1
|
Ali MS, Mekonen EG. Yellow fever vaccine coverage and associated factors among under-five children in Kenya: Data from Kenyan Demographic and Health Survey 2022. Hum Vaccin Immunother 2024; 20:2391596. [PMID: 39165035 PMCID: PMC11340740 DOI: 10.1080/21645515.2024.2391596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024] Open
Abstract
Yellow fever is a vaccine preventable hemorrhagic disease that leads to morbidity and mortality in the affected individuals. The only options for preventing and controlling its spread are through vaccination. Therefore, this study was conducted to estimate yellow fever vaccination coverage and associated factors among under-five children in Kenya. The total weighted samples of 2,844 children aged under-five were included in this study. The data were taken from the Kenyan Demographic and Health Survey 2022. In the multivariable analysis, the adjusted odds ratio with a 95% CI was used to declare significant associations of yellow fever vaccine. The yellow fever vaccine coverage among children aged under-five in Kenya was 18.50%. The significant factors associated with yellow fever vaccine coverage were: the age of the child older than 24 months (AOR = 1.7; 95% CI (1.17-2.58)); higher odds of yellow fever vaccination coverage was observed among older children, place of residence (AOR = 1.76; 95% CI (1.04-2.97)); higher odds was observed among urban residents, maternal education; primary education (AOR = 1.99; 95% CI (1.04-2.97)), secondary education (AOR = 2.85; 95% CI (1.41-5.76)), mothers who attended primary or secondary education have higher odds of yellow fever vaccination coverage, wealth index (AOR = 2.38; 95% CI (1.15-4.91)); higher odds of vaccination coverage was observed among poor households. Yellow fever vaccine coverage among under-five children in Kenya was low and has become an important public health concern. Policymakers and other stakeholders are recommended to focus on vaccination programs to prevent yellow fever disease.
Collapse
Affiliation(s)
- Mohammed Seid Ali
- Department of Pediatrics and Child Health Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Enyew Getaneh Mekonen
- Department of Surgical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
2
|
Lani R, Thariq IM, Suhaimi NS, Hassandarvish P, Abu Bakar S. From defense to offense: Modulating toll-like receptors to combat arbovirus infections. Hum Vaccin Immunother 2024; 20:2306675. [PMID: 38263674 DOI: 10.1080/21645515.2024.2306675] [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: 09/08/2023] [Accepted: 01/14/2024] [Indexed: 01/25/2024] Open
Abstract
Arboviruses are a significant threat to global public health, with outbreaks occurring worldwide. Toll-like receptors (TLRs) play a crucial role in the innate immune response against these viruses by recognizing pathogen-associated molecular patterns and initiating an inflammatory response. Significantly, TLRs commonly implicated in the immune response against viral infections include TLR2, TLR4, TLR6, TLR3, TLR7, and TLR8; limiting or allowing them to replicate and spread within the host. Modulating TLRs has emerged as a promising approach to combat arbovirus infections. This review summarizes recent advances in TLR modulation as a therapeutic target in arbovirus infections. Studies have shown that the activation of TLRs can enhance the immune response against arbovirus infections, leading to increased viral clearance and protection against disease. Conversely, inhibition of TLRs can reduce the excessive inflammation and tissue damage associated with arbovirus infection. Modulating TLRs represents a potential therapeutic strategy to combat arbovirus infections.
Collapse
Affiliation(s)
- Rafidah Lani
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ilya Maisarah Thariq
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Nuramira Syazreen Suhaimi
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Pouya Hassandarvish
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Sazaly Abu Bakar
- Tropical Infectious Diseases Research and Education Centre, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
de Sousa FTG, Warnes CM, Manuli ER, Tjang LV, Carneiro PH, Maria de Oliveira Pinto L, Ng A, Bhat S, Zambrana JV, D'Elia Zanella LGFAB, Ho YL, Romano CM, Beatty PR, Biering SB, Kallas EG, Sabino EC, Harris E. Yellow fever disease severity and endothelial dysfunction are associated with elevated serum levels of viral NS1 protein and syndecan-1. EBioMedicine 2024; 109:105409. [PMID: 39454515 DOI: 10.1016/j.ebiom.2024.105409] [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: 02/11/2023] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Yellow fever virus (YFV) infections are a major global disease concern with high mortality in humans, and as such it is critical to identify clinical correlates of disease severity. While nonstructural protein 1 (NS1) of the related dengue virus is implicated in contributing to vascular leak, little is known about the role of YFV NS1 in severe YF and mechanisms of vascular dysfunction in YFV infections. METHODS Using serum samples from laboratory-confirmed YF patients with severe (n = 39) or non-severe (n = 18) disease in a well-defined hospital observational cohort in Brazil, plus samples from healthy uninfected controls (n = 11), we investigated factors associated with disease severity and endothelial dysfunction. FINDINGS We found significantly increased levels of NS1, as well as syndecan-1, a marker of vascular leak, in serum from severe YF as compared to non-severe YF or control groups. We also showed that hyperpermeability of endothelial cell monolayers treated with serum from severe YF patients was significantly higher compared to non-severe YF and control groups, as measured by transendothelial electrical resistance (TEER). Further, we demonstrated that YFV NS1 induces shedding of syndecan-1 from the surface of human endothelial cells. Notably, YFV NS1 serum levels significantly correlated with syndecan-1 serum levels, TEER values, and signs of disease severity. Syndecan-1 levels also significantly correlated with clinical laboratory parameters of disease severity, viral load, hospitalization, and death. INTERPRETATION This study provides further evidence for endothelial dysfunction as a mechanism of YF pathogenesis in humans and suggests serum quantification of YFV NS1 and syndecan-1 as valuable tools for disease diagnosis and/or prognosis. FUNDING This work was supported by the US NIH and FAPESP.
Collapse
Affiliation(s)
- Francielle T G de Sousa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA; Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 05403000, Brazil; Laboratório de Investigação Médica, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil.
| | - Colin M Warnes
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Erika R Manuli
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 05403000, Brazil; Laboratório de Investigação Médica, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil
| | - Laurentia V Tjang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Pedro H Carneiro
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA.
| | - Luzia Maria de Oliveira Pinto
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA; Laboratório das Interações Vírus-Hospedeiros (LIVH), Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil.
| | - Arash Ng
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720-3200, USA.
| | - Samhita Bhat
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Jose Victor Zambrana
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Luiz G F A B D'Elia Zanella
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil
| | - Yeh-Li Ho
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil.
| | - Camila M Romano
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 05403000, Brazil; Laboratório de Investigação Médica, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil.
| | - P Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA; Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720-3200, USA.
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Esper G Kallas
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 05403000, Brazil; Laboratório de Investigação Médica, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil; Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil.
| | - Ester C Sabino
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 05403000, Brazil; Laboratório de Investigação Médica, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP 05403000, Brazil.
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA; Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720-3200, USA.
| |
Collapse
|
4
|
Liu Y, Xu M, Xia B, Qiao Z, He Y, Liu Y, Pan Z, Zhang C, Peng H, Liang X, Zhao P, Tang H, Zheng X. Nifuroxazide Prevents Chikungunya Virus Infection Both In Vitro and In Vivo via Suppressing Viral Replication. Viruses 2024; 16:1322. [PMID: 39205296 PMCID: PMC11360488 DOI: 10.3390/v16081322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging arbovirus causing disease on a global scale, and the potential for its epidemics remains high. CHIKV has caused millions of cases and heavy economic burdens around the world, while there are no available approved antiviral therapies to date. In this study, nifuroxazide, an FDA-approved antibiotic for acute diarrhea or colitis, was found to significantly inhibit a variety of arboviruses, although its antiviral activity varied among different target cell types. Nifuroxazide exhibited relatively high inhibitory efficiency in yellow fever virus (YFV) infection of the hepatoma cell line Huh7, tick-borne encephalitis virus (TBEV) and west nile virus (WNV) infection of the vascular endothelial cell line HUVEC, and CHIKV infection of both Huh7 cells and HUVECs, while it barely affected the viral invasion of neurons. Further systematic studies on the action stage of nifuroxazide showed that nifuroxazide mainly inhibited in the viral replication stage. In vivo, nifuroxazide significantly reduced the viral load in muscles and protected mice from CHIKV-induced footpad swelling, an inflammation injury within the arthrosis of infected mice. These results suggest that nifuroxazide has a potential clinical application as an antiviral drug, such as in the treatment of CHIKV infection.
Collapse
Affiliation(s)
- Yangang Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Mingxiao Xu
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai 200433, China
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Zhuoyue Qiao
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, China
| | - Yanhua He
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Yan Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Zhendong Pan
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Congcong Zhang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Haoran Peng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Xuesong Liang
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai 200433, China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Xu Zheng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
5
|
Zhu F, Sun MX, Zhao SQ, Qin CF, Wang JH, Deng YQ. Immunogenicity and Protective Efficacy of Aerosolized Live-Attenuated Yellow Fever 17D Vaccine in Mice. Vaccines (Basel) 2024; 12:856. [PMID: 39203982 PMCID: PMC11360090 DOI: 10.3390/vaccines12080856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Yellow fever (YF), caused by the yellow fever virus (YFV), continually spreads and causes epidemics worldwide, posing a great threat to human health. The live-attenuated YF 17D vaccine (YF-17D) has been licensed for preventing YFV infection and administrated via the intramuscular (i.m.) route. In this study, we sought to determine the immunogenicity and protective efficacy of aerosolized YF-17D via the intratracheal (i.t.) route in mice. YF-17D stocks in liquids were successfully aerosolized into particles of 6 μm. Further in vitro phenotype results showed the aerosolization process did not abolish the infectivity of YF-17D. Meanwhile, a single i.t. immunization with aerosolized YF-17D induced robust humoral and cellular immune responses in A129 mice, which is comparable to that received i.p. immunization. Notably, the aerosolized YF-17D also triggered specific secretory IgA (SIgA) production in bronchoalveolar lavage. Additionally, all immunized animals survived a lethal dose of YFV challenge in mice. In conclusion, our results support further development of aerosolized YF-17D in the future.
Collapse
Affiliation(s)
- Feng Zhu
- School of Life Sciences, Southwest Forestry University, Kunming 650224, China;
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (M.-X.S.); (S.-Q.Z.); (C.-F.Q.)
| | - Meng-Xu Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (M.-X.S.); (S.-Q.Z.); (C.-F.Q.)
| | - Suo-Qun Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (M.-X.S.); (S.-Q.Z.); (C.-F.Q.)
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (M.-X.S.); (S.-Q.Z.); (C.-F.Q.)
| | - Jin-Hua Wang
- School of Life Sciences, Southwest Forestry University, Kunming 650224, China;
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China; (M.-X.S.); (S.-Q.Z.); (C.-F.Q.)
| |
Collapse
|
6
|
Owusu-Akyaw M, Owusu-Asenso CM, Abdulai A, Mohammed AR, Sraku IK, Boadu EN, Aduhene E, Attah SK, Afrane YA. Risk of arboviral transmission and insecticide resistance status of Aedes mosquitoes during a yellow fever outbreak in Ghana. BMC Infect Dis 2024; 24:731. [PMID: 39054464 PMCID: PMC11270840 DOI: 10.1186/s12879-024-09643-z] [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: 05/13/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND In late 2021, Ghana was hit by a Yellow Fever outbreak that started in two districts in the Savannah region and spread to several other Districts in three regions. Yellow fever is endemic in Ghana. However, there is currently no structured vector control programme for Aedes the arboviral vector in Ghana. Knowledge of Aedes bionomics and insecticide susceptibility status is important to control the vectors. This study therefore sought to determine Aedes vector bionomics and their insecticide resistance status during a yellow fever outbreak. METHODS The study was performed in two yellow fever outbreak sites (Wenchi, Larabanga) and two non-outbreak sites (Kpalsogu, Pagaza) in Ghana. Immature Aedes mosquitoes were sampled from water-holding containers in and around human habitations. The risk of disease transmission was determined in each site using stegomyia indices. Adult Aedes mosquitoes were sampled using Biogents Sentinel (BG) traps, Human Landing Catch (HLC), and Prokopack (PPK) aspirators. Phenotypic resistance to permethrin, deltamethrin and pirimiphos-methyl was determined with WHO susceptibility tests using Aedes mosquitoes collected as larvae and reared into adults. Knockdown resistance (kdr) mutations were detected using allele-specific multiplex PCR. RESULTS Among the 2,664 immature Aedes sampled, more than 60% were found in car tyres. Larabanga, an outbreak site, was classified as a high-risk zone for the Yellow Fever outbreak (BI: 84%, CI: 26.4%). Out of 1,507 adult Aedes mosquitoes collected, Aedes aegypti was the predominant vector species (92%). A significantly high abundance of Aedes mosquitoes was observed during the dry season (61.2%) and outdoors (60.6%) (P < 0.001). Moderate to high resistance to deltamethrin was observed in all sites (33.75% to 70%). Moderate resistance to pirimiphos-methyl (65%) was observed in Kpalsogu. Aedes mosquitoes from Larabanga were susceptible (98%) to permethrin. The F1534C kdr, V1016I kdr and V410 kdr alleles were present in all the sites with frequencies between (0.05-0.92). The outbreak sites had significantly higher allele frequencies of F1534C and V1016I respectively compared to non-outbreak sites (P < 0.001). CONCLUSION This study indicates that Aedes mosquitoes in Ghana pose a significant risk to public health. Hence there is a need to continue monitoring these vectors to develop an effective control strategy.
Collapse
Affiliation(s)
- Margaret Owusu-Akyaw
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Christopher Mfum Owusu-Asenso
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Anisa Abdulai
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Abdul Rahim Mohammed
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Isaac Kwame Sraku
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Emmanuel Nana Boadu
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Evans Aduhene
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Simon Kwaku Attah
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Yaw Asare Afrane
- Centre for Vector-Borne Disease Research, Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana.
| |
Collapse
|
7
|
Wanyana MW, King P, Migisha R, Kwesiga B, Okello PE, Kadobera D, Bulage L, Kayiwa J, Nankya AM, Ario AR, Harris JR. Evaluation of the sentinel yellow fever surveillance system in Uganda, 2017-2022: strengths and weaknesses. BMC Infect Dis 2024; 24:686. [PMID: 38982363 PMCID: PMC11234539 DOI: 10.1186/s12879-024-09580-x] [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: 11/30/2023] [Accepted: 07/01/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Uganda has a sentinel surveillance system in seven high-risk sites to monitor yellow fever (YF) patterns and detect outbreaks. We evaluated the performance of this system from 2017 to 2022. METHODS We evaluated selected attributes, including timeliness (lags between different critical time points), external completeness (proportion of expected sentinel sites reporting ≥ 1 suspect case in the system annually), and internal completeness (proportion of reports with the minimum required data elements filled), using secondary data in the YF surveillance database from January 2017-July 2022. We conducted key informant interviews with stakeholders at health facility and national level to assess usefulness, flexibility, simplicity, and acceptability of the surveillance system. RESULTS In total, 3,073 suspected and 15 confirmed YF cases were reported. The median time lag from sample collection to laboratory shipment was 37 days (IQR:21-54). External completeness was 76%; internal completeness was 65%. Stakeholders felt that the surveillance system was simple and acceptable, but were uncertain about flexibility. Most (71%) YF cases in previous outbreaks were detected through the sentinel surveillance system; data were used to inform interventions such as intensified YF vaccination. CONCLUSION The YF sentinel surveillance system was useful in detecting outbreaks and informing public health action. Delays in case confirmation and incomplete data compromised its overall effectiveness and efficiency.
Collapse
Affiliation(s)
- Mercy Wendy Wanyana
- Uganda Public Health Fellowship Program, Uganda National Institute of Public Health, Kampala, Uganda.
| | - Patrick King
- Uganda Public Health Fellowship Program, Uganda National Institute of Public Health, Kampala, Uganda
| | - Richard Migisha
- Uganda Public Health Fellowship Program, Uganda National Institute of Public Health, Kampala, Uganda
| | - Benon Kwesiga
- Uganda Public Health Fellowship Program, Uganda National Institute of Public Health, Kampala, Uganda
| | - Paul Edward Okello
- Uganda Public Health Fellowship Program, Uganda National Institute of Public Health, Kampala, Uganda
| | - Daniel Kadobera
- Uganda Public Health Fellowship Program, Uganda National Institute of Public Health, Kampala, Uganda
| | - Lilian Bulage
- Uganda Public Health Fellowship Program, Uganda National Institute of Public Health, Kampala, Uganda
| | - Joshua Kayiwa
- Ministry of Health, Uganda National Institute of Public Health, Kampala, Uganda
| | | | - Alex Riolexus Ario
- Ministry of Health, Uganda National Institute of Public Health, Kampala, Uganda
| | - Julie R Harris
- US Centers for Disease Control and Prevention, Kampala, Uganda
| |
Collapse
|
8
|
Nahain AA, Li J, Modhiran N, Watterson D, Li JP, Ignjatovic V, Monagle P, Tsanaktsidis J, Vamvounis G, Ferro V. Antiviral Activities of Heparan Sulfate Mimetic RAFT Polymers Against Mosquito-borne Viruses. ACS APPLIED BIO MATERIALS 2024; 7:2862-2871. [PMID: 38699864 DOI: 10.1021/acsabm.3c01223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Mosquito-borne viruses are a major worldwide health problem associated with high morbidity and mortality rates and significant impacts on national healthcare budgets. The development of antiviral drugs for both the treatment and prophylaxis of these diseases is thus of considerable importance. To address the need for therapeutics with antiviral activity, a library of heparan sulfate mimetic polymers was screened against dengue virus (DENV), Yellow fever virus (YFV), Zika virus (ZIKV), and Ross River virus (RRV). The polymers were prepared by RAFT polymerization of various acidic monomers with a target MW of 20 kDa (average Mn ∼ 27 kDa by GPC). Among the polymers, poly(SS), a homopolymer of sodium styrenesulfonate, was identified as a broad spectrum antiviral with activity against all the tested viruses and particularly potent inhibition of YFV (IC50 = 310 pM). Our results further uncovered that poly(SS) exhibited a robust inhibition of ZIKV infection in both mosquito and human cell lines, which points out the potential functions of poly(SS) in preventing mosquito-borne viruses associated diseases by blocking viral transmission in their mosquito vectors and mitigating viral infection in patients.
Collapse
Affiliation(s)
- Abdullah Al Nahain
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jinlin Li
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, University of Uppsala, 75123 Uppsala, Sweden
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, University of Uppsala, 75123 Uppsala, Sweden
| | - Vera Ignjatovic
- Haematology Research, Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Paul Monagle
- Haematology Research, Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria 3052, Australia
- Department of Clinical Haematology, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - John Tsanaktsidis
- CSIRO Manufacturing, Research Way, Clayton, Victoria 3168, Australia
| | - George Vamvounis
- College of Science and Engineering, James Cook University, Townsville, QLD 4811, Australia
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
9
|
Owusu-Akyaw M, Owusu-Asenso CM, Abdulai A, Mohammed AR, Sraku IK, Boadu EN, Aduhene E, Attah SK, Afrane YA. Risk of Arboviral Transmission and Insecticide Resistance Status of Aedes Mosquitoes during a Yellow Fever Outbreak in Ghana. RESEARCH SQUARE 2024:rs.3.rs-4271509. [PMID: 38699327 PMCID: PMC11065086 DOI: 10.21203/rs.3.rs-4271509/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Background In late 2021, Ghana was hit by a Yellow Fever outbreak that started in two (2) districts in the Savannah region and spread to several other Districts in (3) regions (Oti, Bono and Upper West).Yellow fever is endemic in Ghana. However, there is currently no structured vector control programme for the yellow vector, Aedes mosquitoes in Ghana. Knowledge of Aedes bionomics and insecticide susceptibility status is important to control the vectors. This study therefore sought todetermine Aedes vector bionomics and their insecticide resistance status during a yellow fever outbreak. Methods The study was performed in two yellow fever outbreak sites (Wenchi, Larabanga) and two non-outbreak sites (Kpalsogu, Pagaza) in Ghana. Immature Aedes mosquitoes were sampled from water-holding containers in and around human habitations. The risk of disease transmission was determined in each site using stegomyia indices. Adult Aedes mosquitoes were sampled using Biogents Sentinel (BG) traps, Human Landing Catch (HLC), and Prokopack (PPK) aspirators. Phenotypic resistance was determined with WHO susceptibility tests using Aedes mosquitoes collected as larvae and reared into adults. Knockdown resistance (kdr) mutations were detected using allele-specific multiplex PCR. Results Of the 2,664 immature Aedes sampled, more than 60% were found in car tyres. Larabanga, an outbreak site, was classified as a high-risk zone for the Yellow Fever outbreak (BI: 84%, CI: 26.4%). Out of 1,507 adult Aedes mosquitoes collected, Aedes aegypti was the predominant vector species (92%). A significantly high abundance of Aedes mosquitoes was observed during the dry season (61.2%) and outdoors (60.6%) (P < 0.001). Moderate to high resistance to deltamethrin was observed in all sites (33.75% to 70%). Moderate resistance to pirimiphos-methyl (65%) was observed in Kpalsogu. Aedesmosquitoes from Larabanga were susceptible (98%) to permethrin. The F1534C kdr, V1016I kdr and V410 kdr alleles were present in all the sites with frequencies between (0.05-0.92). The outbreak sites had significantly higher allele frequencies of F1534C and V1016I respectively compared to non-outbreak sites (P < 0.001). Conclusion This study indicates that Aedes mosquitoes in Ghana pose a significant risk to public health, and there is a need for continuous surveillance to inform effective vector control strategies.
Collapse
Affiliation(s)
- Margaret Owusu-Akyaw
- Department of Medical Microbiology, Centre for Vector-Borne Disease Research, University of Ghana
| | | | - Anisa Abdulai
- Department of Medical Micro biology, Centre for Vector-Borne Disease Research, University of Ghana
| | - Abdul Rahim Mohammed
- Department of Medical Microbiology, Centre for Vector-Borne Disease Research, University of Ghana
| | - Isaac Kwame Sraku
- Department of Medical Microbiology, Centre for Vector-Borne Disease Research, University of Ghana
| | - Emmanuel Nana Boadu
- Department of Medical Microbiology, Centre for Vector-Borne Disease Research, University of Ghana
| | - Evans Aduhene
- Department of Medical Microbiology, Centre for Vector-Borne Disease Research, University of Ghana
| | - Simon Kwaku Attah
- Department of Medical Microbiology, Centre for Vector-Borne Disease Research, University of Ghana
| | - Yaw Asare Afrane
- Department of Medical Microbiology, Centre for Vector-Borne Disease Research, University of Ghana
| |
Collapse
|
10
|
Ojianwuna CC, Enwemiwe VN, Egwunyenga AO, Agboro A, Owobu E. Sampling efficiency and screening of Aedes albopictus for yellow fever virus in Niger Delta region of Nigeria. Pan Afr Med J 2024; 47:120. [PMID: 38828420 PMCID: PMC11143074 DOI: 10.11604/pamj.2024.47.120.39462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/12/2024] [Indexed: 06/05/2024] Open
Abstract
Introduction Aedes albopictus, like Aedes aegypti, is a virulent vector of arboviruses especially the well-documented spread of yellow fever around the world. Although yellow fever is prevalent in Nigeria, there is a paucity of information in the Niger Delta region on the distribution of Aedes mosquito vectors and molecular detection of the virus in infected mosquitoes. This study sampled Aedes mosquitoes around houses associated with farms from four communities (Otolokpo, Ute-Okpu, Umunede, and Ute Alohen) in Ika North-East Local Government Area of Delta State, Nigeria. Methods various sampling methods were used in Aedes mosquito collection to test their efficacy in the survey. Mosquitoes in holding cages were killed by freezing and morphologically identified. A pool of 15 mosquitoes per Eppendorf tube was preserved in RNAi later for yellow fever virus screening. Two samples were molecularly screened for each location. Results seven hundred and twenty-five (725) mosquitoes were obtained from the various traps. The mean abundance of the mosquitoes was highest in m-HLC (42.9) compared to the mosquitoes sampled using other techniques (p<0.0001). The mean abundance of mosquitoes was lowest in Center for Disease Control (CDC) light traps without attractant (0.29). No yellow fever virus strain was detected in all the mosquitoes sampled at the four locations. Conclusion this study suggests that Aedes albopictus are the mosquitoes commonly biting around houses associated with farms. More so, yellow fever virus was not detected in the mosquitoes probably due to the mass vaccination exercise that was carried out the previous year in the study area. More studies are required using the m-HLC to determine the infection rate in this endemic area.
Collapse
Affiliation(s)
- Chioma Cynthia Ojianwuna
- Department of Animal and Environmental Biology, Faculty of Science, Delta State University, Abraka, Nigeria
| | - Victor Ngozi Enwemiwe
- Department of Animal and Environmental Biology, Faculty of Science, Delta State University, Abraka, Nigeria
| | - Andy Ogochukwu Egwunyenga
- Department of Animal and Environmental Biology, Faculty of Science, Delta State University, Abraka, Nigeria
| | - Akwilla Agboro
- Department of Animal and Environmental Biology, Faculty of Science, Delta State University, Abraka, Nigeria
| | - Emmanuel Owobu
- Department of Animal and Environmental Biology, Faculty of Science, Delta State University, Abraka, Nigeria
| |
Collapse
|
11
|
Yang TH, Gao WC, Ma X, Liu Q, Pang PP, Zheng YT, Jia Y, Zheng CB. A Review on The Pathogenesis of Cardiovascular Disease of Flaviviridea Viruses Infection. Viruses 2024; 16:365. [PMID: 38543730 PMCID: PMC10974792 DOI: 10.3390/v16030365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 05/23/2024] Open
Abstract
Members of the Flaviviridae family, encompassing the Flavivirus and Hepacivirus genera, are implicated in a spectrum of severe human pathologies. These diseases span a diverse spectrum, including hepatitis, vascular shock syndrome, encephalitis, acute flaccid paralysis, and adverse fetal outcomes, such as congenital heart defects and increased mortality rates. Notably, infections by Flaviviridae viruses have been associated with substantial cardiovascular compromise, yet the exploration into the attendant cardiovascular sequelae and underlying mechanisms remains relatively underexplored. This review aims to explore the epidemiology of Flaviviridae virus infections and synthesize their cardiovascular morbidities. Leveraging current research trajectories and our investigative contributions, we aspire to construct a cogent theoretical framework elucidating the pathogenesis of Flaviviridae-induced cardiovascular injury and illuminate prospective therapeutic avenues.
Collapse
Affiliation(s)
- Tie-Hua Yang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China; (T.-H.Y.); (P.-P.P.)
- Key Laboratory of Animal Models and Human Diseases Mechanisms of Chinese Academy of Sciences, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (X.M.); (Y.-T.Z.)
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Wen-Cong Gao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China;
| | - Xin Ma
- Key Laboratory of Animal Models and Human Diseases Mechanisms of Chinese Academy of Sciences, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (X.M.); (Y.-T.Z.)
- College of Modern Biomedical Industry, Kunming Medical University, Kunming 650500, China
| | - Qian Liu
- School of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Pan-Pan Pang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China; (T.-H.Y.); (P.-P.P.)
- Key Laboratory of Animal Models and Human Diseases Mechanisms of Chinese Academy of Sciences, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (X.M.); (Y.-T.Z.)
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Diseases Mechanisms of Chinese Academy of Sciences, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (X.M.); (Y.-T.Z.)
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Yinnong Jia
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China;
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China;
- College of Modern Biomedical Industry, Kunming Medical University, Kunming 650500, China
- Yunnan Vaccine Laboratory, Kunming 650500, China
| |
Collapse
|
12
|
Viral agents (2nd section). Transfusion 2024; 64 Suppl 1:S19-S207. [PMID: 38394038 DOI: 10.1111/trf.17630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 12/02/2023] [Indexed: 02/25/2024]
|
13
|
Chilakam N, Lakshminarayanan V, Keremutt S, Rajendran A, Thunga G, Poojari PG, Rashid M, Mukherjee N, Bhattacharya P, John D. Economic Burden of Mosquito-Borne Diseases in Low- and Middle-Income Countries: Protocol for a Systematic Review. JMIR Res Protoc 2023; 12:e50985. [PMID: 38079215 PMCID: PMC10750235 DOI: 10.2196/50985] [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: 07/24/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Globally, among all the vector-borne diseases, mosquito-borne diseases are responsible for a substantial number of cases and deaths and amount to an economic cost of US $12 billion per year. However, there is a dearth of systematic research conducted on the economic burden of mosquito-borne diseases. To address the lack of comprehensive information on this topic, a systematic review will be conducted to synthesize evidence for informing targeted policies and strategies addressing this growing burden and for better financial protection of households. OBJECTIVE The systematic review aims to review the economic burden of mosquito-borne diseases in low- and middle-income countries (LMICs). The review estimates the total cost, which is the compilation of both the direct costs and indirect costs. Additionally, it reports cost estimates per disease, country, and patient. The review outcome will also discuss the impact of the economic burden in terms of out-of-pocket expenditure, catastrophic health expenditure, impoverishment, and gross domestic product impact due to mosquito-borne diseases in LMICs. METHODS Systematic searches will be conducted in PubMed (MEDLINE), Ovid Embase, Scopus, the cumulative index of nursing and allied health literacy, and Cochrane CENTRAL. Additionally, websites of the World Bank, World Health Organization, and Asian Development Bank as well as grey literature (eg, Malaria No More and the Ministry of Health websites) will be searched to gather comprehensive information on the topic and identify studies published in the English language. The titles and abstracts will be independently screened by 2 reviewers, followed by a full-text review against the inclusion criteria. Disagreements will be resolved through discussion with a third author. The methodological reporting quality of the studies will be evaluated using the Larg and Moss checklist, Cochrane risk-of-bias tool for randomized trials, and the Consensus on Health Economic Criteria. Data will be extracted using a standardized data extraction form. RESULTS The protocol was registered in PROSPERO (CRD42023427111) prior to the initiation of the search strategy. The review is currently ongoing and will synthesize information from the identified studies through a process involving structured screening, data extraction, and critical appraisal in the form of tables and a narrative summary of studies reporting the economic burden incurred due to mosquito-borne diseases in LMICs. CONCLUSIONS This review seeks to report the economic burden of mosquito-borne diseases. It will act as evidence for policymakers to prioritize their decisions regarding containing the prevalence of mosquito-borne diseases and the means to lowering the incidence of diseases spread by mosquitoes. TRIAL REGISTRATION PROSPERO CRD42023427111; https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=427111. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/50985.
Collapse
Affiliation(s)
- Nagavalli Chilakam
- Department of Commerce, Manipal Academy of Higher Education, Manipal, India
| | | | - Sushanth Keremutt
- Department of Commerce, Manipal Academy of Higher Education, Manipal, India
| | - Ambigai Rajendran
- Department of Commerce, Manipal Academy of Higher Education, Manipal, India
| | - Girish Thunga
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Pooja Gopal Poojari
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Muhammed Rashid
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Nirmalya Mukherjee
- Centre for Public Health Research, Manbhum Ananda Ashram Nityananda Trust, Kolkata, India
| | - Paramita Bhattacharya
- Centre for Public Health Research, Manbhum Ananda Ashram Nityananda Trust, Kolkata, India
| | - Denny John
- Centre for Public Health Research, Manbhum Ananda Ashram Nityananda Trust, Kolkata, India
- Faculty of Life and Allied Health Sciences, MS Ramaiah University of Applied Sciences, Bengaluru, India
- School of Public Health, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
14
|
Sura K, Rohilla H, Kumar D, Jakhar R, Ahlawat V, Kaushik D, Dangi M, Chhillar AK. Exploring structural antigens of yellow fever virus to design multi-epitope subunit vaccine candidate by utilizing an immuno-informatics approach. J Genet Eng Biotechnol 2023; 21:161. [PMID: 38051433 DOI: 10.1186/s43141-023-00621-7] [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: 04/21/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Yellow fever is a mosquito-borne viral hemorrhagic disease transmitted by several species of virus-infected mosquitoes endemic to tropical regions of Central and South America and Africa. Earlier in the twentieth century, mass vaccination integrated with mosquito control was implemented to eradicate the yellow fever virus. However, regular outbreaks occur in these regions which pose a threat to travelers and residents of Africa and South America. There is no specific antiviral therapy, but there can be an effective peptide-based vaccine candidate to combat infection caused by the virus. Therefore, the study aims to design a multi-epitope-based subunit vaccine (MESV) construct against the yellow fever virus to reduce the time and cost using reverse vaccinology (RV) approach. METHODS Yellow fever virus contains 10,233 nucleotides that encode for 10 proteins (C, prM, E, NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) including 3 structural and 7 non-structural proteins. Structural proteins-precursor membrane protein (prM) and envelope protein (E)-were taken as a target for B cell and T cell epitope screening. Further, various immunoinformatics approaches were employed to FASTA sequences of structural proteins to retrieve B cell and T cell epitopes. MESV was constructed from these epitopes based on allergenicity, antigenicity and immunogenicity, toxicity, conservancy, and population coverage followed by structure prediction. The efficacy of the MESV construct to bind with human TLR-3, TLR-4, and TLR-8 were evaluated using molecular docking and simulation studies. Finally, in-silico cloning of vaccine construct was performed withpBR322 Escherichia coli expression system using codon optimization. RESULTS Predicted epitopes evaluated and selected for MESV construction were found stable, non-allergenic, highly antigenic, and global population coverage of 68.03% according to in-silico analysis. However, this can be further tested in in-vitro and in-vivo investigations. Epitopes were sequentially merged to construct a MESV consisting of 393 amino acids using adjuvant and linkers. Molecular docking and simulation studies revealed stable and high-affinity interactions. Furthermore, in-silico immune response graphs showed effective immune response generation. Finally, higher CAI value ensured high gene expression of vaccine in the host cell. CONCLUSION The designed MESV construct in the present in-silico study can be effective in generating an immune response against the yellow fever virus. Therefore, to prevent yellow fever, it can be an effective vaccine candidate. However, further downstream, in-vitro study is required.
Collapse
Affiliation(s)
- Kiran Sura
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Himanshi Rohilla
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Dev Kumar
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Ritu Jakhar
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Vaishali Ahlawat
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
- Centre for Biotechnology, M.D. University, Rohtak, Haryana, India
| | | | - Mehak Dangi
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India.
| | | |
Collapse
|
15
|
Ramezannia Z, Shamekh A, Bannazadeh Baghi H. CRISPR-Cas system to discover host-virus interactions in Flaviviridae. Virol J 2023; 20:247. [PMID: 37891676 PMCID: PMC10605781 DOI: 10.1186/s12985-023-02216-7] [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: 03/10/2023] [Accepted: 10/25/2023] [Indexed: 10/29/2023] Open
Abstract
The Flaviviridae virus family members cause severe human diseases and are responsible for considerable mortality and morbidity worldwide. Therefore, researchers have conducted genetic screens to enhance insight into viral dependency and develop potential anti-viral strategies to treat and prevent these infections. The host factors identified by the clustered regularly interspaced short palindromic repeats (CRISPR) system can be potential targets for drug development. Meanwhile, CRISPR technology can be efficiently used to treat viral diseases as it targets both DNA and RNA. This paper discusses the host factors related to the life cycle of viruses of this family that were recently discovered using the CRISPR system. It also explores the role of immune factors and recent advances in gene editing in treating flavivirus-related diseases. The ever-increasing advancements of this technology may promise new therapeutic approaches with unique capabilities, surpassing the traditional methods of drug production and treatment.
Collapse
Affiliation(s)
- Zahra Ramezannia
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Shamekh
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Servadio JL, Convertino M, Fiecas M, Muñoz‐Zanzi C. Weekly Forecasting of Yellow Fever Occurrence and Incidence via Eco-Meteorological Dynamics. GEOHEALTH 2023; 7:e2023GH000870. [PMID: 37885914 PMCID: PMC10599710 DOI: 10.1029/2023gh000870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/31/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
Yellow Fever (YF), a mosquito-borne disease, requires ongoing surveillance and prevention due to its persistence and ability to cause major epidemics, including one that began in Brazil in 2016. Forecasting based on factors influencing YF risk can improve efficiency in prevention. This study aimed to produce weekly forecasts of YF occurrence and incidence in Brazil using weekly meteorological and ecohydrological conditions. Occurrence was forecast as the probability of observing any cases, and incidence was forecast to represent morbidity if YF occurs. We fit gamma hurdle models, selecting predictors from several meteorological and ecohydrological factors, based on forecast accuracy defined by receiver operator characteristic curves and mean absolute error. We fit separate models for data before and after the start of the 2016 outbreak, forecasting occurrence and incidence for all municipalities of Brazil weekly. Different predictor sets were found to produce most accurate forecasts in each time period, and forecast accuracy was high for both time periods. Temperature, precipitation, and previous YF burden were most influential predictors among models. Minimum, maximum, mean, and range of weekly temperature, precipitation, and humidity contributed to forecasts, with optimal lag times of 2, 6, and 7 weeks depending on time period. Results from this study show the use of environmental predictors in providing regular forecasts of YF burden and producing nationwide forecasts. Weekly forecasts, which can be produced using the forecast model developed in this study, are beneficial for informing immediate preparedness measures.
Collapse
Affiliation(s)
- Joseph L. Servadio
- Department of BiologyCenter for Infectious Disease DynamicsPennsylvania State UniversityUniversity ParkPAUSA
- Division of Environmental Health SciencesSchool of Public HealthUniversity of MinnesotaMinneapolisMNUSA
| | | | - Mark Fiecas
- Division of BiostatisticsSchool of Public HealthUniversity of MinnesotaMinneapolisMNUSA
| | - Claudia Muñoz‐Zanzi
- Division of Environmental Health SciencesSchool of Public HealthUniversity of MinnesotaMinneapolisMNUSA
| |
Collapse
|
17
|
Bonney JHK, Sanders T, Pratt D, Agbodzi B, Laryea D, Agyeman NKF, Kumordjie S, Attiku K, Adams PL, Boateng GA, Ohene SA, Tamal C, Mawuli G, Yeboah C, Dadzie S, Kubio C, Asiedu-Bekoe F, Odoom JK. Molecular Characterization of Circulating Yellow Fever Viruses from Outbreak in Ghana, 2021-2022. Emerg Infect Dis 2023; 29:1818-1826. [PMID: 37610174 PMCID: PMC10461649 DOI: 10.3201/eid2909.221671] [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] [Indexed: 08/24/2023] Open
Abstract
Yellow fever virus, transmitted by infected Aedes spp. mosquitoes, causes an acute viral hemorrhagic disease. During October 2021-February 2022, a yellow fever outbreak in some communities in Ghana resulted in 70 confirmed cases with 35 deaths (case-fatality rate 50%). The outbreak started in a predominantly unvaccinated nomadic community in the Savannah region, from which 65% of the cases came. The molecular amplification methods we used for diagnosis produced full-length DNA sequences from 3 confirmed cases. Phylogenetic analysis characterized the 3 sequences within West Africa genotype II; strains shared a close homology with sequences from Cote d'Ivoire and Senegal. We deployed more sensitive advanced molecular diagnostic techniques, which enabled earlier detection, helped control spread, and improved case management. We urge increased efforts from health authorities to vaccinate vulnerable groups in difficult-to-access areas and to educate the population about potential risks for yellow fever infections.
Collapse
|
18
|
Binderup A, Galli A, Fossat N, Fernandez-Antunez C, Mikkelsen LS, Rivera-Rangel LR, Scheel TKH, Fahnøe U, Bukh J, Ramirez S. Differential activity of nucleotide analogs against tick-borne encephalitis and yellow fever viruses in human cell lines. Virology 2023; 585:179-185. [PMID: 37356253 DOI: 10.1016/j.virol.2023.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/25/2023] [Accepted: 06/05/2023] [Indexed: 06/27/2023]
Abstract
With no approved antiviral therapies, the continuous emergence and re-emergence of tick-borne encephalitis virus (TBEV) and yellow fever virus (YFV) is a rising concern. We performed head-to-head comparisons of the antiviral activity of available nucleos(t)ide analogs (nucs) using relevant human cell lines. Eight existing nucs inhibited TBEV and/or YFV with differential activity between cell lines and viruses. Remdesivir, uprifosbuvir and sofosbuvir were the most potent drugs against TBEV and YFV in liver cells, but they had reduced activity in neural cells, whereas galidesivir retained uniform activity across cell lines and viruses. Ribavirin, valopicitabine, molnupiravir and GS-6620 exhibited only moderate antiviral activity. We found antiviral activity for drugs previously reported as inactive, demonstrating the importance of using human cell lines and comparative experimental assays when screening the activity of nucs. The relatively high antiviral activity of remdesivir, sofosbuvir and uprifosbuvir against TBEV and YFV merits further investigation in clinical studies.
Collapse
Affiliation(s)
- Alekxander Binderup
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Galli
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolas Fossat
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carlota Fernandez-Antunez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lotte S Mikkelsen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lizandro René Rivera-Rangel
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Troels K H Scheel
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Santseharay Ramirez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
19
|
Guterres A. Viral load: We need a new look at an old problem? J Med Virol 2023; 95:e29061. [PMID: 37638475 DOI: 10.1002/jmv.29061] [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: 01/27/2023] [Revised: 07/22/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
The concept of viral load was introduced in the 1980s to measure the amount of viral genetic material in a person's blood, primarily for human immunodeficiency virus (HIV). It has since become crucial for monitoring HIV infection progression and assessing the efficacy of antiretroviral therapy. However, during the coronavirus disease 2019 pandemic, the term "viral load" became widely popularized, not only for the scientific community but for the general population. Viral load plays a critical role in both clinical patient management and research, providing valuable insights for antiviral treatment strategies, vaccination efforts, and epidemiological control measures. As measuring viral load is so important, why don't researchers discuss the best way to do it? Is it simply acceptable to use raw Ct values? Relying solely on Ct values for viral load estimation can be problematic due to several reasons. First, Ct values can vary between different quantitative polymerase chain reaction assays, platforms, and laboratories, making it difficult to compare data across studies. Second, Ct values do not directly measure the quantity of viral particles in a sample and they can be influenced by various factors such as initial viral load, sample quality, and assay sensitivity. Moreover, variations in viral RNA extraction and reverse-transcription steps can further impact the accuracy of viral load estimation, emphasizing the need for careful interpretation of Ct values in viral load assessment. Interestingly, we did not observe scientific articles addressing different strategies to quantify viral load. The absence of standardized and validated methods impedes the implementation of viral load monitoring in clinical management. The variability in cell quantities within samples and the variation in viral particle numbers within infected cells further challenge accurate viral load measurement and interpretation. To advance the field and improve patient outcomes, there is an urgent need for the development and validation of tailored, standardized methods for precise viral load quantification.
Collapse
Affiliation(s)
- Alexandro Guterres
- Laboratório de Hantaviroses e Rickettsioses, Instituto Oswaldo Cruz Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Jobe NB, Huijben S, Paaijmans KP. Non-target effects of chemical malaria vector control on other biological and mechanical infectious disease vectors. Lancet Planet Health 2023; 7:e706-e717. [PMID: 37558351 DOI: 10.1016/s2542-5196(23)00136-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 06/09/2023] [Accepted: 06/17/2023] [Indexed: 08/11/2023]
Abstract
Public health insecticides play a crucial role in malaria control and elimination programmes. Many other arthropods, including mechanical and biological vectors of infectious diseases, have similar indoor feeding or resting behaviours, or both, as malaria mosquitoes, and could be exposed to the same insecticides. In this Personal View, we show that little is known about the insecticide susceptibility status and the extent of exposure to malaria interventions of other arthropod species. We highlight that there is an urgent need to better understand the selection pressure for insecticide resistance in those vectors, to ensure current and future active ingredients remain effective in targeting a broad range of arthropod species, allowing us to prevent and control future outbreaks of infectious diseases other than malaria.
Collapse
Affiliation(s)
- Ndey Bassin Jobe
- The Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Silvie Huijben
- The Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA; Simon A Levin Mathematical, Computational and Modeling Sciences Center, Arizona State University, Tempe, AZ, USA
| | - Krijn P Paaijmans
- The Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ, USA; Simon A Levin Mathematical, Computational and Modeling Sciences Center, Arizona State University, Tempe, AZ, USA; The Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, USA; ISGlobal, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique.
| |
Collapse
|
21
|
de Sousa FTG, Warnes CM, Manuli ER, Ng A, D’Elia Zanella LGFAB, Ho YL, Bhat S, Romano CM, Beatty PR, Biering SB, Kallas EG, Sabino EC, Harris E. Yellow fever disease severity and endothelial dysfunction are associated with elevated serum levels of viral NS1 protein and syndecan-1. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.29.23292053. [PMID: 37425955 PMCID: PMC10327263 DOI: 10.1101/2023.06.29.23292053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Yellow fever virus (YFV) infections can cause severe disease manifestations, including hepatic injury, endothelial damage, coagulopathy, hemorrhage, systemic organ failure, and shock, and are associated with high mortality in humans. While nonstructural protein 1 (NS1) of the related dengue virus is implicated in contributing to vascular leak, little is known about the role of YFV NS1 in severe YF and mechanisms of vascular dysfunction in YFV infections. Here, using serum samples from qRT-PCR-confirmed YF patients with severe (n=39) or non-severe (n=18) disease in a well-defined hospital cohort in Brazil, plus samples from healthy uninfected controls (n=11), we investigated factors associated with disease severity. We developed a quantitative YFV NS1 capture ELISA and found significantly increased levels of NS1, as well as syndecan-1, a marker of vascular leak, in serum from severe YF as compared to non-severe YF or control groups. We also showed that hyperpermeability of endothelial cell monolayers treated with serum from severe YF patients was significantly higher compared to non-severe YF and control groups as measured by transendothelial electrical resistance (TEER). Further, we demonstrated that YFV NS1 induces shedding of syndecan-1 from the surface of human endothelial cells. Notably, YFV NS1 serum levels significantly correlated with syndecan-1 serum levels and TEER values. Syndecan-1 levels also significantly correlated with clinical laboratory parameters of disease severity, viral load, hospitalization, and death. In summary, this study points to a role for secreted NS1 in YF disease severity and provides evidence for endothelial dysfunction as a mechanism of YF pathogenesis in humans.
Collapse
Affiliation(s)
- Francielle T. G. de Sousa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, 05403000, Brazil
| | - Colin M. Warnes
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Erika R. Manuli
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, 05403000, Brazil
- Laboratório de Investigação Médica, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo-SP, 05403000, Brazil
| | - Arash Ng
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Luiz G. F. A. B. D’Elia Zanella
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo-SP, 05403000, Brazil
- Instituto de Infectologia Emílio Ribas, São Paulo-SP, 01246-900, Brazil
| | - Yeh-Li Ho
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo-SP, 05403000, Brazil
| | - Samhita Bhat
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Camila M. Romano
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, 05403000, Brazil
- Laboratório de Investigação Médica, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo-SP, 05403000, Brazil
| | - P. Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Esper G. Kallas
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo-SP, 05403000, Brazil
| | - Ester C. Sabino
- Departamento de Doenças Infecciosas e Parasitárias, Instituto de Medicina Tropical, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, 05403000, Brazil
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo-SP, 05403000, Brazil
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
22
|
Silva NIO, Albery GF, Arruda MS, Oliveira GFG, Costa TA, de Mello ÉM, Moreira GD, Reis EV, da Silva SA, Silva MC, de Almeida MG, Becker DJ, Carlson CJ, Vasilakis N, Hanley KA, Drumond BP. Ecological drivers of sustained enzootic yellow fever virus transmission in Brazil, 2017-2021. PLoS Negl Trop Dis 2023; 17:e0011407. [PMID: 37276217 PMCID: PMC10270639 DOI: 10.1371/journal.pntd.0011407] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 06/15/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
Beginning December 2016, sylvatic yellow fever (YF) outbreaks spread into southeastern Brazil, and Minas Gerais state experienced two sylvatic YF waves (2017 and 2018). Following these massive YF waves, we screened 187 free-living non-human primate (NHPs) carcasses collected throughout the state between January 2019 and June 2021 for YF virus (YFV) using RTqPCR. One sample belonging to a Callithrix, collected in June 2020, was positive for YFV. The viral strain belonged to the same lineage associated with 2017-2018 outbreaks, showing the continued enzootic circulation of YFV in the state. Next, using data from 781 NHPs carcasses collected in 2017-18, we used generalized additive mixed models (GAMMs) to identify the spatiotemporal and host-level drivers of YFV infection and intensity (an estimation of genomic viral load in the liver of infected NHP). Our GAMMs explained 65% and 68% of variation in virus infection and intensity, respectively, and uncovered strong temporal and spatial patterns for YFV infection and intensity. NHP infection was higher in the eastern part of Minas Gerais state, where 2017-2018 outbreaks affecting humans and NHPs were concentrated. The odds of YFV infection were significantly lower in NHPs from urban areas than from urban-rural or rural areas, while infection intensity was significantly lower in NHPs from urban areas or the urban-rural interface relative to rural areas. Both YFV infection and intensity were higher during the warm/rainy season compared to the cold/dry season. The higher YFV intensity in NHPs in warm/rainy periods could be a result of higher exposure to vectors and/or higher virus titers in vectors during this time resulting in the delivery of a higher virus dose and higher viral replication levels within NHPs. Further studies are needed to better test this hypothesis and further compare the dynamics of YFV enzootic cycles between different seasons.
Collapse
Affiliation(s)
| | - Gregory F. Albery
- Department of Biology, Georgetown University, Washington, DC, United States of America
| | - Matheus Soares Arruda
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Thaís Alkifeles Costa
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Érica Munhoz de Mello
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriel Dias Moreira
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erik Vinícius Reis
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Simone Agostinho da Silva
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Marlise Costa Silva
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Munique Guimarães de Almeida
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel J. Becker
- Department of Biology, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Colin J. Carlson
- Department of Biology, Georgetown University, Washington, DC, United States of America
- Center for Global Health Science and Security, Georgetown University, Washington, D.C., United States of America
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Vector-Borne and Zoonotic Diseases, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathryn A. Hanley
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Betânia Paiva Drumond
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
23
|
Faggioni G, De Santis R, Moramarco F, Di Donato M, De Domenico A, Molinari F, Petralito G, Fortuna C, Venturi G, Rezza G, Lista F. Pan-Yellow Fever Virus Detection and Lineage Assignment by Real-Time RT-PCR and Amplicon Sequencing. J Virol Methods 2023; 316:114717. [PMID: 36972832 DOI: 10.1016/j.jviromet.2023.114717] [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: 09/20/2022] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Yellow fever disease is a viral zoonosis that may result in a severe hemorrhagic disease. A safe and effective vaccine used in mass immunization campaigns has allowed control and mitigation against explosive outbreaks in endemic areas. Since the 1960's, re-emergent of the yellow fever virus has been observed. The timely implementation of control measures, to avoid or contain an ongoing outbreak requires rapid specific viral detection methods. Here a novel molecular assay, expected to detect all known yellow fever virus strains, is described. The method has demonstrated high sensitivity and specificity in real-time RT-PCR as well as in an endpoint RT-PCR set-up. Sequence alignment and phylogenetic analysis reveal that the amplicon resulting from the novel method covers a genomic region whose mutational profile is completely associated to the yellow fever viral lineages. Therefore, sequencing analysis of this amplicon allows for assignment of the viral lineage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Claudia Fortuna
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Giulietta Venturi
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Giovanni Rezza
- Health Prevention Directorate, Ministry of Health, Rome, Italy.
| | - Florigio Lista
- Army Medical Center, Scientific Department, Rome, Italy.
| |
Collapse
|
24
|
Karnam S, Huang Y, Nguyen N, Yeh S. Ophthalmic consequences of viral hemorrhagic fevers: Insights from the clinic and laboratory. FRONTIERS IN TROPICAL DISEASES 2023. [DOI: 10.3389/fitd.2023.1107786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Viral hemorrhagic fevers (VHFs) are a diverse group of RNA virus-mediated systemic diseases with significant morbidity and mortality and represent a significant public health concern. Given the high systemic morbidity and mortality in a number of these entities, delays in diagnosis can lead to downstream public health consequences. Many viral hemorrhagic fevers have ophthalmic manifestations and ophthalmologists thus play a key role in disease recognition and the management of ocular complications associated with specific hemorrhagic fevers. This review summarizes the key ophthalmic consequences of viral hemorrhagic fevers, viral disease pathogenesis, disease findings, and areas of unmet research need.
Collapse
|
25
|
Perrin A, Pellet J, Bergonzoli L, Christe P, Glaizot O. Amphibian abundance is associated with reduced mosquito presence in human‐modified landscapes. Ecosphere 2023. [DOI: 10.1002/ecs2.4484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023] Open
|
26
|
Bignon E, Dumont E, Monari A. Molecular Basis of the pH-Controlled Maturation of the Tick-Borne Encephalitis Flavivirus. J Phys Chem Lett 2023; 14:1977-1982. [PMID: 36790164 DOI: 10.1021/acs.jpclett.2c03551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Flaviviruses are enveloped viruses causing high public concerns. Their maturation spans several cellular compartments having different pH. Thus, complex control mechanisms are in place to avoid premature maturation. Here we report the dynamical behavior at neutral and acidic pH of the precursor of the membrane fusion protein E of tick-borne encephalitis, showing the different stabilizations of the E dimer and the role played by the small fusion-assisting protomer (pr). The comprehension, at atomic resolution, of the fine regulation of viral maturation will be fundamental to the development of efficient strategies against emerging viral threats.
Collapse
Affiliation(s)
- Emmanuelle Bignon
- Université de Lorraine and CNRS, UMR 7019 LPCT, F-5400, Nancy, France
| | - Elise Dumont
- Université Côte d'Azur, Institut de Chimie de Nice, UMR 7272, Parc Valrose, 28 avenue Valrose, F-06108, Nice, France
- Institut Universitaire de France, 5 rue Descartes, F-75005, Paris, France
| | - Antonio Monari
- Université Paris Cité and CNRS, ITODYS, F-75006, Paris, France
| |
Collapse
|
27
|
Howard-Jones AR, Pham D, Sparks R, Maddocks S, Dwyer DE, Kok J, Basile K. Arthropod-Borne Flaviviruses in Pregnancy. Microorganisms 2023; 11:433. [PMID: 36838398 PMCID: PMC9959669 DOI: 10.3390/microorganisms11020433] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Flaviviruses are a diverse group of enveloped RNA viruses that cause significant clinical manifestations in the pregnancy and postpartum periods. This review highlights the epidemiology, pathophysiology, clinical features, diagnosis, and prevention of the key arthropod-borne flaviviruses of concern in pregnancy and the neonatal period-Zika, Dengue, Japanese encephalitis, West Nile, and Yellow fever viruses. Increased disease severity during pregnancy, risk of congenital malformations, and manifestations of postnatal infection vary widely amongst this virus family and may be quite marked. Laboratory confirmation of infection is complex, especially due to the reliance on serology for which flavivirus cross-reactivity challenges diagnostic specificity. As such, a thorough clinical history including relevant geographic exposures and prior vaccinations is paramount for accurate diagnosis. Novel vaccines are eagerly anticipated to ameliorate the impact of these flaviviruses, particularly neuroinvasive disease manifestations and congenital infection, with consideration of vaccine safety in pregnant women and children pivotal. Moving forward, the geographical spread of flaviviruses, as for other zoonoses, will be heavily influenced by climate change due to the potential expansion of vector and reservoir host habitats. Ongoing 'One Health' engagement across the human-animal-environment interface is critical to detect and responding to emergent flavivirus epidemics.
Collapse
Affiliation(s)
- Annaleise R. Howard-Jones
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - David Pham
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
| | - Rebecca Sparks
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
| | - Susan Maddocks
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
| | - Dominic E. Dwyer
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
- Centre for Infectious Diseases and Microbiology-Public Health, Westmead, NSW 2145, Australia
| | - Jen Kok
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Centre for Infectious Diseases and Microbiology-Public Health, Westmead, NSW 2145, Australia
| | - Kerri Basile
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
| |
Collapse
|
28
|
Montalvo Zurbia-Flores G, Rollier CS, Reyes-Sandoval A. Re-thinking yellow fever vaccines: fighting old foes with new generation vaccines. Hum Vaccin Immunother 2022; 18:1895644. [PMID: 33974507 PMCID: PMC8920179 DOI: 10.1080/21645515.2021.1895644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/07/2021] [Accepted: 02/21/2021] [Indexed: 11/16/2022] Open
Abstract
Despite the existence of a highly efficient yellow fever vaccine, yellow fever reemergence throughout Africa and the Americas has put 900 million people in 47 countries at risk of contracting the disease. Although the vaccine has been key to controlling yellow fever epidemics, its live-attenuated nature comes with a range of contraindications that prompts advising against its administration to pregnant and lactating women, immunocompromised individuals, and those with hypersensitivity to chicken egg proteins. Additionally, large outbreaks have highlighted problems with insufficient vaccine supply, whereby manufacturers rely on slow traditional manufacturing processes that prevent them from ramping up production. These limitations have contributed to an inadequate control of yellow fever and have favored the pursuit of novel yellow fever vaccine candidates that aim to circumvent the licensed vaccine's restrictions. Here, we review the live-attenuated vaccine's limitations and explore the epitome of a yellow fever vaccine, whilst scrutinizing next-generation vaccine candidates.
Collapse
Affiliation(s)
- Gerardo Montalvo Zurbia-Flores
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford. The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford. The Henry Wellcome Building for Molecular Physiology, Oxford, UK
- Instituto Politécnico Nacional, IPN. Av. Luis Enrique Erro S/n. Unidad Adolfo López Mateos. CP, Mexico City, Mexico
| |
Collapse
|
29
|
Salgado Á, de Melo-Minardi RC, Giovanetti M, Veloso A, Morais-Rodrigues F, Adelino T, de Jesus R, Tosta S, Azevedo V, Lourenco J, Alcantara LCJ. Machine learning models exploring characteristic single-nucleotide signatures in yellow fever virus. PLoS One 2022; 17:e0278982. [PMID: 36508435 PMCID: PMC9744328 DOI: 10.1371/journal.pone.0278982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
Yellow fever virus (YFV) is the agent of the most severe mosquito-borne disease in the tropics. Recently, Brazil suffered major YFV outbreaks with a high fatality rate affecting areas where the virus has not been reported for decades, consisting of urban areas where a large number of unvaccinated people live. We developed a machine learning framework combining three different algorithms (XGBoost, random forest and regularized logistic regression) to analyze YFV genomic sequences. This method was applied to 56 YFV sequences from human infections and 27 from non-human primate (NHPs) infections to investigate the presence of genetic signatures possibly related to disease severity (in human related sequences) and differences in PCR cycle threshold (Ct) values (in NHP related sequences). Our analyses reveal four non-synonymous single nucleotide variations (SNVs) on sequences from human infections, in proteins NS3 (E614D), NS4a (I69V), NS5 (R727G, V643A) and six non-synonymous SNVs on NHP sequences, in proteins E (L385F), NS1 (A171V), NS3 (I184V) and NS5 (N11S, I374V, E641D). We performed comparative protein structural analysis on these SNVs, describing possible impacts on protein function. Despite the fact that the dataset is limited in size and that this study does not consider virus-host interactions, our work highlights the use of machine learning as a versatile and fast initial approach to genomic data exploration.
Collapse
Affiliation(s)
- Álvaro Salgado
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail: (AS); (LCJA); (JL)
| | - Raquel C. de Melo-Minardi
- Departamento de Ciência da Computação, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marta Giovanetti
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Adriano Veloso
- Departamento de Ciência da Computação, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Francielly Morais-Rodrigues
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Talita Adelino
- Laboratório Central de Saúde Pública, Fundação Ezequiel Dias, Belo Horizonte, Minas Gerais, Brazil
| | - Ronaldo de Jesus
- Coordenação Geral dos Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, DF, Brazil
| | - Stephane Tosta
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - José Lourenco
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- * E-mail: (AS); (LCJA); (JL)
| | - Luiz Carlos J. Alcantara
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- * E-mail: (AS); (LCJA); (JL)
| |
Collapse
|
30
|
Perrin A, Glaizot O, Christe P. Worldwide impacts of landscape anthropization on mosquito abundance and diversity: A meta-analysis. GLOBAL CHANGE BIOLOGY 2022; 28:6857-6871. [PMID: 36107000 PMCID: PMC9828797 DOI: 10.1111/gcb.16406] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 05/23/2023]
Abstract
In recent decades, the emergence and resurgence of vector-borne diseases have been well documented worldwide, especially in tropical regions where protection and defense tools for human populations are still very limited. In this context, the dynamics of pathogens are influenced by landscape anthropization (i.e., urbanization, deforestation, and agricultural development), and one of the mechanisms through which this occurs is a change in the abundance and/or diversity of the vectors. An increasing number of empirical studies have described heterogeneous effects of landscape anthropization on vector communities; therefore, it is difficult to have an overall picture of these effects on a global scale. Here, we performed a meta-analysis to quantify the impacts of landscape anthropization on a global scale on the presence/abundance and diversity of mosquitoes, the most important arthropods affecting human health. We obtained 338 effect sizes on 132 mosquito species, compiled from 107 studies in 52 countries that covered almost every part of the world. The results of the meta-analysis showed an overall decline of mosquito presence/abundance and diversity in response to urbanization, deforestation, and agricultural development, except for a few mosquito species that have been able to exploit landscape anthropization well. Our results highlighted that these few favored mosquito species are those of global concern. They, thus, provide a better understanding of the overall effect of landscape anthropization on vector communities and, more importantly, suggest a greater risk of emergence and transmission of vector-borne diseases in human-modified landscapes.
Collapse
Affiliation(s)
- Antoine Perrin
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| | - Olivier Glaizot
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
- Museum of ZoologyLausanneSwitzerland
| | - Philippe Christe
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
31
|
Opara NU, Nwagbara UI, Hlongwana KW. The COVID-19 Impact on the Trends in Yellow Fever and Lassa Fever Infections in Nigeria. Infect Dis Rep 2022; 14:932-941. [PMID: 36412749 PMCID: PMC9680345 DOI: 10.3390/idr14060091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Lassa fever (LF) and yellow fever (YF) belong to a group of viral hemorrhagic fevers (VHFs). These viruses have common features and damages the organs and blood vessels; they also impair the body's homeostasis. Some VHFs cause mild disease, while some cause severe disease and death such as in the case of Ebola or Marburg. LF virus and YF virus are two of the most recent emerging viruses in Africa, resulting in severe hemorrhagic fever in humans. Lassa fever virus is continuously on the rise both in Nigeria and neighboring countries in West Africa, with an estimate of over 500,000 cases of LF, and 5000 deaths, annually. YF virus is endemic in temperate climate regions of Africa, Central America (Guatemala, Honduras, Nicaragua, El Salvador), and South America (such as Brazil, Argentina, Peru, and Chile) with an annual estimated cases of 200,000 and 30,000 deaths globally. This review examines the impact of the COVID-19 pandemic on the trend in epidemiology of these two VHFs to delineate responses that are associated with protective or pathogenic outcomes.
Collapse
Affiliation(s)
- Nnennaya U. Opara
- Institute for Academic Medicine, Department of Emergency Medicine, Charleston Area Medical Center, Charleston, WV 25304, USA
- Department of Health Administration, University of Phoenix, Phoenix, AZ 85040, USA
- Correspondence: or
| | - Ugochinyere I. Nwagbara
- Department of Public Health Medicine, College of Health Sciences, University of KwaZulu-Natal, Howard Campus, Durban 4041, South Africa
| | - Khumbulani W. Hlongwana
- Cancer and Infectious Disease Epidemiology Research Unit (CIDERU), College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
32
|
Zida-Compaore WIC, Gbeasor-Komlanvi FA, Tchankoni MK, Halatoko WA, Sadio AJ, Konu YR, Gnatou GYS, Koba AK, Agbonon A, Ekouevi DK. Estimation of yellow fever incidence in Togo between 2010 and 2020. Travel Med Infect Dis 2022; 50:102470. [DOI: 10.1016/j.tmaid.2022.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/16/2022] [Accepted: 10/02/2022] [Indexed: 11/30/2022]
|
33
|
Sang R, Lutomiah J, Chepkorir E, Tchouassi DP. Evolving dynamics of Aedes-borne diseases in Africa: a cause for concern. CURRENT OPINION IN INSECT SCIENCE 2022; 53:100958. [PMID: 35878761 DOI: 10.1016/j.cois.2022.100958] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
Aedes-borne viruses, yellow fever (YF), dengue, Chikungunya and Zika are taking a huge toll on global health as Africa faces re-emergence with potential for massive human catastrophe. Transmission driven by diverse vectors in ecological settings that range from urban to rural and sylvatic habitats with human and nonhuman primate/reservoir activities across such habitats has facilitated virus movement and spillover to susceptible human populations. Approved vaccine exists for YF, although availability for routine and mass vaccination is often constrained. Integrating vector surveillance, understanding disease ecology with rationalised vaccination in high-risk areas (YF) remains important in disease prevention and control. We review trends in disease occurrence in Africa, hinting on gaps in disease detection and management and the prospects for prevention and/or control.
Collapse
Affiliation(s)
- Rosemary Sang
- International Centre of Insect Physiology and Ecology, P.O. Box 30772-00100, Nairobi, Kenya.
| | - Joel Lutomiah
- Center for Virus Research, Kenya Medical Research Institute, Kenya
| | - Edith Chepkorir
- Center for Virus Research, Kenya Medical Research Institute, Kenya
| | - David P Tchouassi
- International Centre of Insect Physiology and Ecology, P.O. Box 30772-00100, Nairobi, Kenya
| |
Collapse
|
34
|
Guimarães A, Oliveira MC, Kierulff MCM, Mendonça-Furtado O, Baptista MNM, Mendes SL, Almada GL. Epidemiologic profile and histopathological findings in Neotropical Primates during and after the yellow fever outbreak in Espírito Santo, Brazil. AN ACAD BRAS CIENC 2022; 94:e20211229. [PMID: 36074428 DOI: 10.1590/0001-3765202220211229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/21/2022] [Indexed: 11/22/2022] Open
Abstract
Yellow fever (YF) is a viral disease whose transmission involves non-human primates (NHP), mosquitoes, and humans. Between 2016 and 2018 occurred the largest YF outbreak in the last 100 years in Brazil. We analyzed epidemiologic profile and geographic distribution of epizootics and described most frequent histopathological findings in NHP that died during YF outbreak in the state of Espírito Santo. We consider 487 epizootics notifications registered at the State Health Department from January 2017 to July 2020. Throughout the state, 51 (65.4%) municipalities reported epizootics, with more cases in central and metropolitan areas. Reverse transcription polymerase chain reaction and immunohistochemistry were laboratory tests performed for diagnosis of yellow fever, with 160 (32.9%) positive results, 314 (64.5%) negative and 13 (2.7%) inconclusive. Histopathological findings were compared statistically between positive and negative animals for YF. The liver was the most affected organ. Hemorrhage, hepatocyte necrosis, steatosis, cholestasis and eosinophilic degeneration were statistically more frequent in positive animals. Tubular necrosis, nephritis, congestion and lymphoid hypoplasia on spleen were statistically correlated to positive animals. Knowledge of pathogenic aspect of YF is necessary to guarantee that samples from Neotropical primates are properly used for YF surveillance purposes, to ensure appropriate diagnoses and subsequent public health responses.
Collapse
Affiliation(s)
- Andresa Guimarães
- Instituto Nacional da Mata Atlântica/INMA, Avenida José Ruschi, 04, Centro, 29650-000 Santa Teresa, ES, Brazil
| | - Mariana C Oliveira
- Universidade Estácio de Sá, Departamento de Medicina Veterinária, Campus Vargem Pequena, Estrada da Boca do Mato, 850, Vargem Pequena, 22783-325 Rio de Janeiro, RJ, Brazil
| | - Maria Cecilia M Kierulff
- Instituto Nacional da Mata Atlântica/INMA, Avenida José Ruschi, 04, Centro, 29650-000 Santa Teresa, ES, Brazil.,Instituto Pri-Matas para a Conservação da Biodiversidade, Rua Dulce Maria, 337, Ipiranga, 31160-250 Belo Horizonte, MG, Brazil
| | - Olivia Mendonça-Furtado
- Instituto Nacional da Mata Atlântica/INMA, Avenida José Ruschi, 04, Centro, 29650-000 Santa Teresa, ES, Brazil
| | - Michelle N M Baptista
- Instituto Nacional da Mata Atlântica/INMA, Avenida José Ruschi, 04, Centro, 29650-000 Santa Teresa, ES, Brazil
| | - Sérgio L Mendes
- Instituto Nacional da Mata Atlântica/INMA, Avenida José Ruschi, 04, Centro, 29650-000 Santa Teresa, ES, Brazil
| | - Gilton Luiz Almada
- Centro de Informações Estratégicas e Respostas em Vigilância em Saúde - Secretaria de Estado da Saúde do Espírito Santo (SESA), Rua Eng. Guilherme José Monjardim Varejão, 225, Ed. Enseada Plaza, Enseada do Suá, 29050-260 Vitória, ES, Brazil
| |
Collapse
|
35
|
Emergence of New Immunopathogenic Factors in Human Yellow Fever: Polarisation of the M1/M2 Macrophage Response in the Renal Parenchyma. Viruses 2022; 14:v14081725. [PMID: 36016347 PMCID: PMC9416648 DOI: 10.3390/v14081725] [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: 07/02/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 12/04/2022] Open
Abstract
Macrophages in the kidney play a pathogenic role in inflammation and fibrosis. Our study aimed to understand the polarisation of the M1 and M2 phenotypic profiles of macrophages in injured kidney tissue retrieved from fatal cases of yellow fever virus (YFV). A total of 11 renal tissue biopsies obtained from patients who died of yellow fever (YF) were analysed. To detect antibodies that promote the classical and alternative pathways of macrophage activation, immunohistochemical analysis was performed to detect CD163, CD68, inducible nitric oxide synthase (iNOS), arginase 1, interleukin (IL)-4, IL-10, interferon (IFN)-γ, IFN-β, tumour necrosis factor (TNF)-α, IL-13, and transforming growth factor (TGF)-β. There was a difference in the marker expression between fatal cases of YFV and control samples, with increased expression in the cortical region of the renal parenchyma. The immunoexpression of CD68 and CD163 receptors suggests the presence of activated macrophages migrating to infectious foci. The rise in IL-10, IL-4, and IL-13 indicated their potential role in the inactivation of the inflammatory macrophage response and phenotypic modulation of M2 macrophages. The altered expression of IFN-γ and IFN-β demonstrates the importance of the innate immune response in combating microorganisms. Our findings indicate that the polarisation of M1 and M2 macrophages plays a vital role in the renal immune response to YFV.
Collapse
|
36
|
Oyono MG, Kenmoe S, Abanda NN, Takuissu GR, Ebogo-Belobo JT, Kenfack-Momo R, Kengne-Nde C, Mbaga DS, Tchatchouang S, Kenfack-Zanguim J, Lontuo Fogang R, Zeuko’o Menkem E, Ndzie Ondigui JL, Kame-Ngasse GI, Magoudjou-Pekam JN, Bowo-Ngandji A, Nkie Esemu S, Ndip L. Epidemiology of yellow fever virus in humans, arthropods, and non-human primates in sub-Saharan Africa: A systematic review and meta-analysis. PLoS Negl Trop Dis 2022; 16:e0010610. [PMID: 35867659 PMCID: PMC9307179 DOI: 10.1371/journal.pntd.0010610] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022] Open
Abstract
Yellow fever (YF) has re-emerged in the last two decades causing several outbreaks in endemic countries and spreading to new receptive regions. This changing epidemiology of YF creates new challenges for global public health efforts. Yellow fever is caused by the yellow fever virus (YFV) that circulates between humans, the mosquito vector, and non-human primates (NHP). In this systematic review and meta-analysis, we review and analyse data on the case fatality rate (CFR) and prevalence of YFV in humans, and on the prevalence of YFV in arthropods, and NHP in sub-Saharan Africa (SSA). We performed a comprehensive literature search in PubMed, Web of Science, African Journal Online, and African Index Medicus databases. We included studies reporting data on the CFR and/or prevalence of YFV. Extracted data was verified and analysed using the random effect meta-analysis. We conducted subgroup, sensitivity analysis, and publication bias analyses using the random effect meta-analysis while I2 statistic was employed to determine heterogeneity. This review was registered with PROSPERO under the identification CRD42021242444. The final meta-analysis included 55 studies. The overall case fatality rate due to YFV was 31.1% (18.3–45.4) in humans and pooled prevalence of YFV infection was 9.4% (6.9–12.2) in humans. Only five studies in West and East Africa detected the YFV in mosquito species of the genus Aedes and in Anopheles funestus. In NHP, YFV antibodies were found only in members of the Cercopithecidae family. Our analysis provides evidence on the ongoing circulation of the YFV in humans, Aedes mosquitoes and NHP in SSA. These observations highlight the ongoing transmission of the YFV and its potential to cause large outbreaks in SSA. As such, strategies such as those proposed by the WHO’s Eliminate Yellow Fever Epidemics (EYE) initiative are urgently needed to control and prevent yellow fever outbreaks in SSA.
Collapse
Affiliation(s)
- Martin Gael Oyono
- Centre for Research on Health and Priority Pathologies, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
- Laboratory of Parasitology and Ecology, Department of Animal Biology and Physiology, University of Yaounde I, Yaounde, Cameroon
| | - Sebastien Kenmoe
- Department of Microbiology and Parasitology, University of Buea, Buea, Cameroon
- * E-mail:
| | - Ngu Njei Abanda
- Virology Department, Centre Pasteur of Cameroon, Yaounde, Cameroon
| | - Guy Roussel Takuissu
- Centre for Food, Food Security and Nutrition Research, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | - Jean Thierry Ebogo-Belobo
- Medical Research Centre, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | - Raoul Kenfack-Momo
- Department of Biochemistry, The University of Yaounde I, Yaounde, Cameroon
| | - Cyprien Kengne-Nde
- Epidemiological Surveillance, Evaluation and Research Unit, National AIDS Control Committee, Douala, Cameroon
| | | | | | | | | | | | | | - Ginette Irma Kame-Ngasse
- Medical Research Centre, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | | | - Arnol Bowo-Ngandji
- Department of Microbiology, The University of Yaounde I, Yaounde, Cameroon
| | | | - Lucy Ndip
- Department of Microbiology and Parasitology, University of Buea, Buea, Cameroon
| |
Collapse
|
37
|
Role of Th17 Cytokines in the Liver’s Immune Response during Fatal Yellow Fever: Triggering Cell Damage Mechanisms. Cells 2022; 11:cells11132053. [PMID: 35805137 PMCID: PMC9265354 DOI: 10.3390/cells11132053] [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: 04/27/2022] [Revised: 06/04/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
Yellow fever (YF) is an infectious and acute viral haemorrhagic disease that triggers a cascade of host immune responses. We investigated the Th17 cytokine profile in the liver tissue of patients with fatal YF. Liver tissue samples were collected from 26 deceased patients, including 21 YF-positive and 5 flavivirus-negative patients, with preserved hepatic parenchyma architecture, who died of other causes. Histopathological and immunohistochemical analysis were performed on the liver samples to evaluate the Th17 profiles (ROR-γ, STAT3, IL-6, TGF-β, IL-17A, and IL-23). Substantial differences were found in the expression levels of these markers between the patients with fatal YF and controls. A predominant expression of Th17 cytokine markers was observed in the midzonal region of the YF cases, the most affected area in the liver acinus, compared with the controls. Histopathological changes in the hepatic parenchyma revealed cellular damage characterised mainly by the presence of inflammatory cell infiltrates, Councilman bodies (apoptotic cells), micro/macrovesicular steatosis, and lytic and coagulative necrosis. Hence, Th17 cytokines play a pivotal role in the immunopathogenesis of YF and contribute markedly to triggering cell damage in patients with fatal disease outcomes.
Collapse
|
38
|
Luria-Pérez R, Sánchez-Vargas LA, Muñoz-López P, Mellado-Sánchez G. Mucosal Vaccination: A Promising Alternative Against Flaviviruses. Front Cell Infect Microbiol 2022; 12:887729. [PMID: 35782117 PMCID: PMC9241634 DOI: 10.3389/fcimb.2022.887729] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae are a family of positive-sense, single-stranded RNA enveloped viruses, and their members belong to a single genus, Flavivirus. Flaviviruses are found in mosquitoes and ticks; they are etiological agents of: dengue fever, Japanese encephalitis, West Nile virus infection, Zika virus infection, tick-borne encephalitis, and yellow fever, among others. Only a few flavivirus vaccines have been licensed for use in humans: yellow fever, dengue fever, Japanese encephalitis, tick-borne encephalitis, and Kyasanur forest disease. However, improvement is necessary in vaccination strategies and in understanding of the immunological mechanisms involved either in the infection or after vaccination. This is especially important in dengue, due to the immunological complexity of its four serotypes, cross-reactive responses, antibody-dependent enhancement, and immunological interference. In this context, mucosal vaccines represent a promising alternative against flaviviruses. Mucosal vaccination has several advantages, as inducing long-term protective immunity in both mucosal and parenteral tissues. It constitutes a friendly route of antigen administration because it is needle-free and allows for a variety of antigen delivery systems. This has promoted the development of several ways to stimulate immunity through the direct administration of antigens (e.g., inactivated virus, attenuated virus, subunits, and DNA), non-replicating vectors (e.g., nanoparticles, liposomes, bacterial ghosts, and defective-replication viral vectors), and replicating vectors (e.g., Salmonella enterica, Lactococcus lactis, Saccharomyces cerevisiae, and viral vectors). Because of these characteristics, mucosal vaccination has been explored for immunoprophylaxis against pathogens that enter the host through mucosae or parenteral areas. It is suitable against flaviviruses because this type of immunization can stimulate the parenteral responses required after bites from flavivirus-infected insects. This review focuses on the advantages of mucosal vaccine candidates against the most relevant flaviviruses in either humans or animals, providing supporting data on the feasibility of this administration route for future clinical trials.
Collapse
Affiliation(s)
- Rosendo Luria-Pérez
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
| | - Luis A. Sánchez-Vargas
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Paola Muñoz-López
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
- Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gabriela Mellado-Sánchez
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Ciudad de México, Mexico
| |
Collapse
|
39
|
de Oliveira Figueiredo P, Stoffella-Dutra AG, Costa GB, de Oliveira JS, Amaral CD, Alves PA, Filho JDA, Paz GF, Tonelli GB, Kroon EG, Drumond BP, Paglia AP, de Oliveira DB, de Souza Trindade G. Absence of yellow fever virus circulation in wildlife rodents from Brazil. Braz J Microbiol 2022; 53:647-654. [PMID: 35133637 PMCID: PMC9151931 DOI: 10.1007/s42770-022-00688-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
Yellow fever (YF), caused by the yellow fever virus (YFV), is an emerging viral zoonosis that affects humans and non-human primates (NHP). In South America, YF is naturally maintained through enzootic/sylvatic cycles involving NHPs and mosquitoes (Haemagogus and Sabethes). In this study, we retrospectively analyzed wildlife rodents to better understand their role in a potential alternative YF sylvatic cycle. The plaque reduction neutralization test was performed to detect anti-YFV antibodies, while qPCR targeting the NS5 region of flaviviruses and standard PCR targeting the CprM region were applied to detect YFV RNA in tissue and blood samples. YFV was not evidenced in any of the tested samples. These findings provide additional information regarding sylvatic YFV and emphasize the importance of YFV surveillance in wild animals as potential reservoirs/hosts given the well-established enzootic cycle in the studied areas, mainly in the Atlantic Forest.
Collapse
Affiliation(s)
- Poliana de Oliveira Figueiredo
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Ana Gabriella Stoffella-Dutra
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| | - Galileu Barbosa Costa
- Departamento de Ciências da Saúde, Universidade Estadual de Santa Cruz, Campus Soane Nazaré de Andrade, Rodovia Jorge Amado, Km 16, Salobrinho, Ilhéus, BA, 45662-900, Brazil.
| | - Jaqueline Silva de Oliveira
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Carolina Dourado Amaral
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Pedro Augusto Alves
- Grupo de Imunologia de Doenças Virais, Instituto René Rachou, Fiocruz-Minas, Belo Horizonte, MG, Brazil
| | | | - Gustavo Fontes Paz
- Grupo de Estudos em Leishmanioses, Instituto René Rachou, Fiocruz-Minas, Belo Horizonte, MG, Brazil
| | - Gabriel Barbosa Tonelli
- Grupo de Estudos em Leishmanioses, Instituto René Rachou, Fiocruz-Minas, Belo Horizonte, MG, Brazil
| | - Erna Geessien Kroon
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Betânia Paiva Drumond
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Adriano Pereira Paglia
- Laboratório de Ecologia e Conservação, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danilo Bretas de Oliveira
- Centro Integrado de Pesquisa em Saúde, Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Giliane de Souza Trindade
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|
40
|
Caldwell HS, Pata JD, Ciota AT. The Role of the Flavivirus Replicase in Viral Diversity and Adaptation. Viruses 2022; 14:1076. [PMID: 35632818 PMCID: PMC9143365 DOI: 10.3390/v14051076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 02/04/2023] Open
Abstract
Flaviviruses include several emerging and re-emerging arboviruses which cause millions of infections each year. Although relatively well-studied, much remains unknown regarding the mechanisms and means by which these viruses readily alternate and adapt to different hosts and environments. Here, we review a subset of the different aspects of flaviviral biology which impact host switching and viral fitness. These include the mechanism of replication and structural biology of the NS3 and NS5 proteins, which reproduce the viral genome; rates of mutation resulting from this replication and the role of mutational frequency in viral fitness; and the theory of quasispecies evolution and how it contributes to our understanding of genetic and phenotypic plasticity.
Collapse
Affiliation(s)
- Haley S. Caldwell
- The Arbovirus Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, NY 12159, USA;
- Department of Biomedical Sciences, State University of New York at Albany, School of Public Health, Rensselaer, NY 12144, USA;
| | - Janice D. Pata
- Department of Biomedical Sciences, State University of New York at Albany, School of Public Health, Rensselaer, NY 12144, USA;
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - Alexander T. Ciota
- The Arbovirus Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, NY 12159, USA;
- Department of Biomedical Sciences, State University of New York at Albany, School of Public Health, Rensselaer, NY 12144, USA;
| |
Collapse
|
41
|
Mubemba B, Mburu MM, Changula K, Muleya W, Moonga LC, Chambaro HM, Kajihara M, Qiu Y, Orba Y, Hayashida K, Sutcliffe CG, Norris DE, Thuma PE, Ndubani P, Chitanga S, Sawa H, Takada A, Simulundu E. Current knowledge of vector-borne zoonotic pathogens in Zambia: A clarion call to scaling-up "One Health" research in the wake of emerging and re-emerging infectious diseases. PLoS Negl Trop Dis 2022; 16:e0010193. [PMID: 35120135 PMCID: PMC8849493 DOI: 10.1371/journal.pntd.0010193] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/16/2022] [Accepted: 01/24/2022] [Indexed: 12/19/2022] Open
Abstract
Background Although vector-borne zoonotic diseases are a major public health threat globally, they are usually neglected, especially among resource-constrained countries, including those in sub-Saharan Africa. This scoping review examined the current knowledge and identified research gaps of vector-borne zoonotic pathogens in Zambia. Methods and findings Major scientific databases (Web of Science, PubMed, Scopus, Google Scholar, CABI, Scientific Information Database (SID)) were searched for articles describing vector-borne (mosquitoes, ticks, fleas and tsetse flies) zoonotic pathogens in Zambia. Several mosquito-borne arboviruses have been reported including Yellow fever, Ntaya, Mayaro, Dengue, Zika, West Nile, Chikungunya, Sindbis, and Rift Valley fever viruses. Flea-borne zoonotic pathogens reported include Yersinia pestis and Rickettsia felis. Trypanosoma sp. was the only tsetse fly-borne pathogen identified. Further, tick-borne zoonotic pathogens reported included Crimean-Congo Haemorrhagic fever virus, Rickettsia sp., Anaplasma sp., Ehrlichia sp., Borrelia sp., and Coxiella burnetii. Conclusions This study revealed the presence of many vector-borne zoonotic pathogens circulating in vectors and animals in Zambia. Though reports of human clinical cases were limited, several serological studies provided considerable evidence of zoonotic transmission of vector-borne pathogens in humans. However, the disease burden in humans attributable to vector-borne zoonotic infections could not be ascertained from the available reports and this precludes the formulation of national policies that could help in the control and mitigation of the impact of these diseases in Zambia. Therefore, there is an urgent need to scale-up “One Health” research in emerging and re-emerging infectious diseases to enable the country to prepare for future epidemics, including pandemics. Despite vector-borne zoonoses being a major public health threat globally, they are often overlooked, particularly among resource-constrained countries in sub-Saharan Africa, including Zambia. Therefore, we reviewed the current knowledge and identified research gaps of vector-borne zoonotic pathogens in Zambia. We focussed on mosquito-, tick-, flea- and tsetse fly-borne zoonotic pathogens reported in the country. Although we found evidence of circulation of several vector-borne zoonotic pathogens among vectors, animals and humans, clinical cases in humans were rarely reported. This suggests sparse capacity for diagnosis of vector-borne pathogens in healthcare facilities in the country and possibly limited awareness and knowledge of the local epidemiology of these infectious agents. Establishment of facility-based surveillance of vector-borne zoonoses in health facilities could provide valuable insights on morbidity, disease severity, and mortalities associated with infections as well as immune responses. In addition, there is also need for increased genomic surveillance of vector-borne pathogens in vectors and animals and humans for a better understanding of the molecular epidemiology of these diseases in Zambia. Furthermore, vector ecology studies aimed at understanding the drivers of vector abundance, pathogen host range (i.e., including the range of vectors and reservoirs), parasite-host interactions and factors influencing frequency of human-vector contacts should be prioritized. The study revealed the need for Zambia to scale-up One Health research in emerging and re-emerging infectious diseases to enable the country to be better prepared for future epidemics, including pandemics.
Collapse
Affiliation(s)
- Benjamin Mubemba
- Department of Wildlife Sciences, School of Natural Resources, Copperbelt University, Kitwe, Zambia
- Department of Biomedical Sciences, School of Medicine, Copperbelt University, Ndola, Zambia
| | | | - Katendi Changula
- Department of Paraclinical Studies, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Walter Muleya
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
| | - Lavel C. Moonga
- Division of Collaboration and Education, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Herman M. Chambaro
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Masahiro Kajihara
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yongjin Qiu
- Division of International Research Promotion, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Kyoko Hayashida
- Division of Collaboration and Education, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Catherine G. Sutcliffe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Douglas E. Norris
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | | | | | - Simbarashe Chitanga
- Department of Paraclinical Studies, School of Veterinary Medicine, University of Namibia, Windhoek, Namibia
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
- School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of International Research Promotion, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Disease Control, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
- Africa Centre of Excellence for Infectious Diseases of Humans and Animals, University of Zambia, Lusaka, Zambia
- Global Virus Network, Baltimore, Maryland, United States of America
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Ayato Takada
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Disease Control, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
- Africa Centre of Excellence for Infectious Diseases of Humans and Animals, University of Zambia, Lusaka, Zambia
- * E-mail: (AT); (ES)
| | - Edgar Simulundu
- Macha Research Trust, Choma, Zambia
- Department of Disease Control, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
- * E-mail: (AT); (ES)
| |
Collapse
|
42
|
Endothelium Activation during Severe Yellow Fever Triggers an Intense Cytokine-Mediated Inflammatory Response in the Liver Parenchyma. Pathogens 2022; 11:pathogens11010101. [PMID: 35056050 PMCID: PMC8779659 DOI: 10.3390/pathogens11010101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 01/27/2023] Open
Abstract
Yellow fever (YF) is a pansystemic disease caused by the yellow fever virus (YFV), the prototype species of the family Flaviviridae and genus Flavivirus, and has a highly complex host-pathogen relationship, in which endothelial dysfunction reflects viral disease tropism. In this study, the in situ endothelial response was evaluated. Liver tissue samples were collected from 21 YFV-positive patients who died due to the disease and five flavivirus-negative controls who died of other causes and whose hepatic parenchyma architecture was preserved. Immunohistochemical analysis of tissues in the hepatic parenchyma of YF cases showed significantly higher expression of E-selectin, P-selectin, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and very late antigen-4 in YFV-positive cases than in flavivirus-negative controls. These results indicate that endothelium activation aggravates the inflammatory response by inducing the expression of adhesion molecules that contribute to the rolling, recruitment, migration, and construction of the inflammatory process in the hepatic parenchyma in fatal YF cases.
Collapse
|
43
|
An evaluation of the diagnostic performance characteristics of the Yellow Fever IgM immunochromatographic rapid diagnostic test kit from SD Biosensor in Ghana. PLoS One 2022; 17:e0262312. [PMID: 34995319 PMCID: PMC8741057 DOI: 10.1371/journal.pone.0262312] [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: 06/27/2021] [Accepted: 12/21/2021] [Indexed: 12/03/2022] Open
Abstract
Yellow fever is endemic in Ghana and outbreaks occur periodically. The prodromal signs due to Yellow Fever Virus (YFV) infection are non-specific, making clinical signs unreliable as the sole criteria for diagnosis. Accurate laboratory confirmation of suspected yellow fever cases is therefore vital in surveillance programs. Reporting of ELISA IgM testing results by laboratories can delay due to late arrival of samples from the collection sites as well as limited availability of ELISA kits. In this study, the diagnostic performance characteristics of a rapid immunochromatographic Standard Q Yellow Fever IgM test kit (SD Biosensor) was evaluated for the rapid diagnosis of Yellow Fever infection in Ghana. A panel of 275 sera, comprising 81 confirmed YFV positives and 194 negatives were re-tested in this study using the Standard Q Yellow Fever IgM test kit. Using the CDC/WHO Yellow Fever IgM capture ELISA as a benchmark, the sensitivity, specificity and accuracy of the Standard Q Yellow Fever test kit were 96.3%, 97.9% and 97.5%, respectively. The false positivity rate was 5.1% and there was no cross-reactivity when the Standard Q Yellow Fever test kit was tested against dengue, malaria and hepatitis B and C positive samples. In addition, inter-reader variability and invalid rate were both zero. The results indicate that the diagnostic performance of the Standard Q Yellow Fever IgM test kit on serum or plasma is comparable to the serum IgM detection by ELISA and can be used as a point of care rapid diagnostic test kit for YFV infection in endemic areas.
Collapse
|
44
|
Rampazzo RDCP, Zambenedetti MR, Alexandrino F, Jacomasso T, Tschá MK, de Fillipis AMB, Morello LG, Marchini FK. Development, verification, and validation of an RT-qPCR-based protocol for Yellow Fever diagnosis. Int J Infect Dis 2022; 119:34-37. [PMID: 34990800 DOI: 10.1016/j.ijid.2021.12.361] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Yellow fever (YF) is a public health threat with frequent outbreaks in tropical and subtropical areas, despite the existence of a safe and effective vaccine. The diagnosis of acute infection of the etiologic agent relies mainly on real-time reverse transcription-polymerase chain reaction (RT-qPCR)-based assays. OBJECTIVES The aim of this study was to evaluate and compare this novel protocol for yellow fever virus (YFV) diagnosis against assays developed in-house by reference laboratories for arboviruses. METHODS We developed a novel molecular protocol for the detection of YFV that includes an Internal Control to validate the reaction and an External Control to monitor the RNA extraction efficiency. RESULTS AND DISCUSSION Our assay detects one viral genome per reaction and displays no cross-reactions with dengue (1-4), Zika, or Chikungunya viruses. This novel assay yielded 95% of agreement with the reference method recommended by the Pan American Health Organization when analyzing 204 clinical samples and cultured viruses, these samples were analyzed in 3 different diagnosis centers for arboviruses in Brazil. The data suggest the use of the proposed multiplex assay protocol to do routine tests in a clinical laboratory. This product adds higher specificity and sensitivity in addition to reduced cost per test due to hands-on time and reagent spending.
Collapse
Affiliation(s)
- Rita de Cássia Pontello Rampazzo
- Instituto de Biologia Molecular do Paraná (IBMP), Rua Professor Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil
| | - Miriam Ribas Zambenedetti
- Instituto de Biologia Molecular do Paraná (IBMP), Rua Professor Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil
| | - Fabiana Alexandrino
- Instituto de Biologia Molecular do Paraná (IBMP), Rua Professor Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil
| | - Thiago Jacomasso
- Instituto de Biologia Molecular do Paraná (IBMP), Rua Professor Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil
| | - Marcel Kruchelski Tschá
- Instituto de Biologia Molecular do Paraná (IBMP), Rua Professor Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil
| | - Ana Maria Bispo de Fillipis
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil, 4365, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luis Gustavo Morello
- Instituto de Biologia Molecular do Paraná (IBMP), Rua Professor Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil; Instituto Carlos Chagas (ICC), FIOCRUZ-PR, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil
| | - Fabricio Klerynton Marchini
- Instituto de Biologia Molecular do Paraná (IBMP), Rua Professor Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil; Instituto Carlos Chagas (ICC), FIOCRUZ-PR, Rua Prof. Algacyr Munhoz Mader, 3775 CIC, 81350-010, Curitiba, Paraná, Brazil.
| |
Collapse
|
45
|
Review of -omics studies on mosquito-borne viruses of the Flavivirus genus. Virus Res 2022; 307:198610. [PMID: 34718046 DOI: 10.1016/j.virusres.2021.198610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/18/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023]
Abstract
Arboviruses are transmitted by arthropods (arthropod-borne virus) which can be mosquitoes or other hematophagous arthropods, in which their life cycle occurs before transmission to other hosts. Arboviruses such as Dengue, Zika, Saint Louis Encephalitis, West Nile, Yellow Fever, Japanese Encephalitis, Rocio and Murray Valley Encephalitis viruses are some of the arboviruses transmitted biologically among vertebrate hosts by blood-taking vectors, mainly Aedes and Culex sp., and are associated with neurological, viscerotropic, and hemorrhagic reemerging diseases, posing as significant health and socioeconomic concern, as they become more and more adaptive to new environments, to arthropods vectors and human hosts. One of the main families that include mosquito-borne viruses is Flaviviridae, and here, we review the case of the Flavivirus genus, which comprises the viruses cited above, using a variety of research approaches published in literature, including genomics, transcriptomics, proteomics, metabolomics, etc., to better understand their structures as well as virus-host interactions, which are essential for development of future antiviral therapies.
Collapse
|
46
|
Gava C, Silva TCCD, Lyra DGP, Ardisson KS, Marques CS, Almada GL, Corrêa LMC, Siqueira PC, Rodrigues GAP, Moura LD, Cruz OG, Maciel ELN, Camacho LAB. Prevenção e controle da febre amarela: avaliação de ações de vigilância em área indene no Brasil. CAD SAUDE PUBLICA 2022; 38:e00000521. [DOI: 10.1590/0102-311x00000521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022] Open
Abstract
A partir da reemergência da febre amarela em 2014/2015, o Brasil registrou nos anos sequentes sua maior epidemia de febre amarela das últimas décadas, atingindo principalmente a região sudeste. A febre amarela, doença viral hemorrágica, é causada por um flavivírus, transmitido por mosquitos silvestres (Haemagogus; Sabethes). Na ocorrência do ciclo urbano, erradicado no Brasil desde 1942, a transmissão se dá pelo Aedes aegypti. Primatas não humanos são os principais hospedeiros do vírus e constituem “sentinelas” na vigilância da febre amarela. Este artigo descreve as ações de controle e prevenção desencadeadas durante a epidemia de febre amarela no Estado do Espírito Santo, Brasil, e a implementação da vacinação por meio de um estudo ecológico com abordagem espacial. O estudo evidenciou a falha na detecção de epizootias em primatas não humanos pelos serviços de vigilância do Espírito Santo, sendo simultânea à detecção em humanos. Apresentou a evolução das ações de vacinação, com alcance de 85% de cobertura vacinal geral para o estado em seis meses, sendo heterogênea entre os municípios (de 59% a 122%). Destaca-se que 55% dos municípios com ações de imunização em tempo oportuno, considerando o intervalo adotado para este estudo, não apresentaram casos em humanos. A intensificação das ações de vigilância, interlocução entre as áreas e equipes multidisciplinares na condução da epidemia otimizou a detecção e o diagnóstico dos casos em humanos e viabilizou o controle da epidemia. Foi possível reconhecer avanços, apontar algumas medidas tardias e lacunas na vigilância que necessitam melhorias.
Collapse
Affiliation(s)
- Caroline Gava
- Fundação Oswaldo Cruz, Brazil; Ministério da Saúde, Brazil
| | | | | | | | | | - Gilton Luiz Almada
- Secretaria de Estado da Saúde do Espírito Santo, Brasil; Universidade de Vila Velha, Brazil
| | | | | | | | - Lenildo de Moura
- Organização Pan-Americana da Saúde, Brasil; Organização Mundial da Saúde, Brasil
| | | | | | | |
Collapse
|
47
|
Arantes MF, Seabra VF, Lins PRG, Rodrigues CE, Reichert BV, Silveira MAD, Li HY, Malbouisson LM, Andrade L. Risk Factors for Acute Kidney Injury and Death in Patients Infected With the Yellow Fever Virus During the 2018 Outbreak in São Paulo, Brazil. Kidney Int Rep 2021; 7:601-609. [PMID: 35257072 PMCID: PMC8897308 DOI: 10.1016/j.ekir.2021.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/23/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Introduction Methods Results Conclusion
Collapse
Affiliation(s)
- Márcia Fernanda Arantes
- Division of Nephrology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Victor Faria Seabra
- Division of Nephrology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Paulo Ricardo Gessolo Lins
- Division of Nephrology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Camila Eleuterio Rodrigues
- Division of Nephrology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Bernardo Vergara Reichert
- Division of Nephrology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Ho Yeh Li
- Intensive Care Unit, Department of Infectious and Parasitic Diseases, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Luiz Marcelo Malbouisson
- Division of Anesthesiology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Lúcia Andrade
- Division of Nephrology, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
- Correspondence: Lúcia Andrade, Division of Nephrology, Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3 andar, sala 3310, São Paulo, SP 01246-903, Brazil.
| |
Collapse
|
48
|
Asebe G, Mamo G, Wieland B, Medhin G, Tilahun G, Abegaz WE, Legesse M. Community awareness and experiences of health workers concerning mosquito-borne viral diseases in selected districts of Gambella Region, Southwestern Ethiopia. Infect Ecol Epidemiol 2021; 11:1988453. [PMID: 34745448 PMCID: PMC8567928 DOI: 10.1080/20008686.2021.1988453] [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] [Indexed: 11/06/2022] Open
Abstract
In this study, we assessed community awareness and experiences of health workers about mosquito-borne viral diseases in selected districts of the Gambella Region, South Western Ethiopia. A community and health facility-based qualitative study involving 11 focus group discussions (FGDs) with community dmembers and two FGDs with health workers was conducted between November 2017 to January 2018. A total of 122 community members and 16 health workers participated in the study. All the discussants mentioned malaria, typhoid fever, unknown causes of diarrhea and skin diseases as the major public health problems in the area. Using pictures of Anopheles and Aedes mosquitoes, participants confirmed that both mosquitoes are present in the area. They identified Anopheles as the vector of malaria. However, community discussants could not mention the name of a disease that can be transmitted by Aedes mosquito though they mentioned that Aedes mosquito bites both humans and animals during the day time in forest areas and causes skin itching to humans. Meanwhile, community participants from Pakag, a village bordering South Sudan, expressed concern that Aedes mosquito can cause a malaria-like disease which can kill within a few days. Health workers from Itang health center described that in 2016, an outbreak of an unknown disease that causes fever and jaundice occurred and killed seven individuals in a village called Akula, which is closer to a South Sudan refugee camp. Overall, the findings showed that community members and health workers in the area do not have adequate information on mosquito-borne viral diseases. Creating awareness, improving laboratory services and further epidemiological studies would be important for early warning and preparedness for outbreaks in the area.
Collapse
Affiliation(s)
- Getahun Asebe
- Addis Ababa University College of Veterinary Medicine, Department of Veterinary Microbiology, Immunology and Public Health, Bishoftu, Ethiopia.,College of Agriculture and Natural Resources, Gambella University, Gambella, Ethiopia
| | - Gezahegne Mamo
- Addis Ababa University College of Veterinary Medicine, Department of Veterinary Microbiology, Immunology and Public Health, Bishoftu, Ethiopia
| | - Barbara Wieland
- International Livestock Research Institute, Addis Ababa, Ethiopia
| | - Girmay Medhin
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Getachew Tilahun
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Woldaregay Erku Abegaz
- College of Health Sciences, School of Medicine, Department of Microbiology, Immunology & Parasitology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Mengistu Legesse
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
49
|
Perkins TA, Huber JH, Tran QM, Oidtman RJ, Walters MK, Siraj AS, Moore SM. Burden is in the eye of the beholder: Sensitivity of yellow fever disease burden estimates to modeling assumptions. SCIENCE ADVANCES 2021; 7:eabg5033. [PMID: 34644110 DOI: 10.1126/sciadv.abg5033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Estimates of disease burden are important for setting public health priorities. These estimates involve numerous modeling assumptions, whose uncertainties are not always well described. We developed a framework for estimating the burden of yellow fever in Africa and evaluated its sensitivity to modeling assumptions that are often overlooked. We found that alternative interpretations of serological data resulted in a nearly 20-fold difference in burden estimates (range of central estimates, 8.4 × 104 to 1.5 × 106 deaths in 2021–2030). Uncertainty about the vaccination status of serological study participants was the primary driver of this uncertainty. Even so, statistical uncertainty was even greater than uncertainty due to modeling assumptions, accounting for a total of 87% of variance in burden estimates. Combined with estimates that most infections go unreported (range of 95% credible intervals, 99.65 to 99.99%), our results suggest that yellow fever’s burden will remain highly uncertain without major improvements in surveillance.
Collapse
Affiliation(s)
- T Alex Perkins
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - John H Huber
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Quan M Tran
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel J Oidtman
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Magdalene K Walters
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Amir S Siraj
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sean M Moore
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
50
|
Segura NA, Muñoz AL, Losada-Barragán M, Torres O, Rodríguez AK, Rangel H, Bello F. Minireview: Epidemiological impact of arboviral diseases in Latin American countries, arbovirus-vector interactions and control strategies. Pathog Dis 2021; 79:6354781. [PMID: 34410378 DOI: 10.1093/femspd/ftab043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
Mosquitoes are the most crucial insects in public health due to their vector capacity and competence to transmit pathogens, including arboviruses, bacterias and parasites. Re-emerging and emerging arboviral diseases, such as yellow fever virus (YFV), dengue virus (DENV), zika virus (ZIKV), and chikungunya virus (CHIKV), constitute one of the most critical health public concerns in Latin America. These diseases present a significant incidence within the human settlements increasing morbidity and mortality events. Likewise, among the different genus of mosquito vectors of arboviruses, those of the most significant medical importance corresponds to Aedes and Culex. In Latin America, the mosquito vector species of YFV, DENV, ZIKV, and CHIKV are mainly Aedes aegypti and Ae. Albopictus. Ae. aegypti is recognized as the primary vector in urban environments, whereas Ae. albopictus, recently introduced in the Americas, is more prone to rural settings. This minireview focuses on what is known about the epidemiological impact of mosquito-borne diseases in Latin American countries, with particular emphasis on YFV, DENV, ZIKV and CHIKV, vector mosquitoes, geographic distribution, and vector-arbovirus interactions. Besides, it was analyzed how climate change and social factors have influenced the spread of arboviruses and the control strategies developed against mosquitoes in this continent.
Collapse
Affiliation(s)
- Nidya A Segura
- Faculty of Science, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| | - Ana L Muñoz
- PhD Program of Health Science, Universidad Antonio Nariño (UAN), Bogotá 110231, Colombia
| | | | - Orlando Torres
- Faculty of Veterinary, Universidad Antonio Nariño (UAN), Bogotá 110231, Colombia
| | - Anny K Rodríguez
- Faculty of Science, Universidad Antonio Nariño (UAN), Bogotá 110231, Colombia
| | - Héctor Rangel
- Laboratory of Molecular Virology, Instituto Venezolano de Investigaciones Científicas, Caracas 1204, Venezuela
| | - Felio Bello
- Faculty of Agricultural and Livestock Sciences, Program of Veterinary Medicine, Universidad de La Salle, Bogotá 110141, Colombia
| |
Collapse
|