1
|
Oliveira SRDM, da Cruz EDRM, Prestes NGDO, da Silva FS, de Araújo MTF, Amador Neto OP, Lima MDLG, de Alcântara BN, Dias DD, de Sousa JR, Filho AJM, Casseb LMN, Medeiros DBDA. Histopathological Changes and Immune Response Profile in the Brains of Non-Human Primates Naturally Infected with Yellow Fever Virus. Viruses 2025; 17:386. [PMID: 40143314 PMCID: PMC11946203 DOI: 10.3390/v17030386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
In the history of yellow fever (YF) outbreaks in Brazil, howler monkeys (Alouatta sp.) and marmosets (Callithrix sp.) have been among the most affected genera, exhibiting significant hepatic injuries similar to those seen in humans. However, limited information exists regarding yellow fever virus (YFV) infection in their central nervous system (CNS). To address this gap, an epidemiological study was conducted to assess tissue changes, viral detection, and cytokine profiles in the brains of both neotropical primate species when they are naturally infected with YFV. A total of 22 brain samples from these species (8 from Alouatta sp. and 14 from Callithrix sp.) showing infection with YFV in the liver via immunohistochemistry (IHC) were selected. From them, YFV antigen detection occurred in 35.7% (5/14) of Callithrix sp. brain samples and 87.5% (7/8) of Alouatta sp. samples, with a higher frequency of viral antigen quantification in Callithrix sp. Both species exhibited similar CNS lesions, characterized by congestion, low hemorrhage, limited inflammatory infiltration interstitial and perivascular edema associated with neuronal degeneration, neurophagy, and higher cell death (necrosis and apoptosis) quantification. Pro- and anti-inflammatory cytokine profiles were balanced, with TNF-α and IL-1β playing a key role in inflammation, while IL-10 and IL-13 exhibited a prominent role in immunomodulation, suggesting an anti-inflammatory modulation typical of flaviviruses occurs. This study demonstrates that YFV can induce CNS lesions in neotropical primates, establishing it as a secondary target of viral tropism. These findings highlight the importance of collecting nervous tissue during epizootics, particularly in Callithrix sp., as such tissue is often overlooked despite its critical role in disease monitoring.
Collapse
Affiliation(s)
- Suzana Ribeiro de Melo Oliveira
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| | - Ermelinda do Rosário Moutinho da Cruz
- Department of Clinical and Experimental Pathology, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (E.d.R.M.d.C.); (M.T.F.d.A.); (O.P.A.N.); (M.d.L.G.L.); (A.J.M.F.)
| | - Nelielma Garcia de Oliveira Prestes
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| | - Fábio Silva da Silva
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| | - Marialva Tereza Ferreira de Araújo
- Department of Clinical and Experimental Pathology, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (E.d.R.M.d.C.); (M.T.F.d.A.); (O.P.A.N.); (M.d.L.G.L.); (A.J.M.F.)
| | - Orlando Pereira Amador Neto
- Department of Clinical and Experimental Pathology, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (E.d.R.M.d.C.); (M.T.F.d.A.); (O.P.A.N.); (M.d.L.G.L.); (A.J.M.F.)
| | - Maria de Lourdes Gomes Lima
- Department of Clinical and Experimental Pathology, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (E.d.R.M.d.C.); (M.T.F.d.A.); (O.P.A.N.); (M.d.L.G.L.); (A.J.M.F.)
| | - Bianca Nascimento de Alcântara
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| | - Daniel Damous Dias
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| | - Jorge Rodrigues de Sousa
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| | - Arnaldo Jorge Martins Filho
- Department of Clinical and Experimental Pathology, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (E.d.R.M.d.C.); (M.T.F.d.A.); (O.P.A.N.); (M.d.L.G.L.); (A.J.M.F.)
| | - Livia Medeiros Neves Casseb
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| | - Daniele Barbosa de Almeida Medeiros
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Rodovia BR-316 km 7 s/n—Levilândia, Ananindeua 67030-000, Pará, Brazil; (S.R.d.M.O.); (N.G.d.O.P.); (B.N.d.A.); (D.D.D.); (J.R.d.S.); (D.B.d.A.M.)
| |
Collapse
|
2
|
Ho YL, Nukui Y, Villaça PR, Okazaki E, Tatsui NH, Netto LC, Joelsons D, da Rocha TRF, de Mello Malta F, Pinho JRR, Segurado AAC, Rocha V. Intensive Therapeutic Plasma Exchange-New Approach to Treat and Rescue Patients with Severe Form of Yellow Fever. Trop Med Infect Dis 2025; 10:39. [PMID: 39998043 PMCID: PMC11860207 DOI: 10.3390/tropicalmed10020039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Severe yellow fever (YF) can result in acute liver failure (ALF) and high mortality. The role of therapeutic plasma exchange (TPE) in managing YF-ALF remains unclear. This study evaluated the impact of TPE strategies in severe YF. METHODS This observational case-series study evaluated three groups of patients classified according to treatment: G1 (standard intensive care support [ICS]), G2 (ICS + high-volume-TPE [HV-TPE]), and G3 (ICS + intensive TPE). HV-TPE was performed during 3 consecutive days with extra sessions of one plasma-volume, if necessary, whereas intensive TPE consisted of one plasma volume/session performed twice daily, with additional fresh frozen plasma infusion. Hemostatic agents, including tranexamic acid, platelets, and cryoprecipitate, were administered as needed. TPE was de-escalated based on clinical and laboratory parameters. The primary outcome was mortality. RESULTS Sixty-six patients were included (G1: 41, G2: 11, G3: 14). Groups had similar baseline characteristics. Mortality was significantly lower in G3 (14%) compared to G2 (82%) and G1 (85%) (p < 0.001). Additionally, G3 patients showed a higher frequency of undetectable YF viral load. CONCLUSIONS Intensive TPE is a feasible and effective intervention for severe YF, achieving an 84% reduction in mortality. The limitations of our results are the small sample size, observational and single-center study. Further studies are warranted to elucidate intensive TPE's role in YF management.
Collapse
Affiliation(s)
- Yeh-Li Ho
- Departamento de Infectologia e Medicina Tropical, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil (D.J.)
| | - Youko Nukui
- Serviço de Hematologia, Hemoterapia e Terapia Celular, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Paula Ribeiro Villaça
- Serviço de Hematologia, Hemoterapia e Terapia Celular, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Erica Okazaki
- Serviço de Hematologia, Hemoterapia e Terapia Celular, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Nelson Hidekazu Tatsui
- Serviço de Hematologia, Hemoterapia e Terapia Celular, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Lucas Chaves Netto
- Departamento de Infectologia e Medicina Tropical, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil (D.J.)
| | - Daniel Joelsons
- Departamento de Infectologia e Medicina Tropical, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil (D.J.)
| | - Tania Rubia Flores da Rocha
- Serviço de Hematologia, Hemoterapia e Terapia Celular, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Fernanda de Mello Malta
- LIM07, Departamento de Gastroenterologia, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - João Renato Rebello Pinho
- LIM07, Departamento de Gastroenterologia, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Aluisio Augusto Cotrim Segurado
- Departamento de Infectologia e Medicina Tropical, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil (D.J.)
| | - Vanderson Rocha
- Serviço de Hematologia, Hemoterapia e Terapia Celular, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-000, Brazil
- Fundacao Pro-Sangue, Sao Paulo 05403-000, Brazil
- Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| |
Collapse
|
3
|
McClure M, de Rezende IM, Pereira LS, Dutra MRT, Fradico JRB, Macedo R, Marçal MC, Fonte Boa LSC, Bragato AMC, Faria FADA, Pamplona L, Said RFDC, Calzavara-Silva CE, Ramalho DB, Magalhães CLDB, Alves PA, Gama TDP, Cota GF, Monath TP, Martins-Filho OA, Pascoal-Xavier MA, Teixeira-Carvalho A, Drumond BP, LaBeaud AD. Risk factors associated with in-hospital mortality during yellow fever outbreak in Brazil. Front Med (Lausanne) 2025; 12:1505005. [PMID: 39931562 PMCID: PMC11807806 DOI: 10.3389/fmed.2025.1505005] [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: 10/01/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Objective To characterize the clinical manifestations of yellow fever disease and identify risk factors for mortality. Methods A retrospective study was conducted in the referral center for infectious diseases (Hospital Eduardo de Menezes-HEM) in Belo Horizonte, Minas Gerais, Brazil. Analysis included data from 283 patients with confirmed YF infection older than 13 years old who presented to HEM between January 2017 and June 2018. In-hospital mortality (hypothesis formulated after data collection), demographic factors and clinical and laboratory assessments were used. Results Study patients were mainly men (87.6%), with a median age of 46.0 (IQR 36.5, 57.0). 131 (46.3%) patients were admitted to the ICU, and 62 (22.0%) used invasive mechanical ventilation for a median of 2 days (IQR 1, 3). The median (IQR) total length of stay (LOS) in the ICU was 6 days (IQR 4, 8). The in-hospital mortality rate was 24.0%. Age was significantly higher in fatal (median 49.5, IQR 41.0, 61.0]) than in non-fatal cases [46 (36, 55)] (p < 0.01). Male sex was associated with an increased risk of death (RR 4.66, 95% CI 1.19, 18.2; p < 0.01). Most common symptoms and signs on admission to HEM were fever (31.9%), myalgia (27.8%), jaundice (24.3%), headache (23.9%), abdominal pain (16.1%), vomiting (12.2%), weakness (10.4%), and arthralgias (10.0%). Initial viral load above the cutoff of 4.45 log10 copies/mL was significantly associated with death prior to discharge (OR 12.2; CI 2.83, 92.3). Five factors were significantly related to increased odds of death prior to discharge: log-transformed AST (OR 3.65; CI 2.02, 7.81; p < 0.001), log-transformed INR (OR 7.40; CI 1.31, 33.0; p = 0.010), log-transformed lactate (OR 4.57; CI 1.48, 17.1; p = 0.013), log-transformed WBC (OR 4.33; CI 1.19, 18.5; p = 0.034), and age (OR 1.06; CI 1.01, 1.12; p = 0.026). Conclusions and relevance AST, INR, lactate, WBC, and age are statistically associated with death prior to discharge in YF patients. These clinical markers should be applied to improve patient screening and management during future YF epidemics.
Collapse
Affiliation(s)
- Max McClure
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Izabela Maurício de Rezende
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Pedro Augusto Alves
- Immunology of Viral Diseases, Instituto René Rachou, Fundação Oswaldo Cruz (Fiocruz), Belo Horizonte, Brazil
| | | | | | | | - Olindo Assis Martins-Filho
- Integrated Group of Biomarkers Research, Instituto René Rachou, Fundação Oswaldo Cruz (Fiocruz), Belo Horizonte, Brazil
| | | | - Andrea Teixeira-Carvalho
- Integrated Group of Biomarkers Research, Instituto René Rachou, Fundação Oswaldo Cruz (Fiocruz), Belo Horizonte, Brazil
| | - Betânia Paiva Drumond
- Laboratory of Viruses, Microbiology Department, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - A. Desiree LaBeaud
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
4
|
Puerta-Guardo H, Biering SB, Castillo-Rojas B, DiBiasio-White MJ, Lo NT, Espinosa DA, Warnes CM, Wang C, Cao T, Glasner DR, Beatty PR, Kuhn RJ, Harris E. Flavivirus NS1-triggered endothelial dysfunction promotes virus dissemination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.625931. [PMID: 39651279 PMCID: PMC11623691 DOI: 10.1101/2024.11.29.625931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The Flaviviridae are a family of viruses that include the important arthropod-borne human pathogens dengue virus (DENV), West Nile virus, Zika virus, Japanese encephalitis virus, and yellow fever virus. Flavivirus nonstructural protein 1 (NS1) is essential for virus replication but is also secreted from virus-infected cells. Extracellular NS1 acts as a virulence factor during flavivirus infection in multiple ways, including triggering endothelial dysfunction and vascular leak via interaction with endothelial cells. While the role of NS1 in inducing vascular leak and exacerbating pathogenesis is well appreciated, if and how NS1-triggered endothelial dysfunction promotes virus infection remains obscure. Flaviviruses have a common need to disseminate from circulation into specific tissues where virus-permissive cells reside. Tissue-specific dissemination is associated with disease manifestations of a given flavivirus, but mechanisms dictating virus dissemination are unclear. Here we show that NS1-mediated endothelial dysfunction promotes virus dissemination in vitro and in vivo . In mouse models of DENV infection, we show that anti-NS1 antibodies decrease virus dissemination, while the addition of exogenous NS1 promotes virus dissemination. Using an in vitro system, we show that NS1 promotes virus dissemination in two distinct ways: (1) promoting crossing of barriers and (2) increasing infectivity of target cells in a tissue- and virus-specific manner. The capacity of NS1 to modulate infectivity correlates with a physical association between virions and NS1, suggesting a potential NS1-virion interaction. Taken together, our study indicates that flavivirus NS1 promotes virus dissemination across endothelial barriers, providing an evolutionary basis for virus-triggered vascular leak. Author Summary The Flaviviridae contain numerous medically important human pathogens that cause potentially life-threatening infections. Over half of the world's population is at risk of flavivirus infection, and this number is expected to increase as climate change expands the habitats of the arthropod vectors that transmit these flaviviruses. There are few effective vaccines and no therapeutics approved for prevention or treatment of flavivirus infection, respectively. Given these challenges, understanding how and why flaviviruses cause pathogenesis is critical for identifying targets for therapeutic intervention. The secreted nonstructural protein 1 (NS1) of flaviviruses is a conserved virulence factor that triggers endothelial dysfunction in a tissue-specific manner. It is unknown if this endothelial dysfunction provides any benefit for virus infection. Here we provide evidence that NS1-triggered endothelial dysfunction facilitates virus crossing of endothelial barriers and augments infection of target cells in vitro and promotes virus dissemination in vivo . This study provides an evolutionary explanation for flaviviruses evolving the capacity to trigger barrier dysfunction and highlights NS1 and the pathways governing endothelial dysfunction, as therapeutic targets to prevent flavivirus dissemination.
Collapse
|
5
|
Shinde DP, Plante JA, Scharton D, Mitchell B, Walker J, Azar SR, Campos RK, Sacchetto L, Drumond BP, Vasilakis N, Plante KS, Weaver SC. Potential role of heterologous flavivirus immunity in preventing urban transmission of yellow fever virus. Nat Commun 2024; 15:9728. [PMID: 39523371 PMCID: PMC11551182 DOI: 10.1038/s41467-024-54146-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
During the recent yellow fever (YF) epidemics in Brazil, human cases were attributed to spillover infections via sylvatic mosquito transmission. Despite YF virus (YFV) transmission in major urban centers with insufficient vaccination coverage and abundant populations of the domestic vector, Aedes aegypti, there was no evidence of human-amplified transmission. Furthermore, the historic absence of YF in Asia, despite abundant Ae. aegypti and an immunologically naive human population, is unexplained. We tested the hypothesis that pre-existing, heterologous flavivirus immunity, specifically from dengue (DENV) and Zika (ZIKV) viruses, limits YFV viremia and transmission by Ae. aegypti. We infected cynomolgus macaques with DENV or ZIKV, then challenged them 6-9 months later with YFV. We then measured viremia and disease and allowed Ae. aegypti mosquitoes to feed during peak macaque viremia. Although prior heterologous immunity had variable effects on disease, DENV and ZIKV immunity consistently suppressed YFV viremia. Despite no statistical difference due to a small sample size, the suppression in viremia led to a significant reduction in Ae. aegypti infection and a lack of transmission potential. These results support the hypothesis that, in DENV- and ZIKV-endemic regions such as South America and Asia, human flavivirus immunity suppresses YFV human amplification potential, reducing the risk of urban outbreaks.
Collapse
Affiliation(s)
- Divya P Shinde
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Center for Vector-borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Jessica A Plante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Dionna Scharton
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Brooke Mitchell
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Jordyn Walker
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
| | - Sasha R Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Tissue Engineering, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Rafael K Campos
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lívia Sacchetto
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | - Betânia P Drumond
- Deparment of Microbiology-Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nikos Vasilakis
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Center for Vector-borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Kenneth S Plante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| | - Scott C Weaver
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA.
- Center for Vector-borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
6
|
Wu H, Fujioka Y, Sakaguchi S, Suzuki Y, Nakano T. Electron Tomography as a Tool to Study SARS-CoV-2 Morphology. Int J Mol Sci 2024; 25:11762. [PMID: 39519314 PMCID: PMC11547116 DOI: 10.3390/ijms252111762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel betacoronavirus, is the causative agent of COVID-19, which has caused economic and social disruption worldwide. To date, many drugs and vaccines have been developed for the treatment and prevention of COVID-19 and have effectively controlled the global epidemic of SARS-CoV-2. However, SARS-CoV-2 is highly mutable, leading to the emergence of new variants that may counteract current therapeutic measures. Electron microscopy (EM) is a valuable technique for obtaining ultrastructural information about the intracellular process of virus replication. In particular, EM allows us to visualize the morphological and subcellular changes during virion formation, which would provide a promising avenue for the development of antiviral agents effective against new SARS-CoV-2 variants. In this review, we present our recent findings using transmission electron microscopy (TEM) combined with electron tomography (ET) to reveal the morphologically distinct types of SARS-CoV-2 particles, demonstrating that TEM and ET are valuable tools for visually understanding the maturation status of SARS-CoV-2 in infected cells. This review also discusses the application of EM analysis to the evaluation of genetically engineered RNA viruses.
Collapse
Affiliation(s)
- Hong Wu
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka 565-0871, Japan; (Y.F.); (S.S.); (T.N.)
| | | | | | - Youichi Suzuki
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka 565-0871, Japan; (Y.F.); (S.S.); (T.N.)
| | | |
Collapse
|
7
|
Schult P, Kümmerer BM, Hafner M, Paeschke K. Viral hijacking of hnRNPH1 unveils a G-quadruplex-driven mechanism of stress control. Cell Host Microbe 2024; 32:1579-1593.e8. [PMID: 39094585 DOI: 10.1016/j.chom.2024.07.006] [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: 04/17/2023] [Revised: 04/02/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
Viral genomes are enriched with G-quadruplexes (G4s), non-canonical structures formed in DNA or RNA upon assembly of four guanine stretches into stacked quartets. Because of their critical roles, G4s are potential antiviral targets, yet their function remains largely unknown. Here, we characterize the formation and functions of a conserved G4 within the polymerase coding region of orthoflaviviruses of the Flaviviridae family. Using yellow fever virus, we determine that this G4 promotes viral replication and suppresses host stress responses via interactions with hnRNPH1, a host nuclear protein involved in RNA processing. G4 binding to hnRNPH1 causes its cytoplasmic retention with subsequent impacts on G4-containing tRNA fragments (tiRNAs) involved in stress-mediated reductions in translation. As a result, these host stress responses and associated antiviral effects are impaired. These data reveal that the interplay between hnRNPH1 and both host and viral G4 targets controls the integrated stress response and viral replication.
Collapse
Affiliation(s)
- Philipp Schult
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, 53127 Bonn, Germany; Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Beate Mareike Kümmerer
- Institute of Virology, Medical Faculty, University of Bonn, 53127 Bonn, Germany; German Centre for Infection Research, Partner Site Bonn-Cologne, 53127 Bonn, Germany
| | - Markus Hafner
- RNA Molecular Biology Laboratory, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20892, USA
| | - Katrin Paeschke
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, 53127 Bonn, Germany; Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany.
| |
Collapse
|
8
|
Zarate-Sanchez E, George SC, Moya ML, Robertson C. Vascular dysfunction in hemorrhagic viral fevers: opportunities for organotypic modeling. Biofabrication 2024; 16:032008. [PMID: 38749416 PMCID: PMC11151171 DOI: 10.1088/1758-5090/ad4c0b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
The hemorrhagic fever viruses (HFVs) cause severe or fatal infections in humans. Named after their common symptom hemorrhage, these viruses induce significant vascular dysfunction by affecting endothelial cells, altering immunity, and disrupting the clotting system. Despite advances in treatments, such as cytokine blocking therapies, disease modifying treatment for this class of pathogen remains elusive. Improved understanding of the pathogenesis of these infections could provide new avenues to treatment. While animal models and traditional 2D cell cultures have contributed insight into the mechanisms by which these pathogens affect the vasculature, these models fall short in replicatingin vivohuman vascular dynamics. The emergence of microphysiological systems (MPSs) offers promising avenues for modeling these complex interactions. These MPS or 'organ-on-chip' models present opportunities to better mimic human vascular responses and thus aid in treatment development. In this review, we explore the impact of HFV on the vasculature by causing endothelial dysfunction, blood clotting irregularities, and immune dysregulation. We highlight how existing MPS have elucidated features of HFV pathogenesis as well as discuss existing knowledge gaps and the challenges in modeling these interactions using MPS. Understanding the intricate mechanisms of vascular dysfunction caused by HFV is crucial in developing therapies not only for these infections, but also for other vasculotropic conditions like sepsis.
Collapse
Affiliation(s)
- Evelyn Zarate-Sanchez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| |
Collapse
|
9
|
Naveed A, Eertink LG, Wang D, Li F. Lessons Learned from West Nile Virus Infection:Vaccinations in Equines and Their Implications for One Health Approaches. Viruses 2024; 16:781. [PMID: 38793662 PMCID: PMC11125849 DOI: 10.3390/v16050781] [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: 03/11/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Humans and equines are two dead-end hosts of the mosquito-borne West Nile virus (WNV) with similar susceptibility and pathogenesis. Since the introduction of WNV vaccines into equine populations of the United States of America (USA) in late 2002, there have been only sporadic cases of WNV infection in equines. These cases are generally attributed to unvaccinated and under-vaccinated equines. In contrast, due to the lack of a human WNV vaccine, WNV cases in humans have remained steadily high. An average of 115 deaths have been reported per year in the USA since the first reported case in 1999. Therefore, the characterization of protective immune responses to WNV and the identification of immune correlates of protection in vaccinated equines will provide new fundamental information about the successful development and evaluation of WNV vaccines in humans. This review discusses the comparative epidemiology, transmission, susceptibility to infection and disease, clinical manifestation and pathogenesis, and immune responses of WNV in humans and equines. Furthermore, prophylactic and therapeutic strategies that are currently available and under development are described. In addition, the successful vaccination of equines against WNV and the potential lessons for human vaccine development are discussed.
Collapse
Affiliation(s)
| | | | | | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546, USA; (A.N.); (L.G.E.); (D.W.)
| |
Collapse
|
10
|
Shinde DP, Plante JA, Scharton D, Mitchell B, Walker J, Azar SR, Campos RK, Sacchetto L, Drumond BP, Vasilakis N, Plante KS, Weaver SC. Yellow Fever Emergence: Role of Heterologous Flavivirus Immunity in Preventing Urban Transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583168. [PMID: 38463973 PMCID: PMC10925309 DOI: 10.1101/2024.03.03.583168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
During major, recent yellow fever (YF) epidemics in Brazil, human cases were attributed only to spillover infections from sylvatic transmission with no evidence of human amplification. Furthermore, the historic absence of YF in Asia, despite abundant peridomestic Aedes aegypti and naive human populations, represents a longstanding enigma. We tested the hypothesis that immunity from dengue (DENV) and Zika (ZIKV) flaviviruses limits YF virus (YFV) viremia and transmission by Ae. aegypti . Prior DENV and ZIKV immunity consistently suppressed YFV viremia in experimentally infected macaques, leading to reductions in Ae. aegypti infection when mosquitoes were fed on infected animals. These results indicate that, in DENV- and ZIKV-endemic regions such as South America and Asia, flavivirus immunity suppresses YFV human amplification potential, reducing the risk of urban outbreaks. One-Sentence Summary Immunity from dengue and Zika viruses suppresses yellow fever viremia, preventing infection of mosquitoes and reducing the risk of epidemics.
Collapse
|
11
|
Coelho Ferraz A, Bueno da Silva Menegatto M, Lameira Souza Lima R, Samuel Ola-Olub O, Caldeira Costa D, Carlos de Magalhães J, Maurício Rezende I, Desiree LaBeaud A, P Monath T, Augusto Alves P, Teixeira de Carvalho A, Assis Martins-Filho O, P Drumond B, Magalhães CLDB. Yellow fever virus infection in human hepatocyte cells triggers an imbalance in redox homeostasis with increased reactive oxygen species production, oxidative stress, and decreased antioxidant enzymes. Free Radic Biol Med 2024; 213:266-273. [PMID: 38278309 PMCID: PMC10911966 DOI: 10.1016/j.freeradbiomed.2024.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 01/28/2024]
Abstract
Yellow fever (YF) presents a wide spectrum of severity, with clinical manifestations in humans ranging from febrile and self-limited to fatal cases. Although YF is an old disease for which an effective and safe vaccine exists, little is known about the viral- and host-specific mechanisms that contribute to liver pathology. Several studies have demonstrated that oxidative stress triggered by viral infections contributes to pathogenesis. We evaluated whether yellow fever virus (YFV), when infecting human hepatocytes cells, could trigger an imbalance in redox homeostasis, culminating in oxidative stress. YFV infection resulted in a significant increase in reactive oxygen species (ROS) levels from 2 to 4 days post infection (dpi). When measuring oxidative parameters at 4 dpi, YFV infection caused oxidative damage to lipids, proteins, and DNA, evidenced by an increase in lipid peroxidation/8-isoprostane, carbonyl protein, and 8-hydroxy-2'-deoxyguanosine, respectively. Furthermore, there was a significant reduction in the activity of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx), in addition to a reduction in the ratio of reduced to oxidized glutathione (GSH/GSSG), indicating a pro-oxidant environment. However, no changes were observed in the enzymatic activity of the enzyme catalase (CAT) or in the gene expression of SOD isoforms (1/2/3), CAT, or GPx. Therefore, our results show that YFV infection generates an imbalance in redox homeostasis, with the overproduction of ROS and depletion of antioxidant enzymes, which induces oxidative damage to cellular constituents. Moreover, as it has been demonstrated that oxidative stress is a conspicuous event in YFV infection, therapeutic strategies based on antioxidant biopharmaceuticals may be new targets for the treatment of YF.
Collapse
Affiliation(s)
- Ariane Coelho Ferraz
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Marília Bueno da Silva Menegatto
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Rafaela Lameira Souza Lima
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Oluwashola Samuel Ola-Olub
- Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Daniela Caldeira Costa
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - José Carlos de Magalhães
- Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Departamento de Química, Biotecnologia e Engenharia de Bioprocessos, Universidade Federal de São João del-Rei, Ouro Branco, Minas Gerais, Brazil
| | - Izabela Maurício Rezende
- Pandemic Preparedenss Hub, Divison of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Angelle Desiree LaBeaud
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, California, United States
| | | | - Pedro Augusto Alves
- Imunologia de Doenças Virais, Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Andréa Teixeira de Carvalho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Betânia P Drumond
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cintia Lopes de Brito Magalhães
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
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
|
Ferreira MS, Martins LC, de Melo KFL, da Silva WB, Imbeloni AA, Muniz JAPC, de Oliveira CF, Freitas MNO, Dos Santos ÉB, Chagas LL, Luz MBM, de Queiroz LAD, Tesh RB, Vasconcelos PFC. Experimental Yellow Fever in the Squirrel Monkey ( Saimiri spp.): Hematological, Biochemical, and Immunological Findings. Viruses 2023; 15:v15030613. [PMID: 36992323 PMCID: PMC10052740 DOI: 10.3390/v15030613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 03/31/2023] Open
Abstract
Between 2016 and 2018, Brazil experienced the largest sylvatic epidemic of yellow fever virus (YFV). Despite to the magnitude and rapid spread of the epidemic, little is known about YFV dispersion. The study evaluated whether the squirrel monkey is a good model for yellow fever (YF) studies. Methods: Ten animals were infected with 1 × 106 PFU/mL of YFV, with one negative control. Blood samples were collected daily during the first 7 days and at 10, 20 and 30 days post infection (dpi) for detection of viral load and cytokines by RT-qPCR; measurements of AST, ALT, urea and creatinine were taken; IgM/IgG antibodies were detected by ELISA, and hemagglutination inhibition and neutralization tests were performed. The animals exhibited fever, flushed appearance, vomiting and petechiae, and one animal died. Viremia was detected between 1 and 10 dpi, and IgM/IgG antibodies appeared between 4 and 30 dpi. The levels of AST, ALT and urea increased. The immune responses were characterized by expression of S100 and CD11b cells; endothelial markers (VCAM-1, ICAM-1 and VLA-4), cell death and stress (Lysozyme and iNOS); and pro-inflammatory cytokines (IL-8, TNF-α, and IFN-γ) and anti-inflammatory cytokines (IL-10 and TGF-β). The squirrel monkeys showed changes similar to those described in humans with YF, and are a good experimental model for the study of YF.
Collapse
Affiliation(s)
- Milene S Ferreira
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
- Postgraduate Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém 66075110, PA, Brazil
| | - Lívia C Martins
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | - Karla F L de Melo
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | | | - Aline A Imbeloni
- National Primate Center, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | | | - Camille F de Oliveira
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | - Maria Nazaré O Freitas
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | - Éder B Dos Santos
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | - Liliane L Chagas
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | - Márcia B M Luz
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | - Luiz A D de Queiroz
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
| | - Robert B Tesh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Pedro F C Vasconcelos
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030000, PA, Brazil
- Department of Pathology, Pará State University, Belém 66050540, PA, Brazil
| |
Collapse
|
14
|
Mechanisms of Neuroinvasion and Neuropathogenesis by Pathologic Flaviviruses. Viruses 2023; 15:v15020261. [PMID: 36851477 PMCID: PMC9965671 DOI: 10.3390/v15020261] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/07/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Flaviviruses are present on every continent and cause significant morbidity and mortality. In many instances, severe cases of infection with flaviviruses involve the invasion of and damage to the central nervous system (CNS). Currently, there are several mechanisms by which it has been hypothesized flaviviruses reach the brain, including the disruption of the blood-brain barrier (BBB) which acts as a first line of defense by blocking the entry of many pathogens into the brain, passing through the BBB without disruption, as well as travelling into the CNS through axonal transport from peripheral nerves. After flaviviruses have entered the CNS, they cause different neurological symptoms, leading to years of neurological sequelae or even death. Similar to neuroinvasion, there are several identified mechanisms of neuropathology, including direct cell lysis, blockage of the cell cycle, indication of apoptosis, as well as immune induced pathologies. In this review, we aim to summarize the current knowledge in the field of mechanisms of both neuroinvasion and neuropathogenesis during infection with a variety of flaviviruses and examine the potential contributions and timing of each discussed pathway.
Collapse
|
15
|
Frassetto FP, Rosemberg S. Neuropathology of yellow fever autopsy cases. Trop Dis Travel Med Vaccines 2023; 9:1. [PMID: 36707912 PMCID: PMC9883951 DOI: 10.1186/s40794-022-00187-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 12/14/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Yellow fever is a viral hemorrhagic fever caused by yellow fever virus, a mosquito-borne flavivirus. Despite an effective vaccine, major outbreaks continue to occur around the world. Even though it is not a proven neurotropic virus, neurological symptoms in more severe clinical forms are frequent. The understanding of this apparent paradox is still rarely addressed in literature. METHODS The brains of thirty-eight patients with yellow fever confirmed by RT-PCR, who underwent autopsy, were analyzed morphologically to identify and characterize neuropathological changes. The data were compared with brains collected from individuals without the disease, as a control group. Both cases and controls were subdivided according to the presence or absence of co-concurrent septic shock, to exclude changes of the sepsis associated encephalopathy. To verify possible morphological differences between the yellow fever cases groups, between the control groups, and between the cases and the controls, we applied the statistical tests Fisher's exact test and chi-square, with p values < 0.05 considered statistically significant. RESULTS All cases and controls presented, at least focally, neuropathological changes, which included edema, meningeal and parenchymal inflammatory infiltrate and hemorrhages, and perivascular inflammatory infiltrate. We did not find an unequivocal aspect of encephalitis. The only parameter that, after statistical analysis, can be attributed to yellow fever was the perivascular inflammatory infiltrate. CONCLUSIONS The neuropathological findings are sufficient to justify the multiple clinical neurologic disturbances detected in the YF cases. Since most of the parameters evaluated did not show statistically significant difference between cases and controls, an explanation for most of the neuropathological findings may be the vascular changes, consequent to shock induced endotheliopathy, associated with stimulation of the immune system inherent to systemic infectious processes. The statistical difference obtained in yellow fever cases regarding perivascular infiltrate can be can be explained by the immune activation inherent to the condition.
Collapse
Affiliation(s)
- Fernando Pereira Frassetto
- grid.11899.380000 0004 1937 0722Department of Pathology, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, SP Brazil ,grid.261331.40000 0001 2285 7943Present Address: Department of Radiation Oncology, Ohio State University, OH Columbus, United States of America
| | - Sergio Rosemberg
- grid.11899.380000 0004 1937 0722Department of Pathology, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, SP Brazil
| |
Collapse
|
16
|
Bailey AL, Diamond MS. Hepatopathology of flaviviruses. J Hepatol 2022; 77:1711-1713. [PMID: 35981935 DOI: 10.1016/j.jhep.2022.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/04/2022]
Affiliation(s)
- Adam L Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Heath, University of Wisconsin-Madison, Madison, WI, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
17
|
Exotic viral hepatitis: A review on epidemiology, pathogenesis, and treatment. J Hepatol 2022; 77:1431-1443. [PMID: 35817222 DOI: 10.1016/j.jhep.2022.06.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/14/2022] [Accepted: 06/19/2022] [Indexed: 12/04/2022]
Abstract
Certain "exotic" viruses are known to cause clinical diseases with potential liver involvement. These include viruses, beyond regular hepatotropic viruses (hepatitis A, -B(D), -C, -E, cytomegalovirus, Epstein-Barr virus), that can be found in (sub)tropical areas and can cause "exotic viral hepatitis". Transmission routes typically involve arthropods (Crimean Congo haemorrhagic fever, dengue, Rift Valley fever, yellow fever). However, some of these viruses are transmitted by the aerosolised excreta of rodents (Hantavirus, Lassa fever), or via direct contact or contact with bodily fluids (Ebola). Although some exotic viruses are associated with high fatality rates, such as Ebola for example, the clinical presentation of most exotic viruses can range from mild flu-like symptoms, in most cases, right through to being potentially fatal. A smaller percentage of people develop severe disease with haemorrhagic fever, possibly with (fulminant) hepatitis. Liver involvement is often caused by direct tropism for hepatocytes and Kupffer cells, resulting in virus-mediated and/or immune-mediated necrosis. In all exotic hepatitis viruses, PCR is the most sensitive diagnostic method. The determination of IgM/IgG antibodies is a reasonable alternative, but cross-reactivity can be a problem in the case of flaviviruses. Licenced vaccines are available for yellow fever and Ebola, and they are currently under development for dengue. Therapy for exotic viral hepatitis is predominantly supportive. To ensure that preventive measures can be introduced to control possible outbreaks, the timely detection of these viruses is very important.
Collapse
|
18
|
Flórez-Álvarez L, de Souza EE, Botosso VF, de Oliveira DBL, Ho PL, Taborda CP, Palmisano G, Capurro ML, Pinho JRR, Ferreira HL, Minoprio P, Arruda E, de Souza Ferreira LC, Wrenger C, Durigon EL. Hemorrhagic fever viruses: Pathogenesis, therapeutics, and emerging and re-emerging potential. Front Microbiol 2022; 13:1040093. [PMID: 36386719 PMCID: PMC9640979 DOI: 10.3389/fmicb.2022.1040093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/06/2022] [Indexed: 01/29/2023] Open
Abstract
Hemorrhagic fever viruses (HFVs) pose a threat to global public health owing to the emergence and re-emergence of highly fatal diseases. Viral hemorrhagic fevers (VHFs) caused by these viruses are mostly characterized by an acute febrile syndrome with coagulation abnormalities and generalized hemorrhage that may lead to life-threatening organ dysfunction. Currently, the events underlying the viral pathogenicity associated with multiple organ dysfunction syndrome still underexplored. In this minireview, we address the current knowledge of the mechanisms underlying VHFs pathogenesis and discuss the available development of preventive and therapeutic options to treat these infections. Furthermore, we discuss the potential of HFVs to cause worldwide emergencies along with factors that favor their spread beyond their original niches.
Collapse
Affiliation(s)
| | | | | | | | - Paulo Lee Ho
- Virology Laboratory, Butantan Institute, São Paulo, Brazil
| | | | - Giuseppe Palmisano
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - João Renato Rebello Pinho
- Albert Einstein Institute for Teaching and Research (IIEP), Hospital Israelita Albert Einstein, São Paulo, Brazil,Hospital das Clínicas da Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Helena Lage Ferreira
- Faculty of Animal Science and Food Engineering, University of São Paulo, São Paulo, Brazil
| | | | - Eurico Arruda
- Faculty of Medicine of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Luís Carlos de Souza Ferreira
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,Scientific Platform Pasteur-USP, São Paulo, Brazil
| | - Carsten Wrenger
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,*Correspondence: Carsten Wrenger, ; Edison Luiz Durigon,
| | - Edison Luiz Durigon
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,Scientific Platform Pasteur-USP, São Paulo, Brazil,*Correspondence: Carsten Wrenger, ; Edison Luiz Durigon,
| |
Collapse
|
19
|
de Rezende IM, Cenachi ARC, Costa TA, Oliveira GFG, Rabelo L, Menezes LM, Penido I, Pereira LS, Arruda MS, Gonç alves AP, Alves PA, Kroon EG, Calzavara-Silva CE, Ramalho DB, Martins-Filho OA, Teixeira-Carvalho A, LaBeaud AD, Drumond BP. Wild-type Yellow fever virus in cerebrospinal fluid from fatal cases in Brazil, 2018. FRONTIERS IN VIROLOGY (LAUSANNE, SWITZERLAND) 2022; 2:936191. [PMID: 37461745 PMCID: PMC10351615 DOI: 10.3389/fviro.2022.936191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Yellow fever virus (YFV) is the causative agent of yellow fever (YF), a hemorrhagic and viscerotropic acute disease. Severe YF has been described in approximately 15-25% of YF patients, with 20-50% of severe YF cases being fatal. Here we analyzed cerebrospinal fluid (CSF) samples collected during the YF outbreak in Brazil in 2018, aiming to investigate CNS neuroinvasion in fatal YFV cases. YFV RNA was screened by RT-qPCR targeting the 3'UTR region of the YFV genome in CSF. CSF samples were tested for the presence of anti-YFV IgM and neutralizing antibodies, coupled with routine laboratory examinations. Among the 13 patients studied, we detected anti-YFV IgM in CSF from eight patients and YFV RNA in CSF from five patients. YFV RNA genomic load in CSF samples ranged from 1.75×103 to 5.42×103 RNA copies/mL. We genotyped YFV from three CSF samples that grouped with other YFV samples from the 2018 outbreak in Brazil within the South-American I genotype. Even though descriptions of neurologic manifestations due to wild type YFV (WT-YFV) infection are rare, since the last YF outbreak in Brazil in 2017-2018, a few studies have demonstrated WT-YFV RNA in CSF samples from YF fatal cases. Serological tests indicated the presence of IgM and neutralizing antibodies against YFV in CSF samples from two patients. Although the presence of viral RNA, IgM and neutralizing antibodies in CSF samples could indicate neuroinvasiveness, further studies are needed to better elucidate the role of YFV neuroinvasion and possible impacts in disease pathogenesis.
Collapse
Affiliation(s)
- Izabela Mauricio de Rezende
- Laboratory of Viruses, Microbiology Department, Biological Sciences Institute, Federal University of Minas Gerais, Minas Gerais, Brazil
- Department of Pediatrics, Division of Infectious Disease, Stanford University School of Medicine, Stanford, CA, United States
| | | | - Thais Alkifeles Costa
- Laboratory of Viruses, Microbiology Department, Biological Sciences Institute, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Gabriela Fernanda Garcia Oliveira
- Laboratory of Viruses, Microbiology Department, Biological Sciences Institute, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Livia Rabelo
- Eduardo de Menezes Hospital, Belo Horizonte, Brazil
| | | | | | - Leonardo Soares Pereira
- Eduardo de Menezes Hospital, Belo Horizonte, Brazil
- Bendigo Heath Hospital, Bendigo, VIC, Australia
| | - Matheus Soares Arruda
- Laboratory of Viruses, Microbiology Department, Biological Sciences Institute, Federal University of Minas Gerais, Minas Gerais, Brazil
| | | | - Pedro Augusto Alves
- Immunology of Viruses Diseases, René Rachou Institute, Oswaldo Cruz Foundation/FIOCRUZ, Minas Gerais, Brazil
| | - Erna Geessien Kroon
- Laboratory of Viruses, Microbiology Department, Biological Sciences Institute, Federal University of Minas Gerais, Minas Gerais, Brazil
| | | | | | - Olindo Assis Martins-Filho
- Integrated Group of Biomarkers Research, René Rachou Institute, Oswaldo Cruz Foundation/FIOCRUZ, Minas Gerais, Brazil
| | - Andrea Teixeira-Carvalho
- Integrated Group of Biomarkers Research, René Rachou Institute, Oswaldo Cruz Foundation/FIOCRUZ, Minas Gerais, Brazil
| | - A. Desiree LaBeaud
- Department of Pediatrics, Division of Infectious Disease, Stanford University School of Medicine, Stanford, CA, United States
| | - Betânia Paiva Drumond
- Laboratory of Viruses, Microbiology Department, Biological Sciences Institute, Federal University of Minas Gerais, Minas Gerais, Brazil
| |
Collapse
|
20
|
Shinde DP, Plante JA, Plante KS, Weaver SC. Yellow Fever: Roles of Animal Models and Arthropod Vector Studies in Understanding Epidemic Emergence. Microorganisms 2022; 10:1578. [PMID: 36013996 PMCID: PMC9412558 DOI: 10.3390/microorganisms10081578] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 02/08/2023] Open
Abstract
Yellow fever virus (YFV) is a mosquito-borne flavivirus circulating throughout the tropical and sub-tropical regions of Africa and South America. It is responsible for an estimated 30,000 deaths annually, and while there is a highly successful vaccine, coverage is incomplete, and there is no approved treatment for YFV infection. Despite advancements in the field, animal models for YFV infection remain scarce, and care must be taken to select an appropriate model for a given hypothesis. Small animal models require either adapted YFV strains or immunocompromised hosts. Non-human primates (NHPs) recapitulate human disease, but they require specialized facilities and training, are often in short supply and cost-prohibitive, and can present ethical concerns. The limitations in studying the mosquito vectors for YFV infection include inconsistency in the laboratory environment, the requirement for a high containment insectary, and difficulty in maintaining sylvatic mosquitoes. In this review, we discuss the roles of animal models and arthropod vector studies in understanding epidemic emergence.
Collapse
Affiliation(s)
- Divya P. Shinde
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jessica A. Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kenneth S. Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Scott C. Weaver
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
21
|
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
|
22
|
Reichling J. Antiviral and Virucidal Properties of Essential Oils and Isolated Compounds - A Scientific Approach. PLANTA MEDICA 2022; 88:587-603. [PMID: 34144626 DOI: 10.1055/a-1382-2898] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Essential oils and isolated essential oil compounds are known to exert various pharmacological effects, such as antibacterial, antifungal, antiviral, anti-inflammatory, anti-immunomodulatory, antioxidant, and wound healing effects. Based on selected articles, this review deals with the potential antiviral and virucidal activities of essential oils and essential oil compounds together with their mechanism of action as well as in silico studies involving viral and host cell-specific target molecules that are indispensable for virus cell adsorption, penetration, and replication. The reported in vitro and in vivo studies highlight the baseline data about the latest findings of essential oils and essential oil compounds antiviral and virucidal effects on enveloped and non-enveloped viruses, taking into account available biochemical and molecular biological tests. The results of many in vitro studies revealed that several essential oils and essential oil compounds from different medicinal and aromatic plants are potent antiviral and virucidal agents that inhibit viral progeny by blocking different steps of the viral infection/replication cycle of DNA and RNA viruses in various host cell lines. Studies in mice infected with viruses causing respiratory diseases showed that different essential oils and essential oil compounds were able to prolong the life of infected animals, reduce virus titers in brain and lung tissues, and significantly inhibit the synthesis of proinflammatory cytokines and chemokines. In addition, some in vitro studies on hydrophilic nano-delivery systems encapsulating essential oils/essential oil compounds exhibited a promising way to improve the chemical stability and enhance the water solubility, bioavailabilty, and antiviral efficacy of essential oils and essential oil compounds.
Collapse
Affiliation(s)
- Jürgen Reichling
- Formerly Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
23
|
Eberle RJ, Olivier DS, Amaral MS, Pacca CC, Nogueira ML, Arni RK, Willbold D, Coronado MA. Riboflavin, a Potent Neuroprotective Vitamin: Focus on Flavivirus and Alphavirus Proteases. Microorganisms 2022; 10:1331. [PMID: 35889050 PMCID: PMC9315535 DOI: 10.3390/microorganisms10071331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 12/01/2022] Open
Abstract
Several neurotropic viruses are members of the flavivirus and alphavirus families. Infections caused by these viruses may cause long-term neurological sequelae in humans. The continuous emergence of infections caused by viruses around the world, such as the chikungunya virus (CHIKV) (Alphavirus genus), the zika virus (ZIKV) and the yellow fever virus (YFV) (both of the Flavivirus genus), warrants the development of new strategies to combat them. Our study demonstrates the inhibitory potential of the water-soluble vitamin riboflavin against NS2B/NS3pro of ZIKV and YFV and nsP2pro of CHIKV. Riboflavin presents a competitive inhibition mode with IC50 values in the medium µM range of 79.4 ± 5.0 µM for ZIKV NS2B/NS3pro and 45.7 ± 2.9 μM for YFV NS2B/NS3pro. Against CHIKV nsP2pro, the vitamin showed a very strong effect (93 ± 5.7 nM). The determined dissociation constants (KD) are significantly below the threshold value of 30 µM. The ligand binding increases the thermal stability between 4 °C and 8 °C. Unexpectedly, riboflavin showed inhibiting activity against another viral protein; the molecule was also able to inhibit the viral entry of CHIKV. Molecular dynamics simulations indicated great stability of riboflavin in the protease active site, which validates the repurposing of riboflavin as a promising molecule in drug development against the viruses presented here.
Collapse
Affiliation(s)
- Raphael J. Eberle
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany;
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße, 40225 Düsseldorf, Germany
| | - Danilo S. Olivier
- Center of Integrated Sciences, Campus Cimba, Federal University of Tocantins, Araguaína 77824-838, TO, Brazil;
| | - Marcos S. Amaral
- Institute of Physics, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil;
| | - Carolina C. Pacca
- Instituto Superior de Educação Ceres, FACERES Medical School, São José do Rio Preto 15090-305, SP, Brazil;
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto-FAMERP, São José do Rio Preto 15090-000, SP, Brazil;
| | - Mauricio L. Nogueira
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto-FAMERP, São José do Rio Preto 15090-000, SP, Brazil;
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Raghuvir K. Arni
- Multiuser Center for Biomolecular Innovation, Department of Physics, IBILCE, São Paulo State University, São Jose do Rio Preto 15054-000, SP, Brazil;
| | - Dieter Willbold
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany;
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße, 40225 Düsseldorf, Germany
- JuStruct: Jülich Centre for Structural Biology, Forchungszentrum Jülich, 52428 Jülich, Germany
| | - Monika A. Coronado
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany;
| |
Collapse
|
24
|
Evaluation of Whatman FTA cards for the preservation of yellow fever virus RNA for use in molecular diagnostics. PLoS Negl Trop Dis 2022; 16:e0010487. [PMID: 35704565 PMCID: PMC9200311 DOI: 10.1371/journal.pntd.0010487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/10/2022] [Indexed: 12/31/2022] Open
Abstract
Yellow fever virus (YFV) is a flavivirus that frequently causes outbreaks of hemorrhagic fever in Africa and South America and is considered a reemerging public health threat. Accurate diagnosis of yellow fever (YF) disease is critical as one confirmed case constitutes an outbreak and may trigger a mass vaccination campaign. Highly sensitive and specific molecular diagnostics have been developed; however, these assays require maintenance of cold-chain during transport of specimens to prevent the degradation of viral RNA prior to testing. Such cold-chain requirements are difficult to meet in some regions. In this study, we investigated Whatman FTA cards as an alternative stabilization method of YFV RNA for use in molecular diagnosis. Using contrived specimens, linear regression analysis showed that RNA detection from a single 6mm FTA card punch was significantly less sensitive than traditional RNA extraction; however, pooling RNA extracted from two FTA punches significantly lowered the limit of detection to be equal to that of the traditional RNA extraction gold standard. In experiments addressing the ability of FTA card methodology to stabilize YFV RNA at variable temperature, RNA could be detected for more than two weeks following storage at 25°C. Even more promising, YFV RNA was detectable on cards held at 37°C from two days to over two weeks depending on viral input. FTA cards were also shown to stabilize YFV RNA at high humidity if cards were desiccated prior to inoculation. These results support that FTA cards could be cost effective and easy to use in molecular diagnosis of YF, preserving viral RNA to allow for positive diagnoses in situations where maintaining cold-chain is not feasible.
Collapse
|
25
|
Desantis J, Felicetti T, Cannalire R. An overview on small molecules acting as broad spectrum-agents for yellow fever infection. Expert Opin Drug Discov 2022; 17:755-773. [PMID: 35638299 DOI: 10.1080/17460441.2022.2084529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Yellow Fever virus (YFV) is a mosquito-borne flavivirus, endemic in 47 countries in Africa and South America, which causes febrile symptoms that can evolve in 15% of the patients to serious haemorrhagic conditions, liver injury, and multiorgan failure. Although a highly effective vaccine (YF-17D vaccine) is available, to date, no antiviral drugs have been approved for the prevention and treatment of YFV infections. AREAS COVERED This review article focuses on the description of viral targets that have been considered within YFV and flavivirus drug discovery studies and on the most relevant candidates reported so far that elicit broad-spectrum inhibition against relevant strains and mutants of YFV. EXPERT OPINION Considering the growing interest on (re)emerging vector-borne viral infections, it is expected that flavivirus drug discovery will quickly deliver potential candidates for clinical evaluation. Due to similarity among flaviviral targets, several candidates identified against different flaviviruses have shown broad-spectrum activity, thus exhibiting anti-YFV activity, as well. In this regard, it would be desirable to routinely include the assessment of antiviral activity against different YFV strains. On the other hand, the development of host targeting agents are still at an initial stage and deserve further focused efforts.
Collapse
Affiliation(s)
- Jenny Desantis
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123, Perugia, Italy
| | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Rolando Cannalire
- Department of Pharmacy, University of Napoli "Federico II", Via D. Montesano 49, 80131, Napoli, Italy
| |
Collapse
|
26
|
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
|
27
|
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: 0.5] [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
|
28
|
Viral and Prion Infections Associated with Central Nervous System Syndromes in Brazil. Viruses 2021; 13:v13071370. [PMID: 34372576 PMCID: PMC8310075 DOI: 10.3390/v13071370] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Virus-induced infections of the central nervous system (CNS) are among the most serious problems in public health and can be associated with high rates of morbidity and mortality, mainly in low- and middle-income countries, where these manifestations have been neglected. Typically, herpes simplex virus 1 and 2, varicella-zoster, and enterovirus are responsible for a high number of cases in immunocompetent hosts, whereas other herpesviruses (for example, cytomegalovirus) are the most common in immunocompromised individuals. Arboviruses have also been associated with outbreaks with a high burden of neurological disorders, such as the Zika virus epidemic in Brazil. There is a current lack of understanding in Brazil about the most common viruses involved in CNS infections. In this review, we briefly summarize the most recent studies and findings associated with the CNS, in addition to epidemiological data that provide extensive information on the circulation and diversity of the most common neuro-invasive viruses in Brazil. We also highlight important aspects of the prion-associated diseases. This review provides readers with better knowledge of virus-associated CNS infections. A deeper understanding of these infections will support the improvement of the current surveillance strategies to allow the timely monitoring of the emergence/re-emergence of neurotropic viruses.
Collapse
|
29
|
McEntire CRS, Song KW, McInnis RP, Rhee JY, Young M, Williams E, Wibecan LL, Nolan N, Nagy AM, Gluckstein J, Mukerji SS, Mateen FJ. Neurologic Manifestations of the World Health Organization's List of Pandemic and Epidemic Diseases. Front Neurol 2021; 12:634827. [PMID: 33692745 PMCID: PMC7937722 DOI: 10.3389/fneur.2021.634827] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/25/2021] [Indexed: 01/02/2023] Open
Abstract
The World Health Organization (WHO) monitors the spread of diseases globally and maintains a list of diseases with epidemic or pandemic potential. Currently listed diseases include Chikungunya, cholera, Crimean-Congo hemorrhagic fever, Ebola virus disease, Hendra virus infection, influenza, Lassa fever, Marburg virus disease, Neisseria meningitis, MERS-CoV, monkeypox, Nipah virus infection, novel coronavirus (COVID-19), plague, Rift Valley fever, SARS, smallpox, tularemia, yellow fever, and Zika virus disease. The associated pathogens are increasingly important on the global stage. The majority of these diseases have neurological manifestations. Those with less frequent neurological manifestations may also have important consequences. This is highlighted now in particular through the ongoing COVID-19 pandemic and reinforces that pathogens with the potential to spread rapidly and widely, in spite of concerted global efforts, may affect the nervous system. We searched the scientific literature, dating from 1934 to August 2020, to compile data on the cause, epidemiology, clinical presentation, neuroimaging features, and treatment of each of the diseases of epidemic or pandemic potential as viewed through a neurologist's lens. We included articles with an abstract or full text in English in this topical and scoping review. Diseases with epidemic and pandemic potential can be spread directly from human to human, animal to human, via mosquitoes or other insects, or via environmental contamination. Manifestations include central neurologic conditions (meningitis, encephalitis, intraparenchymal hemorrhage, seizures), peripheral and cranial nerve syndromes (sensory neuropathy, sensorineural hearing loss, ophthalmoplegia), post-infectious syndromes (acute inflammatory polyneuropathy), and congenital syndromes (fetal microcephaly), among others. Some diseases have not been well-characterized from a neurological standpoint, but all have at least scattered case reports of neurological features. Some of the diseases have curative treatments available while in other cases, supportive care remains the only management option. Regardless of the pathogen, prompt, and aggressive measures to control the spread of these agents are the most important factors in lowering the overall morbidity and mortality they can cause.
Collapse
Affiliation(s)
- Caleb R. S. McEntire
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - Kun-Wei Song
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - Robert P. McInnis
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - John Y. Rhee
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - Michael Young
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - Erika Williams
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - Leah L. Wibecan
- Massachusetts General Hospital (MGH)-Brigham Pediatric Neurology Residency Program, Boston, MA, United States
| | - Neal Nolan
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - Amanda M. Nagy
- Massachusetts General Hospital (MGH)-Brigham Pediatric Neurology Residency Program, Boston, MA, United States
| | - Jeffrey Gluckstein
- Massachusetts General Hospital (MGH)-Brigham Neurology Residency Program, Boston, MA, United States
| | - Shibani S. Mukerji
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Farrah J. Mateen
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
30
|
Bailey AL, Kang LI, de Assis Barros D'Elia Zanella LGF, Silveira CGT, Ho YL, Foquet L, Bial G, McCune BT, Duarte-Neto AN, Thomas A, Raué HP, Byrnes K, Kallas EG, Slifka MK, Diamond MS. Consumptive coagulopathy of severe yellow fever occurs independently of hepatocellular tropism and massive hepatic injury. Proc Natl Acad Sci U S A 2020; 117:32648-32656. [PMID: 33268494 PMCID: PMC7768776 DOI: 10.1073/pnas.2014096117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Yellow fever (YF) is a mosquito-transmitted viral disease that causes tens of thousands of deaths each year despite the long-standing deployment of an effective vaccine. In its most severe form, YF manifests as a hemorrhagic fever that causes severe damage to visceral organs. Although coagulopathy is a defining feature of severe YF in humans, the mechanism by which it develops remains uncertain. Hepatocytes are a major target of yellow fever virus (YFV) infection, and the coagulopathy in severe YF has long been attributed to massive hepatocyte infection and destruction that results in a defect in clotting factor synthesis. However, when we analyzed blood from Brazilian patients with severe YF, we found high concentrations of plasma D-dimer, a fibrin split product, suggestive of a concurrent consumptive process. To define the relationship between coagulopathy and hepatocellular tropism, we compared infection and disease in Fah-/-, Rag2-/-, and Il2rɣ-/- mice engrafted with human hepatocytes (hFRG mice) and rhesus macaques using a highly pathogenic African YFV strain. YFV infection of macaques and hFRG mice caused substantial hepatocyte infection, liver damage, and coagulopathy as defined by virological, clinical, and pathological criteria. However, only macaques developed a consumptive coagulopathy whereas YFV-infected hFRG mice did not. Thus, infection of cell types other than hepatocytes likely contributes to the consumptive coagulopathy associated with severe YF in primates and humans. These findings expand our understanding of viral hemorrhagic disease and associated coagulopathy and suggest directions for clinical management of severe YF cases.
Collapse
Affiliation(s)
- Adam L Bailey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| | - Liang-I Kang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | | | - Cássia G T Silveira
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, Sao Paulo, Brazil 01246 903
| | - Yeh-Li Ho
- Department of Infectious and Parasitic Diseases, School of Medicine, University of São Paulo, Sao Paulo, Brazil 01246 903
| | | | - Greg Bial
- Yecuris Corporation, Tualatin, OR 97062
| | - Broc T McCune
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Amaro Nunes Duarte-Neto
- Department of Pathology, Clinical Hospital, School of Medicine, University of São Paulo, Sao Paulo, Brazil 01246 903
| | - Archana Thomas
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Kathleen Byrnes
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Esper G Kallas
- Department of Infectious and Parasitic Diseases, School of Medicine, University of São Paulo, Sao Paulo, Brazil 01246 903
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
31
|
Puerta-Guardo H, Glasner DR, Espinosa DA, Biering SB, Patana M, Ratnasiri K, Wang C, Beatty PR, Harris E. Flavivirus NS1 Triggers Tissue-Specific Vascular Endothelial Dysfunction Reflecting Disease Tropism. Cell Rep 2020; 26:1598-1613.e8. [PMID: 30726741 PMCID: PMC6934102 DOI: 10.1016/j.celrep.2019.01.036] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/27/2018] [Accepted: 01/09/2019] [Indexed: 01/22/2023] Open
Abstract
Flaviviruses cause systemic or neurotropic-encephalitic pathology in humans. The flavivirus nonstructural protein 1 (NS1) is a secreted glycoprotein involved in viral replication, immune evasion, and vascular leakage during dengue virus infection. However, the contribution of secreted NS1 from related flaviviruses to viral pathogenesis remains unknown. Here, we demonstrate that NS1 from dengue, Zika, West Nile, Japanese encephalitis, and yellow fever viruses selectively binds to and alters permeability of human endothelial cells from lung, dermis, umbilical vein, brain, and liver in vitro and causes tissue-specific vascular leakage in mice, reflecting the pathophysiology of each flavivirus. Mechanistically, each flavivirus NS1 leads to differential disruption of endothelial glycocalyx components, resulting in endothelial hyperpermeability. Our findings reveal the capacity of a secreted viral protein to modulate endothelial barrier function in a tissue-specific manner both in vitro and in vivo, potentially influencing virus dissemination and pathogenesis and providing targets for antiviral therapies and vaccine development. Puerta-Guardo et al. discover that five flavivirus NS1 proteins trigger hyperpermeability and vascular dysfunction in human endothelial cells and mice in a manner reflecting disease tropism. This tissue-specific tropism is partially determined by the capacity of NS1 to bind endothelial cells and is characterized by disruption of endothelial glycocalyx components.
Collapse
Affiliation(s)
- Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Dustin R Glasner
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Diego A Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, 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
| | - Mark Patana
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Kalani Ratnasiri
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Chunling Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - P Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
32
|
da Silva FC, Magaldi FM, Sato HK, Bevilacqua E. Yellow Fever Vaccination in a Mouse Model Is Associated With Uninterrupted Pregnancies and Viable Neonates Except When Administered at Implantation Period. Front Microbiol 2020; 11:245. [PMID: 32153534 PMCID: PMC7044120 DOI: 10.3389/fmicb.2020.00245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 02/03/2020] [Indexed: 02/05/2023] Open
Abstract
The potential risk of yellow fever (YF) infection in unvaccinated pregnant women has aroused serious concerns. In this study, we evaluated the effect of the YF vaccine during gestation using a mouse model, analyzing placental structure, immunolocalization of the virus antigen, and viral activity at the maternal-fetal barrier and in the maternal liver and fetus. The YF vaccine (17DD) was administered subcutaneously at a dose of 2.0 log10 PFU to CD-1 mice on gestational days (gd) 0.5, 5.5, and 11.5 (n = 5–10/group). The control group received sterile saline (n = 5–10/group). Maternal liver, implantation sites with fetus, and placentas were collected on gd18.5. The numbers of implantation sites, reabsorbed embryos, and stillborn fetuses were counted, and placentas and live fetuses were weighed. Tissues (placenta, fetuses, and liver) of vaccinated pregnant mice on gd5.5 (n = 15) were paraffin-embedded in 10% buffered-formalin and collected in TRIzol for immunolocalization of YF vaccine virus and PCR, respectively. PCR products were also subjected to automated sequence analysis. Fetal growth restriction (p < 0.0001) and a significant decrease in fetal viability (p < 0.0001) occurred only when the vaccine was administered on gd5.5. In stillbirths, the viral antigen was consistently immunolocalized at the maternal-fetal barrier and in fetal organs, suggesting a transplacental transfer. In stillbirths, RNA of the vaccine virus was also detected by reverse transcriptase-PCR indicating viral activity in the maternal liver and fetal tissues. In conclusion, the findings of this study in the mouse suggest that vaccination did not cause adverse outcomes with respect to fetal development except when administered during the early gestational stage, indicating the implantation period as a susceptible period in which the YF vaccine virus might interfere with pregnancy.
Collapse
Affiliation(s)
- Fernanda C da Silva
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fernanda M Magaldi
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Helena K Sato
- Secretaria do Estado de São Paulo, Epidemiological Surveillance Center, Department of Health, São Paulo, Brazil
| | - Estela Bevilacqua
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
33
|
Afrough B, Dowall S, Hewson R. Emerging viruses and current strategies for vaccine intervention. Clin Exp Immunol 2020; 196:157-166. [PMID: 30993690 PMCID: PMC6468171 DOI: 10.1111/cei.13295] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
During the past decade several notable viruses have suddenly emerged from obscurity or anonymity to become serious global health threats, provoking concern regarding their sustained epidemic transmission in immunologically naive human populations. With each new threat comes the call for rapid vaccine development. Indeed, vaccines are considered a critical component of disease prevention for emerging viral infections because, in many cases, other medical options are limited or non‐existent, or that infections result in such a rapid clinical deterioration that the effectiveness of therapeutics is limited. While classic approaches to vaccine development are still amenable to emerging viruses, the application of molecular techniques in virology has profoundly influenced our understanding of virus biology, and vaccination methods based on replicating, attenuated and non‐replicating virus vector approaches have become useful vaccine platforms. Together with a growing understanding of viral disease emergence, a range of vaccine strategies and international commitment to underpin development, vaccine intervention for new and emerging viruses may become a possibility.
Collapse
Affiliation(s)
- B Afrough
- Virology and Pathogenesis Laboratory, National Infection Service, Public Health England, Salisbury, UK
| | - S Dowall
- Virology and Pathogenesis Laboratory, National Infection Service, Public Health England, Salisbury, UK
| | - R Hewson
- Virology and Pathogenesis Laboratory, National Infection Service, Public Health England, Salisbury, UK
| |
Collapse
|
34
|
Domingo C, Fraissinet J, Ansah PO, Kelly C, Bhat N, Sow SO, Mejía JE. Long-term immunity against yellow fever in children vaccinated during infancy: a longitudinal cohort study. THE LANCET. INFECTIOUS DISEASES 2019; 19:1363-1370. [PMID: 31543249 PMCID: PMC6892259 DOI: 10.1016/s1473-3099(19)30323-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/24/2019] [Accepted: 06/07/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND A single dose of vaccine against yellow fever is routinely administered to infants aged 9-12 months under the Expanded Programme on Immunization, but the long-term outcome of vaccination in this age group is unknown. We aimed to evaluate the long-term persistence of neutralising antibodies to yellow fever virus following routine vaccination in infancy. METHODS We did a longitudinal cohort study, using a microneutralisation assay to measure protective antibodies against yellow fever in Malian and Ghanaian children vaccinated around age 9 months and followed up for 4·5 years (Mali), or 2·3 and 6·0 years (Ghana). Healthy children with available day-0 sera, a complete follow-up history, and no record of yellow fever revaccination were included; children seropositive for yellow fever at baseline were excluded. We standardised antibody concentrations with reference to the yellow fever WHO International Standard. FINDINGS We included 587 Malian and 436 Ghanaian children vaccinated between June 5, 2009, and Dec 26, 2012. In the Malian group, 296 (50·4%, 95% CI 46·4-54·5) were seropositive (antibody concentration ≥0·5 IU/mL) 4·5 years after vaccination. Among the Ghanaian children, 121 (27·8%, 23·5-32·0) were seropositive after 2·3 years. These results show a large decrease from the proportions of seropositive infants 28 days after vaccination, 96·7% in Mali and 72·7% in Ghana, reported by a previous study of both study populations. The number of seropositive children increased to 188 (43·1%, 95% CI 38·5-47·8) in the Ghanaian group 6·0 years after vaccination, but this result might be confounded by unrecorded revaccination or natural infection with wild yellow fever virus during a 2011-12 outbreak in northern Ghana. INTERPRETATION Rapid waning of immunity during the early years after vaccination of 9-month-old infants argues for a revision of the single-dose recommendation for this target population in endemic countries. The short duration of immunity in many vaccinees suggests that booster vaccination is necessary to meet the 80% population immunity threshold for prevention of yellow fever outbreaks. FUNDING Wellcome Trust.
Collapse
Affiliation(s)
- Cristina Domingo
- Robert Koch Institute, Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, WHO Collaborating Centre for Emerging Infections and Biological Threats, Berlin, Germany.
| | - Juliane Fraissinet
- Robert Koch Institute, Highly Pathogenic Viruses (ZBS 1), Centre for Biological Threats and Special Pathogens, WHO Collaborating Centre for Emerging Infections and Biological Threats, Berlin, Germany
| | - Patrick O Ansah
- Navrongo Health Research Centre and Research Laboratory, Navrongo, Ghana
| | | | | | - Samba O Sow
- National Institute of Research on Public Health, Bamako, Mali
| | - José E Mejía
- Centre de Physiopathologie Toulouse-Purpan (CNRS, INSERM, Université Paul Sabatier), Centre Hospitalier Universitaire Purpan, Toulouse, France
| |
Collapse
|
35
|
Kimathi D, Juan A, Bejon P, Grais RF, Warimwe GM. Randomized, double-blinded, controlled non-inferiority trials evaluating the immunogenicity and safety of fractional doses of Yellow Fever vaccines in Kenya and Uganda. Wellcome Open Res 2019; 4:182. [PMID: 31984244 PMCID: PMC6971842 DOI: 10.12688/wellcomeopenres.15579.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2019] [Indexed: 01/22/2023] Open
Abstract
Introduction: Yellow fever is endemic in specific regions of sub-Saharan Africa and the Americas, with recent epidemics occurring on both continents. The yellow fever vaccine is effective, affordable and safe, providing life-long immunity following a single dose vaccination. However, the vaccine production process is slow and cannot be readily scaled up during epidemics. This has led the World Health Organization (WHO) to recommend the use of fractional doses as a dose-sparing strategy during epidemics, but there are no randomized controlled trials of fractional yellow fever vaccine doses in Africa. Methods and analysis: We will recruit healthy adult volunteers, adults living with HIV, and children to a series of randomized controlled trials aiming to determine the immunogenicity and safety of fractional vaccine doses in comparison to the standard vaccine dose. The trials will be conducted across two sites; Kilifi, Kenya and Mbarara, Uganda. Recruited participants will be randomized to receive fractional or standard doses of yellow fever vaccine. Scheduled visits will include blood collection for serum and peripheral blood mononuclear cells (PBMCs) before vaccination and on various days - up to 2 years - post-vaccination. The primary outcome is the rate of seroconversion as measured by the plaque reduction neutralization test (PRNT 50) at 28 days post-vaccination. Secondary outcomes include antibody titre changes, longevity of the immune response, safety assessment using clinical data, the nature and magnitude of the cellular immune response and post-vaccination control of viremia by vaccine dose. Ethics and dissemination: The clinical trial protocols have received approval from the relevant institutional ethics and regulatory review committees in Kenya and Uganda, and the WHO Ethics Review Committee. The research findings will be disseminated through open-access publications and presented at relevant conferences and workshops. Registration: ClinicalTrials.gov NCT02991495 (registered on 13 December 2016) and NCT04059471 (registered on 15 August 2019).
Collapse
Affiliation(s)
- Derick Kimathi
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - George M Warimwe
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Davis EH, Barrett ADT. Structure-Function of the Yellow Fever Virus Envelope Protein: Analysis of Antibody Epitopes. Viral Immunol 2019; 33:12-21. [PMID: 31682201 DOI: 10.1089/vim.2019.0107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Yellow fever virus (YFV) is the prototype member of the genus Flavivirus, which contains more than 60 positive-sense, single-stranded RNA viruses, many of which are considered public health threats. YF disease is controlled by a live attenuated vaccine, 17D, which was generated empirically through serial passage of the wild-type (WT) strain Asibi in chicken tissue. The vaccine, which has been used for over 80 years, is considered to be one of the safest and most effective live attenuated vaccines. It has been shown that the humoral immune response is essential to a positive disease outcome during infection. As such, the neutralizing antibody response and its correlation to long-term protection are a critical measure of 17D efficacy. The primary target of these antibodies is the envelope (E) protein, which is the major component of the virion. Monoclonal antibodies can distinguish WT strain Asibi and vaccine strain 17D by many different measures, including physical binding, hemagglutination inhibition, neutralization, and passive protection. This makes the WT-vaccine pair ideal candidates to study the structure-function relationship of the E protein in the attenuation and immunogenicity of flaviviruses. In this study, we provide an overview of structure-function of YFV E protein and its involvement in protective immunity.
Collapse
Affiliation(s)
- Emily H Davis
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas.,Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas.,Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
37
|
Yellow Fever: Integrating Current Knowledge with Technological Innovations to Identify Strategies for Controlling a Re-Emerging Virus. Viruses 2019; 11:v11100960. [PMID: 31627415 PMCID: PMC6832525 DOI: 10.3390/v11100960] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/30/2019] [Accepted: 10/11/2019] [Indexed: 01/17/2023] Open
Abstract
Yellow fever virus (YFV) represents a re-emerging zoonotic pathogen, transmitted by mosquito vectors to humans from primate reservoirs. Sporadic outbreaks of YFV occur in endemic tropical regions, causing a viral hemorrhagic fever (VHF) associated with high mortality rates. Despite a highly effective vaccine, no antiviral treatments currently exist. Therefore, YFV represents a neglected tropical disease and is chronically understudied, with many aspects of YFV biology incompletely defined including host range, host–virus interactions and correlates of host immunity and pathogenicity. In this article, we review the current state of YFV research, focusing on the viral lifecycle, host responses to infection, species tropism and the success and associated limitations of the YFV-17D vaccine. In addition, we highlight the current lack of available treatments and use publicly available sequence and structural data to assess global patterns of YFV sequence diversity and identify potential drug targets. Finally, we discuss how technological advances, including real-time epidemiological monitoring of outbreaks using next-generation sequencing and CRISPR/Cas9 modification of vector species, could be utilized in future battles against this re-emerging pathogen which continues to cause devastating disease.
Collapse
|
38
|
Duarte‐Neto AN, Cunha MDP, Marcilio I, Song ATW, Martino RB, Ho Y, Pour SZ, Dolhnikoff M, Saldiva PHN, Duarte MIS, Takakura CF, Lima FR, Tanigawa RY, Iglezias SD, Kanamura CT, Santos ABG, Perondi B, Zanotto PMDA, D’Albuquerque LAC, Alves VAF. Yellow fever and orthotopic liver transplantation: new insights from the autopsy room for an old but re‐emerging disease. Histopathology 2019; 75:638-648. [DOI: 10.1111/his.13904] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/12/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Amaro N Duarte‐Neto
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Marielton dos P Cunha
- Laboratório de Evolução Molecular e Bioinformática (LEMB) Instituto de Ciências Biomédicas (ICB)Universidade de São Paulo São PauloBrazil
| | - Izabel Marcilio
- Núcleo de Vigilância Epidemiológica, Hospital das Clínicas, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Alice T W Song
- Divisão de Transplante de Fígado e Órgãos do Aparelho Digestivo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina Universidade de São Paulo São PauloBrazil
| | - Rodrigo B Martino
- Divisão de Transplante de Fígado e Órgãos do Aparelho Digestivo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina Universidade de São Paulo São PauloBrazil
| | - Yeh‐Li Ho
- Unidade de Terapia Intensiva da Divisão de Clínica de Moléstias Infecciosas e Parasitárias Hospital das Clinicas, Faculdade de Medicina São PauloBrazil
| | - Shahab Z Pour
- Laboratório de Evolução Molecular e Bioinformática (LEMB) Instituto de Ciências Biomédicas (ICB)Universidade de São Paulo São PauloBrazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Paulo H N Saldiva
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Maria I S Duarte
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Cleusa F Takakura
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Fabiana R Lima
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Ryan Y Tanigawa
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | | | | | - Angela B G Santos
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| | - Beatriz Perondi
- Comitê de Crise da Febre Amarela, Diretoria Clínica, Hospital das Clinicas HCFMUSP, Faculdade de Medicina Universidade de São Paulo São Paulo Brazil
| | - Paolo M de A Zanotto
- Laboratório de Evolução Molecular e Bioinformática (LEMB) Instituto de Ciências Biomédicas (ICB)Universidade de São Paulo São PauloBrazil
| | - Luiz A C D’Albuquerque
- Divisão de Transplante de Fígado e Órgãos do Aparelho Digestivo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina Universidade de São Paulo São PauloBrazil
| | - Venancio A F Alves
- Departamento de Patologia, Faculdade de MedicinaUniversidade de São Paulo São PauloBrazil
| |
Collapse
|
39
|
Jácome R, Carrasco-Hernández R, Campillo-Balderas JA, López-Vidal Y, Lazcano A, Wenzel RP, Ponce de León S. A yellow flag on the horizon: The looming threat of yellow fever to North America. Int J Infect Dis 2019; 87:143-150. [PMID: 31382047 DOI: 10.1016/j.ijid.2019.07.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES Yellow fever virus historically was a frequent threat to American and European coasts. Medical milestones such as the discovery of mosquitoes as vectors and subsequently an effective vaccine significantly reduced its incidence, in spite of which, thousands of cases of this deathly disease still occur regularly in Sub-Saharan Africa and the Amazonian basin in South America, which are usually not reported. An urban outbreak in Angola, consecutive years of increasing incidence near major Brazilian cities, and imported cases in China, South America and Europe, have brought this virus back to the global spotlight. The aim of this article is to underline that the preventive YFV measures, such as vaccination, need to be carefully revised in order to minimize the risks of new YFV outbreaks, especially in urban or immunologically vulnerable places. Furthermore, this article highlights the diverse factors that have favored the spread of other Aedes spp.-associated arboviral diseases like Dengue, Chikungunya and Zika, to northern latitudes causing epidemics in the United States and Europe, emphasizing the possibility that YFV might follow the path of these viruses unless enhanced surveillance and efficient control systems are urgently initiated.
Collapse
Affiliation(s)
- Rodrigo Jácome
- Laboratorio de Origen de la Vida, Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.P. 04510, Mexico City, Mexico
| | - R Carrasco-Hernández
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.P. 04510, Mexico City, Mexico
| | - José Alberto Campillo-Balderas
- Laboratorio de Origen de la Vida, Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.P. 04510, Mexico City, Mexico
| | - Yolanda López-Vidal
- Programa de Inmunología Molecular Microbiana, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.P. 04510, Mexico City, Mexico
| | - Antonio Lazcano
- Laboratorio de Origen de la Vida, Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.P. 04510, Mexico City, Mexico; Miembro de El Colegio Nacional, Mexico
| | | | - Samuel Ponce de León
- Programa Universitario de Investigación en Salud, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.P. 04510, Mexico City, Mexico.
| |
Collapse
|
40
|
Ho YL, Joelsons D, Leite GFC, Malbouisson LMS, Song ATW, Perondi B, Andrade LC, Pinto LF, D'Albuquerque LAC, Segurado AAC. Severe yellow fever in Brazil: clinical characteristics and management. J Travel Med 2019; 26:5509466. [PMID: 31150098 DOI: 10.1093/jtm/taz040] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/25/2019] [Accepted: 05/27/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Little is known about clinical characteristics and management of severe yellow fever as previous yellow fever epidemics often occurred in times or areas with little access to intensive care units (ICU). We aim to describe the clinical characteristics of severe yellow fever cases requiring admission to the ICU during the 2018 yellow fever outbreak in São Paulo, Brazil. Furthermore, we report on preliminary lessons learnt regarding clinical management of severe yellow fever. METHODS Retrospective descriptive cohort study. Demographic data, laboratory test results on admission, clinical follow-up, and clinical outcomes were evaluated. RESULTS From 10 January to 11 March 2018, 79 patients with laboratory confirmed yellow fever were admitted to the ICU in a tertiary hospital in Sao Paolo because of rapid clinical deterioration. On admission, the median AST was 7,000 IU/L, ALT 3,936 IU/L, total bilirubin 5.3 ml/dL, platelet 74 × 103/mm3, INR 2.24 and factor V 37%. Seizures occurred in 24% of patients, even without substantial intracranial hypertension. The high frequency of pancreatitis and rapidly progressive severe metabolic acidosis were notable findings. 73% of patients required renal replacement therapy. The in-hospital fatality rate was 67%. Patients with diabetes mellitus had a higher case fatality rate (CFR) of 80%, while patients without diabetes had a CFR of 65%. Leading causes of death were severe gastrointestinal bleeding, epileptic status, severe metabolic acidosis, necrohemorrhagic pancreatitis, and multi-organ failure. CONCLUSIONS Severe yellow fever is associated with a high CFR. The following management lessons were learnt: Anticonvulsant drugs in patients with any symptoms of hepatic encephalopathy or arterial ammonia levels >70 μmol/L was commenced which reduced the frequency of seizures from 28% to 17%. Other new therapy strategies included early institution of plasma exchange. Due to the high frequency of gastric bleeding, therapeutic doses of intravenous proton pump inhibitors should be administered.
Collapse
Affiliation(s)
- Yeh-Li Ho
- Departamento e Divisão de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas, Faculdade de Medicina USP (HCFMUSP), Sao Paulo, Brazil
| | - Daniel Joelsons
- Departamento e Divisão de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas, Faculdade de Medicina USP (HCFMUSP), Sao Paulo, Brazil
| | - Gabriel F C Leite
- Departamento e Divisão de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas, Faculdade de Medicina USP (HCFMUSP), Sao Paulo, Brazil
| | | | - Alice T W Song
- Disciplina de Transplantes de Órgãos do Aparelho Digestivo do Departamento de Gastroenterologia, HCFMUSP, Sao Paulo, Brazil
| | | | | | - Lécio F Pinto
- Divisão de Clínica Neurológica, HCFMUSP, Sao Paulo, Brazil
| | - Luiz A C D'Albuquerque
- Disciplina de Transplantes de Órgãos do Aparelho Digestivo do Departamento de Gastroenterologia, HCFMUSP, Sao Paulo, Brazil
| | - Aluisio A C Segurado
- Departamento e Divisão de Moléstias Infecciosas e Parasitárias, Hospital das Clínicas, Faculdade de Medicina USP (HCFMUSP), Sao Paulo, Brazil
| | | |
Collapse
|
41
|
Casadio LVB, Salles APM, Malta FDM, Leite GF, Ho YL, Gomes-Gouvêa MS, Malbouisson LMS, Levin AS, de Azevedo RS, Carrilho FJ, Nastri ACSS, Pinho JRR. Lipase and factor V (but not viral load) are prognostic factors for the evolution of severe yellow fever cases. Mem Inst Oswaldo Cruz 2019; 114:e190033. [PMID: 31116245 PMCID: PMC6528381 DOI: 10.1590/0074-02760190033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/24/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Despite a highly efficacious vaccine, yellow fever (YF) is still a major threat in developing countries and a cause of outbreaks. In 2018, the Brazilian state of São Paulo witnessed a new YF outbreak in areas where the virus has not been detected before. OBJECTIVE The aim is to describe the clinical and laboratorial characteristics of severe cases of YF, evaluate viral to determine markers associated with fatal outcome. METHODS Acute severe YF cases (n = 62) were admitted to the Intensive Care Unit of a reference hospital and submitted to routine laboratorial evaluation on admission. YFV-RNA was detected in serum and urine by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and then sequenced. Patients were classified in two groups: survival or death. FINDINGS In the univariate analysis the following variables were associated with outcome: alanin aminotransferase (ALT), aspartat aminotransferase (AST), AST/ALT ratio, total bilirubin (TB), chronic kidney disease epidemiology collaboration (CKD-EPI), ammonia, lipase, factor V, international normalised ratio (INR), lactate and bicarbonate. Logistic regression model showed two independent variables associated with death: lipase [odds ratio (OR) 1.018, 95% confidence interval (CI) 1.007 to 1.030, p = 0.002], and factor V (OR -0.955, 95% CI 0.929 to 0.982, p = 0.001). The estimated lipase and factor V cut-off values that maximised sensitivity and specificity for death prediction were 147.5 U/L [area under the curve (AUC) = 0.879], and 56.5% (AUC = 0.913). MAIN CONCLUSIONS YF acute severe cases show a generalised involvement of different organs (liver, spleen, heart, kidneys, intestines and pancreas), and different parameters were related to outcome. Factor V and lipase are independent variables associated with death, reinforcing the importance of hemorrhagic events due to fulminant liver failure and pointing to pancreatitis as a relevant event in the outcome of the disease.
Collapse
Affiliation(s)
- Luciana Vilas Boas Casadio
- Universidade de São Paulo, Faculdade de Medicina da São Paulo, Instituto de Medicina Tropical, Departamento de Gastroenterologia, Laboratório de Gastroenterologia e Hepatologia Tropical - LIM/07, São Paulo, SP, Brasil
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Moléstias Infecciosas e Parasitárias, São Paulo, SP, Brasil
| | - Ana Paula Moreira Salles
- Universidade de São Paulo, Faculdade de Medicina da São Paulo, Instituto de Medicina Tropical, Departamento de Gastroenterologia, Laboratório de Gastroenterologia e Hepatologia Tropical - LIM/07, São Paulo, SP, Brasil
| | - Fernanda de Mello Malta
- Universidade de São Paulo, Faculdade de Medicina da São Paulo, Instituto de Medicina Tropical, Departamento de Gastroenterologia, Laboratório de Gastroenterologia e Hepatologia Tropical - LIM/07, São Paulo, SP, Brasil
| | - Gabriel Fialkovitz Leite
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Moléstias Infecciosas e Parasitárias, São Paulo, SP, Brasil
| | - Yeh-Li Ho
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Moléstias Infecciosas e Parasitárias, São Paulo, SP, Brasil
| | - Michele Soares Gomes-Gouvêa
- Universidade de São Paulo, Faculdade de Medicina da São Paulo, Instituto de Medicina Tropical, Departamento de Gastroenterologia, Laboratório de Gastroenterologia e Hepatologia Tropical - LIM/07, São Paulo, SP, Brasil
| | - Luiz Marcelo Sá Malbouisson
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Anna S Levin
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Moléstias Infecciosas e Parasitárias, São Paulo, SP, Brasil
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | | | - Flair José Carrilho
- Universidade de São Paulo, Faculdade de Medicina da São Paulo, Instituto de Medicina Tropical, Departamento de Gastroenterologia, Laboratório de Gastroenterologia e Hepatologia Tropical - LIM/07, São Paulo, SP, Brasil
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Ana Catharina Seixas Santos Nastri
- Universidade de São Paulo, Faculdade de Medicina da São Paulo, Instituto de Medicina Tropical, Departamento de Gastroenterologia, Laboratório de Gastroenterologia e Hepatologia Tropical - LIM/07, São Paulo, SP, Brasil
- Faculdade de Medicina da Universidade de São Paulo, Hospital das Clínicas, Departamento de Moléstias Infecciosas e Parasitárias, São Paulo, SP, Brasil
| | - João Renato Rebello Pinho
- Universidade de São Paulo, Faculdade de Medicina da São Paulo, Instituto de Medicina Tropical, Departamento de Gastroenterologia, Laboratório de Gastroenterologia e Hepatologia Tropical - LIM/07, São Paulo, SP, Brasil
- Hospital Israelita Albert Einstein, Albert Einstein Medicina Diagnóstica, Laboratório de Técnicas Especiais, São Paulo, SP, Brasil
| |
Collapse
|
42
|
Kallas EG, D'Elia Zanella LGFAB, Moreira CHV, Buccheri R, Diniz GBF, Castiñeiras ACP, Costa PR, Dias JZC, Marmorato MP, Song ATW, Maestri A, Borges IC, Joelsons D, Cerqueira NB, Santiago E Souza NC, Morales Claro I, Sabino EC, Levi JE, Avelino-Silva VI, Ho YL. Predictors of mortality in patients with yellow fever: an observational cohort study. THE LANCET. INFECTIOUS DISEASES 2019; 19:750-758. [PMID: 31104909 DOI: 10.1016/s1473-3099(19)30125-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Yellow fever virus infection results in death in around 30% of symptomatic individuals. The aim of this study was to identify predictors of death measured at hospital admission in a cohort of patients admitted to hospital during the 2018 outbreak of yellow fever in the outskirts of São Paulo city, Brazil. METHODS In this observational cohort study, we enrolled patients with yellow fever virus from two hospitals in São Paolo-the Hospital das Clínicas, University of São Paulo and the Infectious Diseases Institute "Emilio Ribas". Patients older than 18 years admitted to hospital with fever or myalgia, headache, arthralgia, oedema, rash, or conjunctivitis were consecutively screened for inclusion in the present study. Consenting patients were included if they had travelled to geographical areas in which yellow fever virus cases had been previously confirmed. Yellow fever infection was confirmed by real-time PCR in blood collected at admission or tissues at autopsy. We sequenced the complete genomes of yellow fever virus from infected individuals and evaluated demographic, clinical, and laboratory findings at admission and investigated whether any of these measurements correlated with patient outcome (death). FINDINGS Between Jan 11, 2018, and May 10, 2018, 118 patients with suspected yellow fever were admitted to Hospital das Clínicas, and 113 patients with suspected yellow fever were admitted to Infectious Diseases Institute "Emilio Ribas". 95 patients with suspected yellow fever were included in the study, and 136 patients were excluded. Three (3%) of 95 patients with suspected yellow fever who were included in the study were excluded because they received a different diagnosis, and 16 patients with undetectable yellow fever virus RNA were excluded. Therefore, 76 patients with confirmed yellow fever virus infection, based on detectable yellow fever virus RNA in blood (74 patients) or yellow fever virus confirmed only at the autopsy report (two patients), were included in our analysis. 27 (36%) of 76 patients died during the 60 day period after hospital admission. We generated 14 complete yellow fever virus genomes from the first 15 viral load-detectable samples. The genomes belonged to a single monophyletic clade of the South America I genotype, sub-genotype E. Older age, male sex, higher leukocyte and neutrophil counts, higher alanine aminotransferase, aspartate transaminase (AST), bilirubin, and creatinine, prolonged prothrombin time, and higher yellow fever virus RNA plasma viral load were associated with higher mortality. In a multivariate regression model, older age, elevated neutrophil count, increased AST, and higher viral load remained independently associated with death. All 11 (100%) patients with neutrophil counts of 4000 cells per mL or greater and viral loads of 5·1 log10 copies/mL or greater died (95% CI 72-100), compared with only three (11%) of 27 (95% CI 2-29) among patients with neutrophil counts of less than 4000 cells per mL and viral loads of less than 5·1 log10 copies/mL. INTERPRETATION We identified clinical and laboratory predictors of mortality at hospital admission that could aid in the care of patients with yellow fever virus. Identification of these prognostic markers in patients could help clinicians prioritise admission to the intensive care unit, as patients often deteriorate rapidly. Moreover, resource allocation could be improved to prioritise key laboratory examinations that might be more useful in determining whether a patient could have a better outcome. Our findings support the important role of the virus in disease pathogenesis, suggesting that an effective antiviral could alter the clinical course for patients with the most severe forms of yellow fever. FUNDING São Paulo Research Foundation (FAPESP).
Collapse
Affiliation(s)
- Esper G Kallas
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Luiz Gonzaga F A B D'Elia Zanella
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Infectious Diseases Institute "Emilio Ribas", São Paulo, Brazil
| | - Carlos Henrique V Moreira
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Infectious Diseases Institute "Emilio Ribas", São Paulo, Brazil
| | - Renata Buccheri
- Infectious Diseases Institute "Emilio Ribas", São Paulo, Brazil
| | | | | | - Priscilla R Costa
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Juliana Z C Dias
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Mariana P Marmorato
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alice T W Song
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alvino Maestri
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Igor C Borges
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Daniel Joelsons
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Natalia B Cerqueira
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ingra Morales Claro
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ester C Sabino
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - José Eduardo Levi
- Tropical Medicine Institute, University of São Paulo, São Paulo, Brazil; DASA Laboratories, São Paulo, Brazil
| | - Vivian I Avelino-Silva
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Yeh-Li Ho
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
43
|
Mustafá YM, Meuren LM, Coelho SVA, de Arruda LB. Pathways Exploited by Flaviviruses to Counteract the Blood-Brain Barrier and Invade the Central Nervous System. Front Microbiol 2019; 10:525. [PMID: 30984122 PMCID: PMC6447710 DOI: 10.3389/fmicb.2019.00525] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
Human infection by different flaviviruses may cause severe neurologic syndromes, through pathogenic mechanisms that are still largely unknown. Japanese encephalitis virus (JEV), West Nile virus (WNV), Zika virus (ZIKV), yellow fever virus (YFV), dengue virus (DENV), and tick-borne encephalitis virus (TBEV) are believed to reach the central nervous system by a hematogenous route, upon crossing the blood-brain barrier. Although the disruption of BBB during flavivirus infection has been largely evidenced in experimental models, the relevance of BBB breakdown for virus entering the brain was not completely elucidated. In vitro models of BBB had demonstrated that these viruses replicated in brain microvascular endothelial cells (BMECs), which induced downregulation of tight junction proteins and increased the permeability of the barrier. Other reports demonstrated that infection of BMECs allowed the basolateral release of infectious particles, without a remarkable cytopathic effect, what might be sufficient for virus invasion. Virus replication and activation of other cells associated to the BBB, mostly astrocytes and microglia, were also reported to affect the endothelial barrier permeability. This event might occur simultaneously or after BMECs infection, being a secondary effect leading to BBB disruption. Importantly, activation of BMECs, astrocytes, and microglia by flaviviruses was associated to the expression and secretion of inflammatory mediators, which are believed to recruit leukocytes to the CNS. The leukocyte infiltrate could further mediate viral invasion through a Trojan horse mechanism and might contribute to BBB breakdown and to neurological alterations. This review discussed the previous studies regarding in vitro and in vivo models of JEV, WNV, ZIKV, YFV, DENV, and TBEV infection and addressed the pathways for BBB overcome and invasion of the CNS described for each virus infection, aiming to increment the knowledge and stimulate further discussion about the role of BBB in the neuropathogenesis of flavivirus infection.
Collapse
Affiliation(s)
- Yasmin Mucunã Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lana Monteiro Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Sharton Vinícius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Bartol KD, Aguirre JL, Labruzzo SV, Henriet RP. Transverse myelitis associated with yellow fever vaccination. Proc (Bayl Univ Med Cent) 2019; 32:283-285. [PMID: 31191156 DOI: 10.1080/08998280.2019.1573405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 10/27/2022] Open
Abstract
Yellow fever is a mosquito-borne viral hemorrhagic fever endemic to sub-Saharan Africa and South America associated with significant morbidity and mortality. The use of a live attenuated vaccine can prevent yellow fever, but vaccine-associated neurologic disease has been reported and is a safety concern. We present the case of a previously healthy 35-year-old active-duty man who received the yellow fever vaccine prior to deployment and subsequently developed progressive neurologic dysfunction consistent with transverse myelitis.
Collapse
Affiliation(s)
- Kyle D Bartol
- Department of Medicine, William Beaumont Army Medical CenterEl PasoTexas
| | - Jose L Aguirre
- Department of Medicine, William Beaumont Army Medical CenterEl PasoTexas
| | | | - Russ P Henriet
- Department of Neurology, Madigan Army Medical CenterJoint Base Lewis-McChordWashington
| |
Collapse
|
45
|
Paixão GM, Nunes MCP, Beato BD, Sable C, Beaton AZ, Oliveira KK, Rezende BD, Rios JPP, Fraga CL, Pereira LS, Teixeira MRD, Oliveira NR, Pascoal-Xavier MA, Maciel GV, Brito CGX, Júnior MRL, Ribeiro ALP, Nascimento BR. Cardiac Involvement by Yellow Fever(from the PROVAR+ Study). Am J Cardiol 2019; 123:833-838. [PMID: 30545483 DOI: 10.1016/j.amjcard.2018.11.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 10/27/2022]
Abstract
Incidence of Yellow Fever (YF) has increased in Brazil, and cardiac findings such as bradyarrhythmias and conduction abnormalities have been described. We aimed to perform a comprehensive cardiac evaluation of patients with YF, and to assess the association between cardiac involvement and disease severity. Patients hospitalized with YF from February to March 2018 underwent clinical and laboratory evaluation, focused bedside echocardiography (GE Vivid IQ), electrocardiogram and, in case of alterations, 24-hours Holter. Patients were divided into 2 groups according to YF severity. Five patients underwent magnetic resonance imaging and 3 had necropsy. Seventy patients had confirmed YF, 69% with severe form. Mean age was 48 ± 14 years, 63 (90%) were males and 5 (7%) died. Significant electrocardiogram abnormalities were present in 52% of patients with mild/moderate form of YF (G1) and 77% of those with severe form (G2), p = 0.046. Sinus bradycardia was observed in 24% (N = 17): G1 23% versus G2 25%, p = 0.67. Among 32 patients who underwent Holter, 14 (44%) had mean HR <60 beats per minute, being 8 from G2. Echocardiogram revealed left ventricular dysfunction in 4 (6%) patients, from G2. Left ventricular wall thickening with a hyper-refringent myocardial texture suggesting infiltration was observed in 17 patients (G1 18% vs G2 27%, p = 0.55). One magnetic resonance (G2) was suggestive of myocarditis, and one necropsy revealed areas of myocardial necrosis and acute myocarditis. In conclusion, cardiac involvement was observed in patients with YF, most commonly bradycardia and myocardial hyper-refringent texture suggestive of infiltration.
Collapse
|
46
|
Petrova E, Gracias S, Beauclair G, Tangy F, Jouvenet N. Uncovering Flavivirus Host Dependency Factors through a Genome-Wide Gain-of-Function Screen. Viruses 2019; 11:v11010068. [PMID: 30650657 PMCID: PMC6356745 DOI: 10.3390/v11010068] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 01/14/2023] Open
Abstract
Flaviviruses, such as dengue (DENV), West Nile (WNV), yellow fever (YFV) and Zika (ZIKV) viruses, are mosquito-borne pathogens that present a major risk to global public health. To identify host factors that promote flavivirus replication, we performed a genome-wide gain-of-function cDNA screen for human genes that enhance the replication of flavivirus reporter particles in human cells. The screen recovered seventeen potential host proteins that promote viral replication, including the previously known dolichyl-diphosphooligosaccharide--protein glycosyltransferase non-catalytic subunit (DDOST). Using silencing approaches, we validated the role of four candidates in YFV and WNV replication: ribosomal protein L19 (RPL19), ribosomal protein S3 (RPS3), DDOST and importin 9 (IPO9). Applying a panel of virological, biochemical and microscopic methods, we validated further the role of RPL19 and DDOST as host factors required for optimal replication of YFV, WNV and ZIKV. The genome-wide gain-of-function screen is thus a valid approach to advance our understanding of flavivirus replication.
Collapse
Affiliation(s)
- Evgeniya Petrova
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology department, Institut Pasteur, 75015 Paris, France.
| | - Ségolène Gracias
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology department, Institut Pasteur, 75015 Paris, France.
| | - Guillaume Beauclair
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology department, Institut Pasteur, 75015 Paris, France.
| | - Frédéric Tangy
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology department, Institut Pasteur, 75015 Paris, France.
| | - Nolwenn Jouvenet
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Virology department, Institut Pasteur, 75015 Paris, France.
| |
Collapse
|
47
|
Laureti M, Narayanan D, Rodriguez-Andres J, Fazakerley JK, Kedzierski L. Flavivirus Receptors: Diversity, Identity, and Cell Entry. Front Immunol 2018; 9:2180. [PMID: 30319635 PMCID: PMC6168832 DOI: 10.3389/fimmu.2018.02180] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Flaviviruses are emerging and re-emerging arthropod-borne pathogens responsible for significant mortality and morbidity worldwide. The genus comprises more than seventy small, positive-sense, single-stranded RNA viruses, which are responsible for a spectrum of human and animal diseases ranging in symptoms from mild, influenza-like infection to fatal encephalitis and haemorrhagic fever. Despite genomic and structural similarities across the genus, infections by different flaviviruses result in disparate clinical presentations. This review focusses on two haemorrhagic flaviviruses, dengue virus and yellow fever virus, and two neurotropic flaviviruses, Japanese encephalitis virus and Zika virus. We review current knowledge on host-pathogen interactions, virus entry strategies and tropism.
Collapse
Affiliation(s)
- Mathilde Laureti
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Divya Narayanan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Julio Rodriguez-Andres
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - John K Fazakerley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
48
|
Phylogenomic analysis unravels evolution of yellow fever virus within hosts. PLoS Negl Trop Dis 2018; 12:e0006738. [PMID: 30188905 PMCID: PMC6143276 DOI: 10.1371/journal.pntd.0006738] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/18/2018] [Accepted: 08/07/2018] [Indexed: 11/30/2022] Open
Abstract
The yellow fever virus (YFV) recently reemerged in the large outbreaks in Africa and Brazil, and the first imported patients into Asia have recalled the concerns of YFV evolution. Here we show phylogenomics of YFV with serial clinical samples of the 2016 YFV infections. Phylogenetics exhibited that the 2016 strains were close to Angola 1971 strains and only three amino acid changes presented new to other lineages. Deep sequencing of viral genomes discovered 101 intrahost single nucleotide variations (iSNVs) and 234 single nucleotide polymorphisms (SNPs). Analysis of iSNV distribution and mutated allele frequency revealed that the coding regions were under purifying selection. Comparison of the evolutionary rates estimated by iSNV and SNP showed that the intrahost rate was ~2.25 times higher than the epidemic rate, and both rates were higher than the long-term YFV substitution rate, as expected. In addition, the result also hinted that short viremia duration of YFV might further hinder the evolution of YFV. The first importation of infections into China in 2016 and the following outbreaks in Africa and Brazil of yellow fever virus (YFV) have raised again the concerns of the potential viral spread into new territories. In this study, we aimed to know the evolution dynamics of YFV by using intrahost phylogenomics and to assess the risk of virus epidemics. Through deep sequencing of consecutive samples from 12 patients, we identified hundreds of genomic variations (iSNVs and SNPs), and noticed the nearly linear accumulation of variations within individuals. The estimated evolutionary rate within host is much higher than the epidemic evolutionary rate. In comparison with Dengue virus (DENV) and Zika virus (ZIKV), which share similar host vectors (Aedes spp.), life cycles, mutation rates and replication strategies to YFV, the lower epidemic evolutionary rate of YFV might have been hindered by the shorter viremia duration, which decreased the accumulated variations to get into the transmission cycle.
Collapse
|
49
|
Klitting R, Fischer C, Drexler JF, Gould EA, Roiz D, Paupy C, de Lamballerie X. What Does the Future Hold for Yellow Fever Virus? (II). Genes (Basel) 2018; 9:E425. [PMID: 30134625 PMCID: PMC6162518 DOI: 10.3390/genes9090425] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 02/06/2023] Open
Abstract
As revealed by the recent resurgence of yellow fever virus (YFV) activity in the tropical regions of Africa and South America, YFV control measures need urgent rethinking. Over the last decade, most reported outbreaks occurred in, or eventually reached, areas with low vaccination coverage but that are suitable for virus transmission, with an unprecedented risk of expansion to densely populated territories in Africa, South America and Asia. As reflected in the World Health Organization's initiative launched in 2017, it is high time to strengthen epidemiological surveillance to monitor accurately viral dissemination, and redefine vaccination recommendation areas. Vector-control and immunisation measures need to be adapted and vaccine manufacturing must be reconciled with an increasing demand. We will have to face more yellow fever (YF) cases in the upcoming years. Hence, improving disease management through the development of efficient treatments will prove most beneficial. Undoubtedly, these developments will require in-depth descriptions of YFV biology at molecular, physiological and ecological levels. This second section of a two-part review describes the current state of knowledge and gaps regarding the molecular biology of YFV, along with an overview of the tools that can be used to manage the disease at the individual, local and global levels.
Collapse
Affiliation(s)
- Raphaëlle Klitting
- Unité des Virus Émergents (UVE: Aix-Marseille Univ⁻IRD 190⁻Inserm 1207⁻IHU Méditerranée Infection), 13385 Marseille CEDEX 05, France.
| | - Carlo Fischer
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany.
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany.
| | - Jan F Drexler
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany.
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany.
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov University, 119991 Moscow, Russia.
| | - Ernest A Gould
- Unité des Virus Émergents (UVE: Aix-Marseille Univ⁻IRD 190⁻Inserm 1207⁻IHU Méditerranée Infection), 13385 Marseille CEDEX 05, France.
| | - David Roiz
- UMR Maladies Infectieuses et Vecteurs: Écologie, Génétique Évolution et Contrôle (MIVEGEC: IRD, CNRS, Univ. Montpellier), 34394 Montpellier, France.
| | - Christophe Paupy
- UMR Maladies Infectieuses et Vecteurs: Écologie, Génétique Évolution et Contrôle (MIVEGEC: IRD, CNRS, Univ. Montpellier), 34394 Montpellier, France.
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille Univ⁻IRD 190⁻Inserm 1207⁻IHU Méditerranée Infection), 13385 Marseille CEDEX 05, France.
| |
Collapse
|
50
|
Maciel GVR, Tavares MCDF, Pereira LS, Silva GLC, de Oliveira NR, Paulino E, Pascoal-Xavier MA. Disseminated mycosis in a patient with yellow fever. AUTOPSY AND CASE REPORTS 2018; 8:e2018038. [PMID: 30123781 PMCID: PMC6089461 DOI: 10.4322/acr.2018.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/19/2018] [Indexed: 12/27/2022] Open
Abstract
Disseminated mycosis (DM)—with cardiac involvement and shock—is an unexpected and severe opportunistic infection in patients with yellow fever. DM can mimic bacterial sepsis and should be considered in the differential diagnosis of causes of systemic inflammatory response syndrome in this group of patients, especially in areas where an outbreak of yellow fever is ongoing. We report the case of a 53-year-old male patient who presented to the emergency department with fever, myalgia, headache, and low back pain. The laboratory investigation revealed a positive molecular test for yellow fever, hepatic injury, and renal failure. During hospitalization, the patient developed hepatic encephalopathy, ascending leukocytosis, and ascites, with signs consistent with peritonitis. On the 11th day of hospitalization, the patient developed atrioventricular block, shock and died. At autopsy, angioinvasive mycosis was evidenced mainly in the heart, lungs, kidneys, and adrenals.
Collapse
Affiliation(s)
- Gustavo Vieira Rodrigues Maciel
- Federal University of Minas Gerais (UFMG), Brazilian Company of Hospital Services, Clinical Hospital, Pathologic Anatomy Laboratory. Belo Horizonte, MG, Brazil
| | - Marcelo Combat de Faria Tavares
- Federal University of Minas Gerais (UFMG), Brazilian Company of Hospital Services, Clinical Hospital, Pathologic Anatomy Laboratory. Belo Horizonte, MG, Brazil
| | - Leonardo Soares Pereira
- Hospital Foundation of Minas Gerais, Hospital Eduardo de Menezes. Belo Horizonte, MG, Brazil
| | | | - Neimy Ramos de Oliveira
- Hospital Foundation of Minas Gerais, Hospital Eduardo de Menezes. Belo Horizonte, MG, Brazil
| | - Eduardo Paulino
- Federal University of Minas Gerais (UFMG), Brazilian Company of Hospital Services, Clinical Hospital, Pathologic Anatomy Laboratory. Belo Horizonte, MG, Brazil.,Federal University of Minas Gerais (UFMG), Faculty of Medicine, Pathologic Anatomy Department. Belo Horizonte, MG, Brazil
| | - Marcelo Antonio Pascoal-Xavier
- Federal University of Minas Gerais (UFMG), Brazilian Company of Hospital Services, Clinical Hospital, Pathologic Anatomy Laboratory. Belo Horizonte, MG, Brazil.,Federal University of Minas Gerais (UFMG), Faculty of Medicine, Pathologic Anatomy Department. Belo Horizonte, MG, Brazil.,Oswaldo Cruz Foundation, René Rachou Institute, Research Group Immunology of Viral Diseases. Belo Horizonte, MG, Brazil
| |
Collapse
|