | - Rodolphe Hamel
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France; (R.H.); (D.M.)
| | - Sakda Yainoy
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand; (N.G.); (S.Y.)
| | - Siwaret Arikit
- Department of Agronomy, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University Kamphaeng Saen Campus, Nakhon Pathom 73140, Thailand;
| | - Chuchard Punsawad
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand;
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France; (R.H.); (D.M.)
| |
Collapse
|