201
|
Pong LY, Yew PN, Lee WL, Lim YY, Sharifah SH. Anti-dengue virus serotype 2 activity of tannins from porcupine dates. Chin Med 2020; 15:49. [PMID: 32467721 PMCID: PMC7238553 DOI: 10.1186/s13020-020-00329-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/12/2020] [Indexed: 11/10/2022] Open
Abstract
Background Dengue fever is currently endemic in tropical and subtropical countries worldwide and effective drug against DENV infection is still unavailable. Porcupine dates, which are traditionally used to treat dengue fever, might contain potential anti-dengue compounds. Two porcupine dates, black date (BD) and powdery date (PD) from Himalayan porcupine (Hystrix brachyura), were investigated for their antiviral activities against DENV-2 in vitro. Methods The methanol crude extracts (MBD and MPD) were prepared from the raw material of porcupine dates. The tannin-rich fractions (BDTF and PDTF) were isolated from their methanol crude extracts using column chromatography. The presence of tannins in BDTF and PDTF extracts was determined by fourier-transform infrared spectroscopy (FTIR) and nuclear magnetic resonance (NMR) analyses. The cytotoxicity and anti-DENV-2 activities including virus yield inhibition, virucidal, virus attachment and pre-treatment assays of the extracts were examined in Vero cells. Results Our findings revealed that all the extracts of porcupine dates exhibited antiviral activity against DENV-2 in Vero cells. The IC50 of BDTF and PDTF were 25 µg/mL and 11 µg/mL respectively, while their methanol crude extracts demonstrated lower antiviral efficacy (IC50 ≈ 101–107 µg/mL). BDTF and PDTF also exerted a similar higher virucidal effect (IC50 of 11 µg/mL) than methanol crude extracts (IC50 ≈ 52–66 µg/mL). Furthermore, all the extracts inhibited the attachment of DENV-2 by at least 80%. Pre-treatments of cells with BDTF and PDTF markedly prevented DENV-2 infection when compared to methanol crude extracts. Conclusion This study suggests that porcupine dates possess antiviral properties against DENV-2, which is attributed to its tannin compounds.
Collapse
Affiliation(s)
- Lian Yih Pong
- 1Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia.,2Infectious Diseases and Health Cluster, Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| | - Peng Nian Yew
- 3School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia.,4Present Address: Department of Bioscience, Faculty of Applied Science, Tunku Abdul Rahman University College, Jalan Genting Kelang, 53300 Kuala Lumpur, Malaysia
| | - Wai Leng Lee
- 3School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| | - Yau Yan Lim
- 3School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| | - Syed Hassan Sharifah
- 1Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia.,2Infectious Diseases and Health Cluster, Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan Malaysia
| |
Collapse
|
202
|
Durbin AP. Dengue vascular leak syndrome: insights into potentially new treatment modalities. J Clin Invest 2020; 129:4072-4073. [PMID: 31449055 DOI: 10.1172/jci131170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Dengue viruses (DENV) are the most common cause of mosquito-borne viral illness in the world, affecting approximately 400 million people annually. Symptomatic illness ranges from a mild, self-limiting febrile illness to one manifested by plasma leakage that can lead to vascular collapse and death. In this issue of the JCI, Rathore et al. report that DENV can cause mast cell degranulation independently of mast cell infection, resulting in the release of the vasoactive mediators chymase and tryptase. The authors showed that recombinant chymase and tryptase increased endothelial permeability in a dose-dependent manner in human microvascular endothelial cells. They went on to evaluate the tryptase inhibitor nafamostat mesylate in a mouse model for severe DENV viremia. Strikingly, the potential therapeutic prevented and reversed the tryptase-induced vascular permeability. As there are currently no licensed drugs for the treatment of dengue, these findings present a possible treatment modality for severe disease.
Collapse
|
203
|
Waggoner JJ, Katzelnick LC, Burger-Calderon R, Gallini J, Moore RH, Kuan G, Balmaseda A, Pinsky BA, Harris E. Antibody-Dependent Enhancement of Severe Disease Is Mediated by Serum Viral Load in Pediatric Dengue Virus Infections. J Infect Dis 2020; 221:1846-1854. [PMID: 32236481 PMCID: PMC7213574 DOI: 10.1093/infdis/jiz618] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/19/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Low preexisting anti-dengue virus (DENV) antibody levels are associated with elevated disease severity. While antibody-dependent enhancement of dengue is thought to be driven by viral load, this has not been conclusively shown. We evaluated the association between preinfection anti-DENV antibody titers, viral load, and disease severity among 133 dengue cases in a Nicaraguan pediatric cohort study. METHODS Viral load was quantified in acute-phase serum by real-time reverse transcription polymerase chain reaction and analyzed in relation to preinfection antibody titer (measured by inhibition enzyme-linked immunosorbent assay) and dengue severity, categorized using 3 definitions. RESULTS Higher viral load was significantly associated with dengue severity; for each increase of 1.0 log10 copies/mL, the odds of severe dengue increased approximately 50%, regardless of severity definition. Viral load at presentation and the odds of severe disease were highest among patients with low to intermediate preinfection antibody titers and lowest among those with the highest antibody titers. We showed the effect of preinfection antibody titer on disease severity was mediated by viral load for each of 3 dengue severity outcomes. CONCLUSIONS This study demonstrates the association between preinfection anti-DENV antibody titer, serum viral load, and disease severity, and provides evidence for the mechanism of antibody-dependent enhancement in dengue cases.
Collapse
Affiliation(s)
- Jesse J Waggoner
- Department of Medicine, Emory University, Atlanta, Georgia, USA
- Department of Global Health, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Leah C Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA
| | | | - Julia Gallini
- Biostatistics Collaboration Core, Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, USA
| | - Renee H Moore
- Biostatistics Collaboration Core, Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, USA
| | - Guillermina Kuan
- Centro de Salud Sócrates Flores Vivas, Ministry of Health, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Benjamin A Pinsky
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
204
|
Esswein SR, Gristick HB, Jurado A, Peace A, Keeffe JR, Lee YE, Voll AV, Saeed M, Nussenzweig MC, Rice CM, Robbiani DF, MacDonald MR, Bjorkman PJ. Structural basis for Zika envelope domain III recognition by a germline version of a recurrent neutralizing antibody. Proc Natl Acad Sci U S A 2020; 117:9865-9875. [PMID: 32321830 PMCID: PMC7211955 DOI: 10.1073/pnas.1919269117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Recent epidemics demonstrate the global threat of Zika virus (ZIKV), a flavivirus transmitted by mosquitoes. Although infection is usually asymptomatic or mild, newborns of infected mothers can display severe symptoms, including neurodevelopmental abnormalities and microcephaly. Given the large-scale spread, symptom severity, and lack of treatment or prophylaxis, a safe and effective ZIKV vaccine is urgently needed. However, vaccine design is complicated by concern that elicited antibodies (Abs) may cross-react with other flaviviruses that share a similar envelope protein, such as dengue virus, West Nile virus, and yellow fever virus. This cross-reactivity may worsen symptoms of a subsequent infection through Ab-dependent enhancement. To better understand the neutralizing Ab response and risk of Ab-dependent enhancement, further information on germline Ab binding to ZIKV and the maturation process that gives rise to potently neutralizing Abs is needed. Here we use binding and structural studies to compare mature and inferred-germline Ab binding to envelope protein domain III of ZIKV and other flaviviruses. We show that affinity maturation of the light-chain variable domain is important for strong binding of the recurrent VH3-23/VK1-5 neutralizing Abs to ZIKV envelope protein domain III, and identify interacting residues that contribute to weak, cross-reactive binding to West Nile virus. These findings provide insight into the affinity maturation process and potential cross-reactivity of VH3-23/VK1-5 neutralizing Abs, informing precautions for protein-based vaccines designed to elicit germline versions of neutralizing Abs.
Collapse
Affiliation(s)
- Shannon R Esswein
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Harry B Gristick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Andrea Jurado
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Avery Peace
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Jennifer R Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Yu E Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Alisa V Voll
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Davide F Robbiani
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125;
| |
Collapse
|
205
|
Rao GK, Prell RA, Laing ST, Burleson SCM, Nguyen A, McBride JM, Zhang C, Sheinson D, Halpern WG. In Vivo Assessment of Antibody-Dependent Enhancement of Influenza B Infection. Toxicol Sci 2020; 169:409-421. [PMID: 30796434 DOI: 10.1093/toxsci/kfz053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A theoretical safety concern proposed in the influenza literature is that therapeutic antiviral antibodies could have the potential for antibody-dependent enhancement (ADE) of infection and disease. ADE may occur when virus-specific antibodies at subtherapeutic, nonneutralizing concentrations facilitate virus uptake and, in some cases, enhance replication, which can lead to an exacerbation of virus-mediated disease. Alternatively, ADE may occur due to antibody-dependent complement activation exacerbating virus-mediated disease in the absence of increased replication. As a result of this theoretical safety concern, safety assessment of anti-influenza antibodies may include an in vivo evaluation of ADE of infection and/or disease. These studies were conducted to investigate the potential of MHAB5553A, a broadly specific, neutralizing therapeutic anti-influenza B antibody, to elicit ADE of infection and disease in mouse models of influenza B infection. In parallel studies, female DBA/2J mice were infected with either influenza B/Victoria/504/2000 or influenza B/Brisbane/60/2008 representing distinct lineages. Assessment of ADE was based on an integration of results from multiple endpoints, including infectious lung viral titers and genomes, body weight, mortality, lung weight, and histopathology. In these studies, the high dose of 15 mg/kg MHAB5553A resulted in substantial attenuation of influenza pneumonia, with more modest effects at 1.5 mg/kg; whereas MHAB5553A treatment at 0.15 or 0.015 mg/kg was generally comparable to vehicle-treated controls. Our results demonstrate that MHAB5553A across a broad range of doses did not enhance primary influenza B infection or disease in this model, and represent a nonclinical de-risking of the ADE potential with this antibody.
Collapse
Affiliation(s)
- Gautham K Rao
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| | - Rodney A Prell
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| | - Steven T Laing
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| | | | | | | | | | - Daniel Sheinson
- Biostatistics, Genentech, Inc., South San Francisco, California 94080
| | - Wendy G Halpern
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
206
|
Boyles SM, Mavian CN, Finol E, Ukhanova M, Stephenson CJ, Hamerlinck G, Kang S, Baumgartner C, Geesey M, Stinton I, Williams K, Mathias DK, Prosperi M, Mai V, Salemi M, Buckner EA, Lednicky JA, Rivers AR, Dinglasan RR. Under-the-Radar Dengue Virus Infections in Natural Populations of Aedes aegypti Mosquitoes. mSphere 2020; 5:e00316-20. [PMID: 32350095 PMCID: PMC7193045 DOI: 10.1128/msphere.00316-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/12/2020] [Indexed: 12/30/2022] Open
Abstract
The incidence of locally acquired dengue infections increased during the last decade in the United States, compelling a sustained research effort concerning the dengue mosquito vector, Aedes aegypti, and its microbiome, which has been shown to influence virus transmission success. We examined the "metavirome" of four populations of Aedes aegypti mosquitoes collected in 2016 to 2017 in Manatee County, FL. Unexpectedly, we discovered that dengue virus serotype 4 (DENV4) was circulating in these mosquito populations, representing the first documented case of such a phenomenon in the absence of a local DENV4 human case in this county over a 2-year period. We confirmed that all of the mosquito populations carried the same DENV4 strain, assembled its full genome, validated infection orthogonally by reverse transcriptase PCR, traced the virus origin, estimated the time period of its introduction to the Caribbean region, and explored the viral genetic signatures and mosquito-specific virome associations that potentially mediated DENV4 persistence in mosquitoes. We discuss the significance of prolonged maintenance of the DENV4 infections in A. aegypti that occurred in the absence of a DENV4 human index case in Manatee County with respect to the inability of current surveillance paradigms to detect mosquito vector infections prior to a potential local outbreak.IMPORTANCE Since 1999, dengue outbreaks in the continental United States involving local transmission have occurred only episodically and only in Florida and Texas. In Florida, these episodes appear to be coincident with increased introductions of dengue virus into the region through human travel and migration from countries where the disease is endemic. To date, the U.S. public health response to dengue outbreaks has been largely reactive, and implementation of comprehensive arbovirus surveillance in advance of predictable transmission seasons, which would enable proactive preventative efforts, remains unsupported. The significance of our finding is that it is the first documented report of DENV4 transmission to and maintenance within a local mosquito vector population in the continental United States in the absence of a human case during two consecutive years. Our data suggest that molecular surveillance of mosquito populations in high-risk, high-tourism areas of the United States may enable proactive, targeted vector control before potential arbovirus outbreaks.
Collapse
Affiliation(s)
- Sean M Boyles
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
| | - Carla N Mavian
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Esteban Finol
- Institute for Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
| | - Maria Ukhanova
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Epidemiology, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Caroline J Stephenson
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
| | - Gabriela Hamerlinck
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Geography, College of Liberal Arts & Sciences, University of Florida, Gainesville, Florida, USA
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
| | - Seokyoung Kang
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
| | | | - Mary Geesey
- Manatee County Mosquito Control District, Palmetto, Florida, USA
| | - Israel Stinton
- Manatee County Mosquito Control District, Palmetto, Florida, USA
| | - Katie Williams
- Manatee County Mosquito Control District, Palmetto, Florida, USA
| | - Derrick K Mathias
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
- Florida Medical Entomology Laboratory, Institute of Food and Agricultural Sciences, University of Florida, Vero Beach, Florida, USA
| | - Mattia Prosperi
- Department of Epidemiology, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Volker Mai
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Epidemiology, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Marco Salemi
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Eva A Buckner
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
- Manatee County Mosquito Control District, Palmetto, Florida, USA
- Florida Medical Entomology Laboratory, Institute of Food and Agricultural Sciences, University of Florida, Vero Beach, Florida, USA
| | - John A Lednicky
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
| | - Adam R Rivers
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
- Genomics and Bioinformatics Research Unit, Agricultural Research Service, United States Department of Agriculture, Gainesville, Florida, USA
| | - Rhoel R Dinglasan
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
- CDC Southeastern Center of Excellence in Vector Borne Diseases, Gainesville, Florida, USA
| |
Collapse
|
207
|
Akther T, Muraduzzaman AKM, Parvin SM, Tabssum S, Munshi SU. Molecular & serological study of dengue virus-infected patients attending a tertiary hospital of Dhaka city, Bangladesh (2013 to 2016). Indian J Med Res 2020; 150:96-100. [PMID: 31571636 PMCID: PMC6798611 DOI: 10.4103/ijmr.ijmr_738_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Tahmina Akther
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Dhaka 1000, Bangladesh
| | - A K M Muraduzzaman
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Dhaka 1000, Bangladesh
| | - S Monira Parvin
- Department of Virology, Dhaka Medical College, Dhaka 1000, Bangladesh
| | - Shahina Tabssum
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Dhaka 1000, Bangladesh
| | - Saif Ullah Munshi
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Dhaka 1000, Bangladesh
| |
Collapse
|
208
|
Arayasongsak U, Naka I, Ohashi J, Patarapotikul J, Nuchnoi P, Kalambaheti T, Sa-Ngasang A, Chanama S, Chaorattanakawee S. Interferon lambda 1 is associated with dengue severity in Thailand. Int J Infect Dis 2020; 93:121-125. [PMID: 31981768 DOI: 10.1016/j.ijid.2020.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES Patients with dengue exhibit a range of symptoms from an acute febrile illness (dengue fever, DF), to dengue hemorrhagic fever (DHF), and to the most severe outcome, dengue shock syndrome (DSS). This study was performed to determine the host genetic factors responsible for dengue severity. Two single nucleotide polymorphisms (SNPs) of the interferon lambda 1 (IFNL1) gene (rs30461 and rs7247086) were analyzed for their association with dengue severity in a Thai population. METHODS This was a case-control association study involving 877 patients under the age of 15 years (DF, n = 386; DHF, n = 416; DSS, n = 75). Genotyping was performed by TaqMan real-time PCR assay. RESULTS It was found that the rs7247086 variant of IFNL1 was associated with DHF, but not DSS. Genotypes CT and TT and the T allele were protective against DHF (p = 0.03, odds ratio 0.62 for CT, odds ratio 0.13 for TT; and p = 0.01, odds ratio 0.54 for the T allele). The other SNP tested was not associated with DHF or DSS. CONCLUSIONS The rs7247086 variant of IFNL1 (the T allele) was found to be protective against DHF, suggesting that IFNL1 may play a role in the pathogenesis of DHF.
Collapse
Affiliation(s)
- Unchana Arayasongsak
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Bangkok 10400, Thailand
| | - Izumi Naka
- Laboratory of Human Genome Diversity, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jun Ohashi
- Laboratory of Human Genome Diversity, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jintana Patarapotikul
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Bangkok 10400, Thailand
| | - Pornlada Nuchnoi
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Thareerat Kalambaheti
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Ratchawithi Road, Bangkok 10400, Thailand
| | - Areerat Sa-Ngasang
- National Institute of Health, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Sumalee Chanama
- National Institute of Health, Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | - Suwanna Chaorattanakawee
- Department of Parasitology and Entomology, Faculty of Public Health, Mahidol University, Ratchawithi Road, Bangkok 10400, Thailand.
| |
Collapse
|
209
|
Non-Human Primate Models of Dengue Virus Infection: A Comparison of Viremia Levels and Antibody Responses during Primary and Secondary Infection among Old World and New World Monkeys. Pathogens 2020; 9:pathogens9040247. [PMID: 32230836 PMCID: PMC7238212 DOI: 10.3390/pathogens9040247] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022] Open
Abstract
Due to the global burden of dengue disease, a vaccine is urgently needed. One of the key points in vaccine development is the development of a robust and reliable animal model of dengue virus infection. Characteristics including the ability to sustain viral replication, demonstration of clinical signs, and immune response that resemble those of human dengue virus infection are vital in animal models. Preclinical studies in vaccine development usually include parameters such as safety evaluation, induction of viremia and antigenemia, immunogenicity, and vaccine effectiveness. Although mice have been used as a model, non-human primates have an advantage over mice because of their relative similarity to humans in their genetic composition and immune responses. This review compares the viremia kinetics and antibody responses of cynomolgus macaques (Macaca fasicularis), common marmosets (Callithrix jacchus), and tamarins (Saguinus midas and Saguinus labitus) and summarize the perspectives and the usefulness along with challenges in dengue vaccine development.
Collapse
|
210
|
Wang WH, Urbina AN, Chang MR, Assavalapsakul W, Lu PL, Chen YH, Wang SF. Dengue hemorrhagic fever - A systemic literature review of current perspectives on pathogenesis, prevention and control. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:963-978. [PMID: 32265181 DOI: 10.1016/j.jmii.2020.03.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/26/2020] [Accepted: 03/08/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Dengue is an arboviral disease caused by dengue virus. Symptomatic dengue infection causes a wide range of clinical manifestations, from mild dengue fever (DF) to potentially fatal disease, such as dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS). We conducted a literature review to analyze the risks of DHF and current perspectives for DHF prevention and control. METHODS According to the PRISMA guidelines, the references were selected from PubMed, Web of Science and Google Scholar database using search strings containing a combination of terms that included dengue hemorrhagic fever, pathogenesis, prevention and control. Quality of references were evaluated by independent reviewers. RESULTS DHF was first reported in the Philippines in 1953 and further transmitted to the countries in the region of South-East Asia and Western Pacific. Plasma leakages is the main pathophysiological hallmark that distinguishes DHF from DF. Severe plasma leakage can result in hypovolemic shock. Various factors are thought to impact disease presentation and severity. Virus virulence, preexisting dengue antibodies, immune dysregulation, lipid change and host genetic susceptibility are factors reported to be correlated with the development of DHF. However, the exact reasons and mechanisms that triggers DHF remains controversial. Currently, no specific drugs and licensed vaccines are available to treat dengue disease in any of its clinical presentations. CONCLUSION This study concludes that antibody-dependent enhancement, cytokine dysregulation and variation of lipid profiles are correlated with DHF occurrence. Prompt diagnosis, appropriate treatment, active and continuous surveillance of cases and vectors are the essential determinants for dengue prevention and control.
Collapse
Affiliation(s)
- Wen-Hung Wang
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Aspiro Nayim Urbina
- Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan.
| | - Max R Chang
- Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan.
| | - Wanchai Assavalapsakul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Po-Liang Lu
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Yen-Hsu Chen
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Sheng-Fan Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
211
|
Kang JY, Aldstadt J, Vandewalle R, Yin D, Wang S. A cyberGIS approach to spatiotemporally explicit uncertainty and global sensitivity analysis for agent-based modeling of vector-borne disease transmission. ANNALS OF THE AMERICAN ASSOCIATION OF GEOGRAPHERS 2020; 110:1855-1873. [PMID: 35106407 PMCID: PMC8803269 DOI: 10.1080/24694452.2020.1723400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/27/2019] [Accepted: 11/04/2019] [Indexed: 06/14/2023]
Abstract
While agent-based models (ABMs) provide an effective means for investigating complex interactions between heterogeneous agents and their environment, they may hinder an improved understanding of phenomena being modeled due to inherent challenges associated with uncertainty in model parameters. This study uses uncertainty analysis and global sensitivity analysis (UA-GSA) to examine the effects of such uncertainty on model outputs. The statistics used in UA-GSA, however, are likely to be affected by the modifiable areal unit problem (MAUP). Therefore, to examine the scale varying-effects of model inputs, UA-GSA needs to be performed at multiple spatiotemporal scales. Unfortunately, performing comprehensive UA-GSA comes with considerable computational cost. In this paper, our cyberGIS-enabled spatiotemporally explicit UA-GSA approach helps to not only resolve the computational burden, but also to measure dynamic associations between model inputs and outputs. A set of computational and modeling experiments shows that input factors have scale-dependent impacts on modeling output variability. In other words, most of the input factors have relatively large impacts in a certain region, but may not influence outcomes in other regions. Furthermore, our spatiotemporally explicit UA-GSA approach sheds light on the effects of input factors on modeling outcomes that are particularly spatially and temporally clustered, such as the occurrence of communicable disease transmission.
Collapse
Affiliation(s)
- Jeon-Young Kang
- CyberGIS Center for Advanced Digital and Spatial Studies; Department of Geography and Geographic Information Science, University of Illinois at Urbana-Champaign
| | - Jared Aldstadt
- Department of Geography, State University of New York at Buffalo
| | - Rebecca Vandewalle
- CyberGIS Center for Advanced Digital and Spatial Studies; Department of Geography and Geographic Information Science, University of Illinois at Urbana-Champaign
| | - Dandong Yin
- CyberGIS Center for Advanced Digital and Spatial Studies; Department of Geography and Geographic Information Science, University of Illinois at Urbana-Champaign
| | - Shaowen Wang
- CyberGIS Center for Advanced Digital and Spatial Studies; Department of Geography and Geographic Information Science, University of Illinois at Urbana-Champaign
| |
Collapse
|
212
|
Non-structural protein 1 (NS1) of dengue virus detection correlates with severity in primary but not in secondary dengue infection. J Clin Virol 2020; 124:104259. [PMID: 31968278 DOI: 10.1016/j.jcv.2020.104259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/27/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Non-structural protein 1 (NS1) of dengue virus circulates in the serum of patients during the acute phase of the disease. OBJECTIVES To determine whether NS1 screening can serve in diagnosing primary and secondary infection and to evaluate its utility as a marker for predicting the severity of dengue in children. STUDY DESIGN Patients ≤15 years of age hospitalized for dengue between 2012-2018, with NS1 determination (Panbio, Australia) were included. Clinical y laboratorial characteristics were collected in a standardized data table for analysis of correlation between serotypes, primary or secondary condition of infection, severity, and presence of NS1. RESULTS Of 709 children hospitalized for dengue with NS1 determination, 479 (67.5 %) had the positive test. Of the 378 primary cases, 320 (85 %) were NS1 (+). while among the 242 secondary cases only 103 (42.5 %) were NS1 (+) (p < 0001). Of the 479 patients with NS1 (+), 344 (72 %) were warnig-signed cases (WSC) and 94 (19 %) were severe cases (SC), being these figures 62 % and 34 %, in the NS1 negative patients respectively (p < 0.001). There was no difference in the frequency of WSC or SC between patients with NS1 positive or negative test in secondary dengue; however, in primary dengue, the figures were 68 % vs 32 % (p < 0.001), and 87 % vs 12 % (p < 0.001), respectively. CONCLUSIONS The presence of NS1 positive test is associated with the condition of infection (primary or secondary) and exhibited an increased risk of developing forms with warning signs or severe dengue in primary cases, but not in secondary cases.
Collapse
|
213
|
|
214
|
Castanha PMS, Erdos G, Watkins SC, Falo LD, Marques ETA, Barratt-Boyes SM. Reciprocal immune enhancement of dengue and Zika virus infection in human skin. JCI Insight 2020; 5:133653. [PMID: 31910161 DOI: 10.1172/jci.insight.133653] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) are closely related mosquito-borne flaviviruses that co-circulate in tropical regions and constitute major threats to global human health. Whether preexisting immunity to one virus affects disease caused by the other during primary or secondary infections is unknown but is critical in preparing for future outbreaks and predicting vaccine safety. Using a human skin explant model, we show that DENV-3 immune sera increased recruitment and infection of Langerhans cells, macrophages, and dermal dendritic cells following inoculation with DENV-2 or ZIKV. Similarly, ZIKV immune sera enhanced infection with DENV-2. Immune sera increased migration of infected Langerhans cells to the dermis and emigration of infected cells out of skin. Heterotypic immune sera increased viral RNA in the dermis almost 10-fold and reduced the amount of virus required to infect a majority of myeloid cells by 100- to 1000-fold. Enhancement was associated with cross-reactive IgG and induction of IL-10 expression and was mediated by both CD32 and CD64 Fcγ receptors. These findings reveal that preexisting heterotypic immunity greatly enhances DENV and ZIKV infection, replication, and spread in human skin. This relevant tissue model will be valuable in assessing the efficacy and risk of dengue and Zika vaccines in humans.
Collapse
Affiliation(s)
- Priscila M S Castanha
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Biological Science Institute and Faculty of Medical Science, University of Pernambuco, Recife, Brazil.,Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | | | - Simon C Watkins
- Center for Biologic Imaging.,Department of Cell Biology, and.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Ernesto T A Marques
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Simon M Barratt-Boyes
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
215
|
Diabetic patients suffering dengue are at risk for development of dengue shock syndrome/severe dengue: Emphasizing the impacts of co-existing comorbidity(ies) and glycemic control on dengue severity. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2020; 53:69-78. [DOI: 10.1016/j.jmii.2017.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 12/18/2017] [Accepted: 12/29/2017] [Indexed: 11/24/2022]
|
216
|
Evaluation of the antiviral activity of orlistat (tetrahydrolipstatin) against dengue virus, Japanese encephalitis virus, Zika virus and chikungunya virus. Sci Rep 2020; 10:1499. [PMID: 32001767 PMCID: PMC6992670 DOI: 10.1038/s41598-020-58468-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/15/2020] [Indexed: 11/20/2022] Open
Abstract
Many mosquito transmitted viruses of the genera Alphavirus and Flavivirus are human pathogens of significant concern, and there is currently no specific antiviral for any member of these two genera. This study sought to investigate the broad utility of orlistat (tetrahydrolipstatin) in reducing virus infection for several mosquito borne viruses including flaviviruses (dengue virus (DENV; nine isolates analyzed), Japanese encephalitis virus (JEV; one isolate analyzed) and Zika virus (ZIKV; 2 isolates analyzed)) as well as an alphavirus (chikungunya virus; CHIKV; 2 isolates analyzed). Three different treatment regimens were evaluated, namely pre-treatment (only), post-treatment (only) and pre- and post-treatment, and three factors were evaluated, namely level of infection, virus titer and genome copy number. Results showed that all three treatment modalities were able to significantly reduce virus titer for all viruses investigated, with the exception of three isolates of DENV in the pre-treatment only regimen. Pre- and post-treatment was more effective in reducing the level of infection and genome copy number of all viruses investigated than either pre-treatment or post-treatment alone. Collectively, these results suggest orlistat has potential as a broad-spectrum agent against multiple mosquito transmitted viruses.
Collapse
|
217
|
Robinson M, Einav S. Towards Predicting Progression to Severe Dengue. Trends Microbiol 2020; 28:478-486. [PMID: 31982232 DOI: 10.1016/j.tim.2019.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/04/2019] [Accepted: 12/09/2019] [Indexed: 12/30/2022]
Abstract
There is an urgent need for prognostic assays to predict progression to severe dengue infection, which is a major global threat. While the majority of symptomatic dengue patients experience an acute febrile illness, 5-20% progress to severe infection associated with significant morbidity and mortality. Early monitoring and administration of supportive care reduce mortality and clinically usable biomarkers to predict severe dengue are needed. Here, we review recent discoveries of gene sets, anti-dengue antibody properties, and inflammatory markers with potential utility as predictors of disease progression. Upon larger scale validation and development of affordable sample-to-answer technologies, some of these biomarkers may be utilized to develop the first prognostic assay for improving patient care and allocating healthcare resources more effectively in dengue endemic countries.
Collapse
Affiliation(s)
- Makeda Robinson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shirit Einav
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
218
|
Humanized Mice in Dengue Research: A Comparison with Other Mouse Models. Vaccines (Basel) 2020; 8:vaccines8010039. [PMID: 31979145 PMCID: PMC7157640 DOI: 10.3390/vaccines8010039] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/10/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Dengue virus (DENV) is an arbovirus of the Flaviviridae family and is an enveloped virion containing a positive sense single-stranded RNA genome. DENV causes dengue fever (DF) which is characterized by an undifferentiated syndrome accompanied by fever, fatigue, dizziness, muscle aches, and in severe cases, patients can deteriorate and develop life-threatening vascular leakage, bleeding, and multi-organ failure. DF is the most prevalent mosquito-borne disease affecting more than 390 million people per year with a mortality rate close to 1% in the general population but especially high among children. There is no specific treatment and there is only one licensed vaccine with restricted application. Clinical and experimental evidence advocate the role of the humoral and T-cell responses in protection against DF, as well as a role in the disease pathogenesis. A lot of pro-inflammatory factors induced during the infectious process are involved in increased severity in dengue disease. The advances in DF research have been hampered by the lack of an animal model that recreates all the characteristics of this disease. Experiments in nonhuman primates (NHP) had failed to reproduce all clinical signs of DF disease and during the past decade, humanized mouse models have demonstrated several benefits in the study of viral diseases affecting humans. In DENV studies, some of these models recapitulate specific signs of disease that are useful to test drugs or vaccine candidates. However, there is still a need for a more complete model mimicking the full spectrum of DENV. This review focuses on describing the advances in this area of research.
Collapse
|
219
|
Chen LH, Wilson ME. Yellow fever control: current epidemiology and vaccination strategies. TROPICAL DISEASES TRAVEL MEDICINE AND VACCINES 2020; 6:1. [PMID: 31938550 PMCID: PMC6954598 DOI: 10.1186/s40794-020-0101-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/05/2020] [Indexed: 12/16/2022]
Abstract
Yellow fever (YF) outbreaks continue, have expanded into new areas and threaten large populations in South America and Africa. Predicting where epidemics might occur must take into account local mosquito populations and specific YF virus strain, as well as ecoclimatic conditions, sociopolitical and demographic factors including population size, density, and mobility, and vaccine coverage. Populations of Aedes aegypti and Aedes albopictus from different regions vary in susceptibility to and capacity to transmit YF virus. YF virus cannot be eliminated today because the virus circulates in animal reservoirs, but human disease could be eliminated with wide use of the vaccine. WHO EYE (Eliminate Yellow Fever Epidemics) is a welcome plan to control YF, with strategies to be carried out from 2017 to 2026: to expand use of YF vaccine, to prevent international spread, and to contain outbreaks rapidly. YF vaccination is the mainstay in controlling YF outbreaks, but global supply is insufficient. Therefore, dose-sparing strategies have been proposed including fractional dosing and intradermal administration. Fractional dosing has been effectively used in outbreak control but currently does not satisfy International Health Regulations; special documentation is needed for international travel. Vector control is another facet in preventing YF outbreaks, and novel methods are being considered and proposed.
Collapse
Affiliation(s)
- Lin H Chen
- 1Mount Auburn Hospital, 330 Mount Auburn Street, Cambridge, MA 02138 USA.,2Harvard Medical School, Boston, MA USA
| | - Mary E Wilson
- 3Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA USA.,4Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, USA
| |
Collapse
|
220
|
Zimmerman MG, Wrammert J, Suthar MS. Cross-Reactive Antibodies during Zika Virus Infection: Protection, Pathogenesis, and Placental Seeding. Cell Host Microbe 2020; 27:14-24. [PMID: 31917957 PMCID: PMC7802743 DOI: 10.1016/j.chom.2019.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Humoral immunity is an essential component of the protective immune response to flavivirus infection. Typically, primary infection generates a robust neutralizing antibody response that mediates viral control and protection. It is becoming increasingly apparent that secondary infection with a closely related flavivirus strain can result in immunological cross-reactivity; however, the consequences to infection outcome remain controversial. Since its introduction to Brazil in 2015, Zika virus (ZIKV) has caused an epidemic of fetal congenital malformations within the Americas. Because ZIKV is a mosquito-borne flavivirus with a high degree of sequence and structural homology to Dengue virus (DENV), the role of immunological cross-reactivity in ZIKV and DENV infections has become a great concern. In this review, we highlight contemporary findings that implicate a role for flavivirus antibodies in mediating protection, contributing to pathogenesis, and seeding the human placenta.
Collapse
Affiliation(s)
- Matthew G Zimmerman
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Jens Wrammert
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Mehul S Suthar
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA.
| |
Collapse
|
221
|
Wong RR, Abd-Aziz N, Affendi S, Poh CL. Role of microRNAs in antiviral responses to dengue infection. J Biomed Sci 2020; 27:4. [PMID: 31898495 PMCID: PMC6941309 DOI: 10.1186/s12929-019-0614-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV) is the etiological agent of dengue fever. Severe dengue could be fatal and there is currently no effective antiviral agent or vaccine. The only licensed vaccine, Dengvaxia, has low efficacy against serotypes 1 and 2. Cellular miRNAs are post-transcriptional regulators that could play a role in direct regulation of viral genes. Host miRNA expressions could either promote or repress viral replications. Induction of some cellular miRNAs could help the virus to evade the host immune response by suppressing the IFN-α/β signaling pathway while others could upregulate IFN-α/β production and inhibit the viral infection. Understanding miRNA expressions and functions during dengue infections would provide insights into the development of miRNA-based therapeutics which could be strategized to act either as miRNA antagonists or miRNA mimics. The known mechanisms of how miRNAs impact DENV replication are diverse. They could suppress DENV multiplication by directly binding to the viral genome, resulting in translational repression. Other miRNA actions include modulation of host factors. In addition, miRNAs that could modulate immunopathogenesis are discussed. Major hurdles lie in the development of chemical modifications and delivery systems for in vivo delivery. Nevertheless, advancement in miRNA formulations and delivery systems hold great promise for the therapeutic potential of miRNA-based therapy, as supported by Miravirsen for treatment of Hepatitis C infection which has successfully completed phase II clinical trial.
Collapse
Affiliation(s)
- Rui Rui Wong
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Noraini Abd-Aziz
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Sarah Affendi
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
222
|
Pare G, Neupane B, Eskandarian S, Harris E, Halstead S, Gresh L, Kuan G, Balmaseda A, Villar L, Rojas E, Osorio JE, Anh DD, De Silva AD, Premawansa S, Premawansa G, Wijewickrama A, Lorenzana I, Parham L, Rodriguez C, Fernandez-Salas I, Sanchez-Casas R, Diaz-Gonzalez EE, Saw Aye K, May WL, Thein M, Bucardo F, Reyes Y, Blandon P, Hirayama K, Weiss L, Singh P, Newton J, Loeb M. Genetic risk for dengue hemorrhagic fever and dengue fever in multiple ancestries. EBioMedicine 2020; 51:102584. [PMID: 31901861 PMCID: PMC6940652 DOI: 10.1016/j.ebiom.2019.11.045] [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] [Received: 08/08/2019] [Revised: 10/28/2019] [Accepted: 11/26/2019] [Indexed: 01/31/2023] Open
Abstract
Background Genetic risk factors for dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS) and dengue fever (DF) are limited, in particular there are sparse data on genetic risk across diverse populations. Methods We conducted a genome-wide association study (GWAS) in a derivation and validation sample of 7, 460 participants of Latin American, South Asian, and South East Asian ancestries. We then developed a weighted polygenic risk score (PRS) for each participant in each of the validation cohorts of the three ancestries to predict the risk of DHF/DSS compared to DF, DHF/DSS compared to controls, and, DF compared to controls. Findings The risk of DHF/DSS was significantly increased, odds ratio [OR] 1.84 (95%CI 1.47 to 2.31) (195 SNPs), compared to DF, fourth PRS quartile versus first quartile, in the validation cohort. The risk of DHF/DSS compared to controls was increased (OR=3.94; 95% CI 2.84 to 5.45) (278 SNPs), as was the risk of DF compared to controls (OR=1.97; 95%CI 1.63 to 2.39) (251 SNPs). Risk increased in a dose-dependent manner with increase in quartiles of PRS across comparisons. Significant associations persisted for PRS built within ancestries and applied to the same or different ancestries as well as for PRS built for one outcome (DHF/DSS or DF) and applied to the other. Interpretation There is a strong genetic effect that predisposes to risk of DHF/DSS and DF. The genetic risk for DHF/DSS is higher than that for DF when compared to controls, and this effect persists across multiple ancestries.
Collapse
Affiliation(s)
- Guillaume Pare
- Department of Pathology and Molecular Medicine, McMaster University, Ontario L8N 3Z5, Canada; Department of Health Research, Methods, Evidence, and Impact, Canada
| | - Binod Neupane
- Department of Pathology and Molecular Medicine, McMaster University, Ontario L8N 3Z5, Canada
| | - Sasha Eskandarian
- Department of Pathology and Molecular Medicine, McMaster University, Ontario L8N 3Z5, Canada
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, United States
| | - Scott Halstead
- Department of Preventive Medicine and Biometrics, Uniformed University of the Health Sciences, Bethesda, MD, United States
| | - Lionel Gresh
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Guillermina Kuan
- Health Center Sócrates Flores Vivas, Ministry of Health, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua; Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Luis Villar
- Clinical Epidemiology Unit, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Elsa Rojas
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas, Bucaramanga, Colombia
| | | | - Dang Duc Anh
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | | | - Sunil Premawansa
- Department of Zoology and Environmental Sciences, University of Colombo, Sri Lanka
| | | | | | - Ivette Lorenzana
- Department of National Autonomous University of Honduras, Tegucigalpa, Honduras
| | - Leda Parham
- Department of National Autonomous University of Honduras, Tegucigalpa, Honduras
| | - Cynthia Rodriguez
- Department of National Autonomous University of Honduras, Tegucigalpa, Honduras
| | | | | | | | | | - Win Lai May
- Medical Research, Ministry of Health, Myanmar
| | - Min Thein
- Medical Research, Ministry of Health, Myanmar
| | - Filemon Bucardo
- The Faculty of Medical Sciences at the National Autonomous University of León, Nicaragua
| | - Yaoska Reyes
- The Faculty of Medical Sciences at the National Autonomous University of León, Nicaragua
| | - Patricia Blandon
- The Faculty of Medical Sciences at the National Autonomous University of León, Nicaragua
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine, Nagasaki University, Nagaski, Japan
| | - Lan Weiss
- Department of Immunogenetics, Institute of Tropical Medicine, Nagasaki University, Nagaski, Japan; Department of Immunology and Microbiology, Pasteur Institute, Ho Chi Minh City, Vietnam
| | - Pardeep Singh
- Department of Pathology and Molecular Medicine, McMaster University, Ontario L8N 3Z5, Canada
| | - Jennifer Newton
- Department of Pathology and Molecular Medicine, McMaster University, Ontario L8N 3Z5, Canada
| | - Mark Loeb
- Department of Pathology and Molecular Medicine, McMaster University, Ontario L8N 3Z5, Canada; Department of Health Research, Methods, Evidence, and Impact, Canada; Institute for Infectious Diseases Research, McMaster University Hamilton, Canada.
| |
Collapse
|
223
|
Abstract
Vaccines are considered one of the most important advances in modern medicine and have greatly improved our quality of life by reducing or eliminating many serious infectious diseases. Successful vaccines have been developed against many of the most common human pathogens, and this success has not been dependent upon any one specific class of vaccine since subunit vaccines, non-replicating whole-virus or whole-bacteria vaccines, and attenuated live vaccines have all been effective for particular vaccine targets. After completing the initial immunization series, one common aspect of successful vaccines is that they induce long-term protective immunity. In contrast, several partially successful vaccines appear to induce protection that is relatively short-lived and it is likely that long-term protective immunity will be critical for making effective vaccines against our most challenging diseases such as AIDS and malaria.
Collapse
Affiliation(s)
- Ian J Amanna
- Najít Technologies, Inc, Beaverton, OR, 97006, USA
| | - Mark K Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA.
| |
Collapse
|
224
|
Adaptive immune responses to primary and secondary dengue virus infections. Nat Rev Immunol 2019; 19:218-230. [PMID: 30679808 DOI: 10.1038/s41577-019-0123-x] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dengue is the leading mosquito-borne viral illness infecting humans. Owing to the circulation of multiple serotypes, global expansion of the disease and recent gains in vaccination coverage, pre-existing immunity to dengue virus is abundant in the human population, and secondary dengue infections are common. Here, we contrast the mechanisms initiating and sustaining adaptive immune responses during primary infection with the immune pathways that are pre-existing and reactivated during secondary dengue. We also discuss new developments in our understanding of the contributions of CD4+ T cells, CD8+ T cells and antibodies to immunity and memory recall. Memory recall may lead to protective or pathological outcomes, and understanding of these processes will be key to developing or refining dengue vaccines to be safe and effective.
Collapse
|
225
|
Nasar S, Rashid N, Iftikhar S. Dengue proteins with their role in pathogenesis, and strategies for developing an effective anti-dengue treatment: A review. J Med Virol 2019; 92:941-955. [PMID: 31784997 DOI: 10.1002/jmv.25646] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022]
Abstract
Dengue virus is an arbovirus belonging to class Flaviviridae Its clinical manifestation ranges from asymptomatic to extreme conditions (dengue hemorrhagic fever/dengue shock syndrome). A lot of research has been done on this ailment, yet there is no effective treatment available for the disease. This review provides the systematic understanding of all dengue proteins, role of its structural proteins (C-protein, E-protein, prM) in virus entry, assembly, and secretion in host cell, and nonstructural proteins (NS1, NS2a, NS2b, NS3, NS4a, NS4b, and NS5) in viral assembly, replication, and immune evasion during dengue progression and pathogenesis. Furthermore, the review has highlighted the controversies related to the only commercially available dengue vaccine, that is, Dengvaxia, and the risk associated with it. Lastly, it provides an insight regarding various approaches for developing an effective anti-dengue treatment.
Collapse
Affiliation(s)
- Sitara Nasar
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Naeem Rashid
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Saima Iftikhar
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
226
|
Blight J, Alves E, Reyes-Sandoval A. Considering Genomic and Immunological Correlates of Protection for a Dengue Intervention. Vaccines (Basel) 2019; 7:E203. [PMID: 31816907 PMCID: PMC6963661 DOI: 10.3390/vaccines7040203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 01/18/2023] Open
Abstract
Over three billion are at risk of dengue infection with more than 100 million a year presenting with symptoms that can lead to deadly haemorrhagic disease. There are however no treatments available and the only licensed vaccine shows limited efficacy and is able to enhance the disease in some cases. These failures have mainly been due to the complex pathology and lack of understanding of the correlates of protection for dengue virus (DENV) infection. With increasing data suggesting both a protective and detrimental effect for antibodies and CD8 T-cells whilst having complex environmental dynamics. This review discusses the roles of genomic and immunological aspects of DENV infection, providing both a historical interpretation and fresh discussion on how this information can be used for the next generation of dengue interventions.
Collapse
Affiliation(s)
- Joshua Blight
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK; (J.B.); (E.A.)
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Eduardo Alves
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK; (J.B.); (E.A.)
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
227
|
Novelo M, Hall MD, Pak D, Young PR, Holmes EC, McGraw EA. Intra-host growth kinetics of dengue virus in the mosquito Aedes aegypti. PLoS Pathog 2019; 15:e1008218. [PMID: 31790509 PMCID: PMC6907869 DOI: 10.1371/journal.ppat.1008218] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/12/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022] Open
Abstract
Dengue virus (DENV) transmission by mosquitoes is a time-dependent process that begins with the consumption of an infectious blood-meal. DENV infection then proceeds stepwise through the mosquito from the midgut to the carcass, and ultimately to the salivary glands, where it is secreted into saliva and then transmitted anew on a subsequent bite. We examined viral kinetics in tissues of the Aedes aegypti mosquito over a finely graded time course, and as per previous studies, found that initial viral dose and serotype strain diversity control infectivity. We also found that a threshold level of virus is required to establish body-wide infections and that replication kinetics in the early and intermediate tissues do not predict those of the salivary glands. Our findings have implications for mosquito GMO design, modeling the contribution of transmission to vector competence and the role of mosquito kinetics in the overall DENV epidemiological landscape. DENV infection in the mosquito is a complex and dynamic process. Following ingestion of an infected blood meal, DENV enters the mosquito midgut epithelial cells, where it replicates. Subsequently, the virus disseminates and infects other tissues, including hemocytes, fat body and reproductive organs, ultimately reaching the salivary glands. The kinetics of infection are influenced by genetic variation in the virus. Comparisons between strains within single serotypes, have revealed variation in infection rates in mosquitoes. To explore the role of infectious dose, serotype and tissue in viral infection kinetics we sampled DENV loads in populations of infected mosquitoes over numerous, sequential time-points. We reveal that the kinetics of DENV infection in the midgut, carcass and salivary glands of the mosquito Aedes aegypti are strikingly different among the strains selected for this study, and that these differences are also driven by the initial infectious dose.
Collapse
Affiliation(s)
- Mario Novelo
- School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
- Center for Infectious Disease Dynamics, Department of Entomology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Matthew D. Hall
- School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Damie Pak
- Center for Infectious Disease Dynamics, Department of Biology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Paul R. Young
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Edward C. Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, School of Life and Environmental Sciences and Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Elizabeth A. McGraw
- School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
- Center for Infectious Disease Dynamics, Department of Entomology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
228
|
Dengue viruses infect human megakaryocytes, with probable clinical consequences. PLoS Negl Trop Dis 2019; 13:e0007837. [PMID: 31765380 PMCID: PMC6901235 DOI: 10.1371/journal.pntd.0007837] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 12/09/2019] [Accepted: 10/10/2019] [Indexed: 11/30/2022] Open
Abstract
One of the most important clinical signs of dengue virus infection is the reduction of white blood cells and platelets in human peripheral blood (leukopenia and thrombocytopenia, respectively), which may significantly impair the clearance of dengue virus by the immune system. The cause of thrombocytopenia and leukopenia during dengue infection is still unknown, but may be related to severe suppression of bone marrow populations including hematopoietic stem cells and megakaryocytes, the progenitors of white blood cells and platelets respectively. Here, we explored the possibility that bone marrow suppression, including ablation of megakaryocyte populations, is caused by dengue virus infection of megakaryocytes. We used three different models to measure dengue virus infection and replication: in vitro, in a human megakaryocyte cell line with viral receptors, ex vivo, in primary human megakaryocytes, and in vivo, in humanized mice. All three systems support dengue virus infection and replication, including virus strains from serotypes 1, 2, and 3, and clinical signs, in vivo; all assays showed viral RNA and/or infectious viruses 7–14 days post-infection. Although we saw no significant decrease in cell viability in vitro, there was significant depletion of mature megakaryocytes in vivo. We conclude that megakaryocytes can produce dengue viruses in the bone marrow niche, and a reduction of cell numbers may affect bone marrow homeostasis. Dengue virus is the most common cause of viral hemorrhagic fever in humans. Over half of the world’s population lives in an at risk area for dengue virus infection, and this number will continue to grow as climate change allows the mosquito vectors of dengue virus to expand their breeding ranges to more temperate climates. Currently, there are no specific treatments for dengue virus infection. Understanding how dengue virus causes hemorrhagic fever could inform the development of these much needed treatments. Populations of important immune system mediators, such as white blood cells and platelets, are significantly dysregulated during dengue virus infection. These cells originate in the bone marrow, which experiences significant suppression, including a complete ablation of megakaryocytes (platelet progenitor cells), during DENV infection. Here, we add to the knowledge on how dengue virus induces bone marrow suppression by investigating whether dengue virus infects human megakaryocytes. We discovered that dengue virus infects human megakaryocytes in vitro, ex vivo, and in vivo models of dengue virus infection; however, dengue virus infection does not appear to directly affect viability of human megakaryocytes. Future studies will investigate whether infected megakaryocytes are still able to perform their functions of producing platelets and maintaining bone marrow homeostasis.
Collapse
|
229
|
Ramesh K, Walvekar VA, Wong B, Sayed AMM, Missé D, Kini RM, Mok YK, Pompon J. Increased Mosquito Midgut Infection by Dengue Virus Recruitment of Plasmin Is Blocked by an Endogenous Kazal-type Inhibitor. iScience 2019; 21:564-576. [PMID: 31726374 PMCID: PMC6854080 DOI: 10.1016/j.isci.2019.10.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/20/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
Dengue symptoms include alteration of blood coagulation and fibrinolysis, causing severe hemorrhage and death. Here, we demonstrate that higher concentration of plasmin, the human fibrinolytic factor, in blood meal enhances dengue virus (DENV) infection in mosquito midgut and dissemination in mosquitoes. We also show that mosquitoes express a plasmin-selective Kazal-type inhibitor (AaTI) in the midgut to inhibit plasmin proteolysis and revert the enhanced infection. Using bio-layer interferometry, we show that DENV, plasmin, and AaTI interact to form a tripartite complex. Eventually, plasmin increases midgut internalization of dextran molecules and this is reverted by AaTI. Our study demonstrates that (1) DENV recruits plasmin to increase local proteolytic activity in the midgut, thus degrading the glycocalyx and enhancing DENV internalization and (2) AaTI can act as a transmission-blocking agent by inhibiting plasmin proteolysis. Our results indicate that dengue pathogenesis enhances DENV fitness by increasing its infectivity to mosquitoes.
Collapse
Affiliation(s)
- Karthik Ramesh
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Varsha A Walvekar
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Benjamin Wong
- Program in Emerging Infectious Disease, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Ahmed Mahmoud Mohammed Sayed
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore; Assiut University, Department of Chemistry, Faculty of Science, Assiut 71516, Egypt
| | - Dorothée Missé
- MIVEGEC, UMR IRD 224-CNRS5290-Université de Montpellier, Montpellier, France
| | - R Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Yu Keung Mok
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore.
| | - Julien Pompon
- Program in Emerging Infectious Disease, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; MIVEGEC, UMR IRD 224-CNRS5290-Université de Montpellier, Montpellier, France.
| |
Collapse
|
230
|
Mapder T, Clifford S, Aaskov J, Burrage K. A population of bang-bang switches of defective interfering particles makes within-host dynamics of dengue virus controllable. PLoS Comput Biol 2019; 15:e1006668. [PMID: 31710599 PMCID: PMC6872170 DOI: 10.1371/journal.pcbi.1006668] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 11/21/2019] [Accepted: 09/27/2019] [Indexed: 02/07/2023] Open
Abstract
The titre of virus in a dengue patient and the duration of this viraemia has a profound effect on whether or not a mosquito will become infected when it feeds on the patient and this, in turn, is a key driver of the magnitude of a dengue outbreak. The assessment of the heterogeneity of viral dynamics in dengue-infected patients and its precise treatment are still uncertain. Infection onset, patient physiology and immune response are thought to play major roles in the development of the viral load. Research has explored the interference and spontaneous generation of defective virus particles, but have not examined both the antibody and defective particles during natural infection. We explore the intrinsic variability in the within-host dynamics of viraemias for a population of patients using the method of population of models (POMs). A dataset from 208 patients is used to initially calibrate 20,000 models for the infection kinetics for each of the four dengue virus serotypes. The calibrated POMs suggests that naturally generated defective particles may interfere with the viraemia, but the generated defective virus particles are not adequate to reduce high fever and viraemia duration. The effect of adding excess defective dengue virus interfering particles to patients as a therapeutic is evaluated using the calibrated POMs in a bang-bang (on-off or two-step) optimal control setting. Bang-bang control is a class of binary feedback control that turns either 'ON' or 'OFF' at different time points, determined by the system feedback. Here, the bang-bang control estimates the mathematically optimal dose and duration of the intervention for each model in the POM set.
Collapse
Affiliation(s)
- Tarunendu Mapder
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Brisbane, Queensland, Australia
- * E-mail: , (TM); (KB)
| | - Sam Clifford
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John Aaskov
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kevin Burrage
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Mathematical and Statistical Frontiers, Queensland University of Technology, Brisbane, Queensland, Australia
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
- * E-mail: , (TM); (KB)
| |
Collapse
|
231
|
Trujillo-Correa AI, Quintero-Gil DC, Diaz-Castillo F, Quiñones W, Robledo SM, Martinez-Gutierrez M. In vitro and in silico anti-dengue activity of compounds obtained from Psidium guajava through bioprospecting. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:298. [PMID: 31694638 PMCID: PMC6836419 DOI: 10.1186/s12906-019-2695-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 09/25/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND For decades, bioprospecting has proven to be useful for the identification of compounds with pharmacological potential. Considering the great diversity of Colombian plants and the serious worldwide public health problem of dengue-a disease caused by the dengue virus (DENV)-in the present study, we evaluated the anti-DENV effects of 12 ethanolic extracts derived from plants collected in the Colombian Caribbean coast, and 5 fractions and 5 compounds derived from Psidium guajava. METHODS The cytotoxicity and antiviral effect of 12 ethanolic extracts derived from plants collected in the Colombian Caribbean coast was evaluated in epithelial VERO cells. Five fractions were obtained by open column chromatography from the ethanolic extract with the highest selectivity index (SI) (derived from P. guajava, SI: 128.2). From the fraction with the highest selectivity (Pg-YP-I-22C, SI: 35.5), five compounds were identified by one- and two-dimensional nuclear magnetic resonance spectroscopy. The antiviral effect in vitro of the fractions and compounds was evaluated by different experimental strategies (Pre- and post-treatment) using non-toxic concentrations calculated by MTT method. The DENV inhibition was evaluated by plate focus assay. The results were analyzed by means of statistical analysis using Student's t-test. Finally the antiviral effect in Silico was evaluated by molecular docking. RESULTS In vitro evaluation of these compounds showed that three of them (gallic acid, quercetin, and catechin) were promising antivirals as they inhibit the production of infectious viral particles via different experimental strategies, with the best antiviral being catechin (100% inhibition with a pre-treatment strategy and 91.8% with a post-treatment strategy). When testing the interactions of these compounds with the viral envelope protein in silico by docking, only naringin and hesperidin had better scores than the theoretical threshold of - 7.0 kcal/mol (- 8.0 kcal/mol and - 8.2 kcal/mol, respectively). All ligands tested except gallic acid showed higher affinity to the NS5 protein than the theoretical threshold. CONCLUSION Even though bioprospecting has recently been replaced by more targeted tools for identifying compounds with pharmacological potential, our results show it is still useful for this purpose. Additionally, combining in vitro and in silico evaluations allowed us to identify promising antivirals as well as their possible mechanisms of action.
Collapse
|
232
|
The Interplay between Dengue Virus and the Human Innate Immune System: A Game of Hide and Seek. Vaccines (Basel) 2019; 7:vaccines7040145. [PMID: 31658677 PMCID: PMC6963221 DOI: 10.3390/vaccines7040145] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
With 40% of the world population at risk, infections with dengue virus (DENV) constitute a serious threat to public health. While there is no antiviral therapy available against this potentially lethal disease, the efficacy of the only approved vaccine is not optimal and its safety has been recently questioned. In order to develop better vaccines based on attenuated and/or chimeric viruses, one must consider how the human immune system is engaged during DENV infection. The activation of the innate immunity through the detection of viruses by cellular sensors is the first line of defence against those pathogens. This triggers a cascade of events which establishes an antiviral state at the cell level and leads to a global immunological response. However, DENV has evolved to interfere with the innate immune signalling at multiple levels, hence dampening antiviral responses and favouring viral replication and dissemination. This review elaborates on the interplay between DENV and the innate immune system. A special focus is given on the viral countermeasure mechanisms reported over the last decade which should be taken into consideration during vaccine development.
Collapse
|
233
|
Carpentier KS, Davenport BJ, Haist KC, McCarthy MK, May NA, Robison A, Ruckert C, Ebel GD, Morrison TE. Discrete viral E2 lysine residues and scavenger receptor MARCO are required for clearance of circulating alphaviruses. eLife 2019; 8:e49163. [PMID: 31596239 PMCID: PMC6839921 DOI: 10.7554/elife.49163] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
The magnitude and duration of vertebrate viremia is a critical determinant of arbovirus transmission, geographic spread, and disease severity. We find that multiple alphaviruses, including chikungunya (CHIKV), Ross River (RRV), and o'nyong 'nyong (ONNV) viruses, are cleared from the circulation of mice by liver Kupffer cells, impeding viral dissemination. Clearance from the circulation was independent of natural antibodies or complement factor C3, and instead relied on scavenger receptor SR-A6 (MARCO). Remarkably, lysine to arginine substitutions at distinct residues within the E2 glycoproteins of CHIKV and ONNV (E2 K200R) as well as RRV (E2 K251R) allowed for escape from clearance and enhanced viremia and dissemination. Mutational analysis revealed that viral clearance from the circulation is strictly dependent on the presence of lysine at these positions. These findings reveal a previously unrecognized innate immune pathway that controls alphavirus viremia and dissemination in vertebrate hosts, ultimately influencing disease severity and likely transmission efficiency.
Collapse
Affiliation(s)
- Kathryn S Carpentier
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Bennett J Davenport
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Kelsey C Haist
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Mary K McCarthy
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Nicholas A May
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Alexis Robison
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Claudia Ruckert
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Gregory D Ebel
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Thomas E Morrison
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| |
Collapse
|
234
|
Abstract
Dengue is the world's most prevalent and important arboviral disease. More than 50% of the world's population lives at daily risk of infection and it is estimated more than 95 million people a year seek medical care following infection. Severe disease can manifest as plasma leakage and potential for clinically significant hemorrhage, shock, and death. Treatment is supportive and there is currently no licensed anti-dengue virus prophylactic or therapeutic compound. A single dengue vaccine, Sanofi Pasteur's Dengvaxia®, has been licensed in 20 countries but uptake has been poor. A safety signal in dengue seronegative vaccine recipients stimulated an international re-look at the vaccine performance profile, new World Health Organization recommendations for use, and controversy in the Philippines involving the government, regulatory agencies, Sanofi Pasteur, clinicians responsible for testing and administering the vaccine, and the parents of vaccinated children. In this review, we provide an overview of Dengvaxia's® development and discuss what has been learned about product performance since its licensure.
Collapse
Affiliation(s)
- Stephen J Thomas
- State University of New York, Upstate Medical University, Division of Infectious Diseases, Institute for Global Health and Translational Sciences , Syracuse , NY , USA
| | - In-Kyu Yoon
- Global Dengue & Aedes-Transmitted Diseases Consortium, International Vaccine Institute, SNU Research Park , Gwanak-gu , Republic of Korea
| |
Collapse
|
235
|
Cui J, Gao L, Chen S, Huang Z, Wang X. Electrochemical voltammetric behaviors of synthetic dengue virus RNAs at ITO sensing electrode. J Electroanal Chem (Lausanne) 2019. [DOI: 10.1016/j.jelechem.2019.113463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
236
|
Dudley DM, Aliota MT, Mohr EL, Newman CM, Golos TG, Friedrich TC, O'Connor DH. Using Macaques to Address Critical Questions in Zika Virus Research. Annu Rev Virol 2019; 6:481-500. [PMID: 31180813 PMCID: PMC7323203 DOI: 10.1146/annurev-virology-092818-015732] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Zika virus (ZIKV) and nonhuman primates have been inextricably linked since the virus was first discovered in a sentinel rhesus macaque in Uganda in 1947. Soon after ZIKV was epidemiologically associated with birth defects in Brazil late in 2015, researchers capitalized on the fact that rhesus macaques are commonly used to model viral immunity and pathogenesis, quickly establishing macaque models for ZIKV infection. Within months, the susceptibility of pregnant macaques to experimental ZIKV challenge and ZIKV-associated abnormalities in fetuses was confirmed. This review discusses key unanswered questions in ZIKV immunity and in the pathogenesis of thecongenital Zika virus syndrome. We focus on those questions that can be best addressed in pregnant nonhuman primates and lessons learned from developing macaque models for ZIKV amid an active epidemic.
Collapse
Affiliation(s)
- Dawn M Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53711, USA; , ,
| | - Matthew T Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Saint Paul, Minnesota 55108, USA;
| | - Emma L Mohr
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53792, USA;
| | - Christina M Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53711, USA; , ,
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA; ,
- Departments of Comparative Biosciences and Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA; ,
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53711, USA; , ,
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA; ,
| |
Collapse
|
237
|
Abstract
Dengue is caused by infection with any one of four dengue viruses (DENV); the risk of severe disease appears to be enhanced by the cross-reactive or subneutralizing levels of antibody from a prior DENV infection. These antibodies opsonize DENV entry through the activating Fc gamma receptors (FcγR), instead of infection through canonical receptor-mediated endocytosis, to result in higher levels of DENV replication. However, whether the enhanced replication is solely due to more efficient FcγR-mediated DENV entry or is also through FcγR-mediated alteration of the host transcriptome response to favor DENV infection remains unclear. Indeed, more efficient viral entry through activation of the FcγR can result in an increased viral antigenic load within target cells and confound direct comparisons of the host transcriptome response under antibody-dependent and antibody-independent conditions. Herein, we show that, despite controlling for the viral antigenic load in primary monocytes, the antibody-dependent and non-antibody-dependent routes of DENV entry induce transcriptome responses that are remarkably different. Notably, antibody-dependent DENV entry upregulated DENV host dependency factors associated with RNA splicing, mitochondrial respiratory chain complexes, and vesicle trafficking. Additionally, supporting findings from other studies, antibody-dependent DENV entry impeded the downregulation of ribosomal genes caused by canonical receptor-mediated endocytosis to increase viral translation. Collectively, our findings support the notion that antibody-dependent DENV entry alters host responses that support the viral life cycle and that host responses to DENV need to be defined in the context of its entry pathway.IMPORTANCE Dengue virus is the most prevalent mosquito-borne viral infection globally, resulting in variable manifestations ranging from asymptomatic viremia to life-threatening shock and multiorgan failure. Previous studies have indicated that the risk of severe dengue in humans can be increased by a specific range of preexisting anti-dengue virus antibody titers, a phenomenon termed antibody-dependent enhancement. There is hence a need to understand how antibodies augment dengue virus infection compared to the alternative canonical receptor-mediated viral entry route. Herein, we show that, besides facilitating viral uptake, antibody-mediated entry increases the expression of early host dependency factors to promote viral infection; these factors include RNA splicing, mitochondrial respiratory chain complexes, vesicle trafficking, and ribosomal genes. These findings will enhance our understanding of how differences in entry pathways can affect host responses and offer opportunities to design therapeutics that can specifically inhibit antibody-dependent enhancement of dengue virus infection.
Collapse
|
238
|
Haltaufderhyde K, Srikiatkhachorn A, Green S, Macareo L, Park S, Kalayanarooj S, Rothman AL, Mathew A. Activation of Peripheral T Follicular Helper Cells During Acute Dengue Virus Infection. J Infect Dis 2019; 218:1675-1685. [PMID: 29917084 DOI: 10.1093/infdis/jiy360] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/12/2018] [Indexed: 12/16/2022] Open
Abstract
Background Follicular helper T cells (TFH) are specialized CD4 T cells required for B-cell help and antibody production. Methods Given the postulated role of immune activation in dengue disease, we measured the expansion and activation of TFH in the circulation (peripheral TFH [pTFH]) collected from Thai children with laboratory-confirmed acute dengue virus (DENV) infection. Results We found significant expansion and activation of pTFH subsets during acute infection with the highest frequencies of activated pTFH (PD1hi pTFH and PD1+CD38+ pTFH) detected during the critical phase of illness. Numbers of activated pTFH were higher in patients with secondary compared with primary infections and in patients with more severe disease. We also found a positive correlation between the frequencies of activated pTFH and the frequencies of plasmablasts. Conclusions To our knowledge, this is the first ex vivo analysis of pTFH activation during acute DENV infection. Overall, our study supports the model that pTFH contribute to disease evolution during the critical stage of illness.
Collapse
Affiliation(s)
- Kirk Haltaufderhyde
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence
| | - Anon Srikiatkhachorn
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence
| | - Sharone Green
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester
| | - Louis Macareo
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sangshin Park
- Center for International Health Research, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence.,Department of Pediatrics, The Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | - Alan L Rothman
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence
| | - Anuja Mathew
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence
| |
Collapse
|
239
|
Jiménez-Morillas F, Gil-Mosquera M, García-Lamberechts EJ. Fever in travellers returning from the tropics. MEDICINA CLINICA (ENGLISH ED.) 2019; 153:205-212. [PMID: 32289079 PMCID: PMC7140248 DOI: 10.1016/j.medcle.2019.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/01/2019] [Indexed: 12/30/2022]
Abstract
The increase in international travel, the growing presence of arbovirus vectors in our country, and notifications of haemorrhagic fever such as the current outbreak of Ebola in D.R. Congo and the cases of Crimea-Congo haemorrhagic fever in our country have again cast the spotlight on tropical diseases. Isolating suspected cases of highly contagious and lethal diseases must be a priority (haemorrhagic fever, MERS-CoV). Assessing the patient, taking a careful medical history based on epidemiological aspects of the area of origin, activities they have carried out, their length of stay in the area and the onset of symptoms, will eventually help us, if not to make a definitive diagnosis, at least to exclude diseases that pose a threat to these patients. Malaria should be ruled out because of its frequency, without forgetting other common causes of fever familiar to emergency doctors.
Collapse
|
240
|
Jiménez-Morillas F, Gil-Mosquera M, García-Lamberechts EJ. Fever in travellers returning from the tropics. Med Clin (Barc) 2019; 153:205-212. [PMID: 31155384 PMCID: PMC7094574 DOI: 10.1016/j.medcli.2019.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 12/30/2022]
Abstract
The increase in international travel, the growing presence of arbovirus vectors in our country, and notifications of haemorrhagic fever such as the current outbreak of Ebola in D.R. Congo and the cases of Crimea-Congo haemorrhagic fever in our country have again cast the spotlight on tropical diseases Isolating suspected cases of highly contagious and lethal diseases must be a priority (Haemorrhagic fever, MERS-CoV). Assessing the patient, taking a careful medical history based on epidemiological aspects of the area of origin, activities they have carried out, their length of stay in the area and the onset of symptoms, will eventually help us, if not to make a definitive diagnosis, at least to exclude diseases that pose a threat to these patients. Malaria should be ruled out because of its frequency, without forgetting other common causes of fever familiar to emergency doctors.
Collapse
|
241
|
Abstract
The process of entry into a host cell is a key step in the life cycle of most viruses. In recent years, there has been a significant increase in our understanding of the routes and mechanisms of entry for a number of these viruses. This has led to the development of novel broad-spectrum antiviral approaches that target host cell proteins and pathways, in addition to strategies focused on individual viruses or virus families. Here we consider a number of these approaches and their broad-spectrum potential.
Collapse
Affiliation(s)
- Michela Mazzon
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Mark Marsh
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
242
|
Kuczera D, Assolini JP, Tomiotto-Pellissier F, Pavanelli WR, Silveira GF. Highlights for Dengue Immunopathogenesis: Antibody-Dependent Enhancement, Cytokine Storm, and Beyond. J Interferon Cytokine Res 2019; 38:69-80. [PMID: 29443656 DOI: 10.1089/jir.2017.0037] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Infection with dengue virus (DENV) can lead to a wide spectrum of clinical presentations, ranging from asymptomatic infection to death. It is estimated that the disease manifests only in 90 million cases out of the total 390 million yearly infections. Even though research has not yet elucidated which are the precise pathophysiological mechanisms that trigger severe forms of dengue, the infection elicits a critical immune response significant for dengue pathogenesis development. Understanding how the immune response to DENV is established and how it can resolve the infection or turn into an immunopathology is of great importance in DENV research. Currently, studies have extensively debated 2 hypotheses involving immune response: antibody-dependent enhancement and cytokine storm. However, despite its undeniable importance in severe forms of the disease, these 2 hypotheses are based on a primed immune status resulting from previous heterologous infection, abstaining them from explaining the severe forms of dengue in naive immune subjects, for example. Thus, it seems that a more intricate arrangement of causes and conditions must be achieved to severe dengue to occur. Among them, the cytokine network signature elicited, in association with viral aspects deserves special attention regarding the establishment of infection and evolution to pathogenesis. In this work, we intend to shed light on how those elements contribute to severe dengue development.
Collapse
Affiliation(s)
- Diogo Kuczera
- 1 Laboratório de Virologia Molecular, Instituto Carlos Chagas , ICC/Fiocruz/PR, Curitiba, Brazil
| | - João Paulo Assolini
- 2 Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina, Londrina, Brazil
| | - Fernanda Tomiotto-Pellissier
- 2 Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina, Londrina, Brazil
| | - Wander Rogério Pavanelli
- 2 Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina, Londrina, Brazil
| | | |
Collapse
|
243
|
Kamaladasa A, Gomes L, Wijesinghe A, Jeewandara C, Toh YX, Jayathilaka D, Ogg GS, Fink K, Malavige GN. Altered monocyte response to the dengue virus in those with varying severity of past dengue infection. Antiviral Res 2019; 169:104554. [PMID: 31288040 DOI: 10.1016/j.antiviral.2019.104554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVE We sought to investigate the differences in monocyte immune responses to the dengue virus (DENV) in those who previously had either severe disease (past SD) or non-severe dengue (past NSD) following a secondary dengue infection. METHOD Monocytes from healthy individuals who had either past SD (n = 6) or past NSD (n = 6) were infected at MOI one with all four DENV serotypes following incubation with autologous serum. 36-hours post infection, levels of inflammatory cytokines and viral loads were measured in the supernatant and expression of genes involved in viral sensing and interferon signaling was determined. RESULTS Monocytes of individuals with past SD produced significantly higher viral loads (p = 0.0426 and cytokines (IL-10 p = 0.008, IL-1β p = 0.008 and IL-6 p = 0.0411) when infected with DENV serotypes they were not immune to, compared to those who has past NSD. Monocytes of individuals with past SD also produced significantly higher viral loads (p = 0.022) and cytokines (IL-10 p < 0.0001, IL-1β < 0.0001 and IL-6 p < 0.0001) when infected with DENV serotypes they were previously exposed to, despite the monocytes being infected in the presence of autologous serum. A significant upregulation of NLRP3 (p = 0.005), RIG-I (0.0004) and IFNB-1 (0.01) genes were observed in those who had past SD compared to past NSD when infected with non-immune DENV serotypes. CONCLUSION Monocytes from those with past SD appear to show marked differences in viral loads, viral sensing and production of inflammatory mediators in response to the DENV, when compared to those who experienced past NSD, suggesting that initial innate immune responses may influence the disease outcome.
Collapse
Affiliation(s)
- Achala Kamaladasa
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | - Laksiri Gomes
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | - Ayesha Wijesinghe
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | | | - Ying Xiu Toh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Deshni Jayathilaka
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | - Graham S Ogg
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre and University of Oxford, OX3 9DS, UK
| | - Katja Fink
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| | - G N Malavige
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre and University of Oxford, OX3 9DS, UK.
| |
Collapse
|
244
|
Robert MA, Christofferson RC, Weber PD, Wearing HJ. Temperature impacts on dengue emergence in the United States: Investigating the role of seasonality and climate change. Epidemics 2019; 28:100344. [PMID: 31175008 PMCID: PMC6791375 DOI: 10.1016/j.epidem.2019.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/02/2019] [Accepted: 05/05/2019] [Indexed: 12/23/2022] Open
Abstract
Tropical mosquito-borne viruses have been expanding into more temperate regions in recent decades. This is partly due to the coupled effects of temperature on mosquito life history traits and viral infection dynamics and warming surface temperatures, resulting in more suitable conditions for vectors and virus transmission. In this study, we use a deterministic ordinary differential equations model to investigate how seasonal and diurnal temperature fluctuations affect the potential for dengue transmission in six U.S. cities. We specifically consider temperature-dependent mosquito larval development, adult mosquito mortality, and the extrinsic incubation period of the virus. We show that the ability of introductions to lead to outbreaks depends upon the relationship between a city's temperature profile and the time of year at which the initial case is introduced. We also investigate how the potential for outbreaks changes with predicted future increases in mean temperatures due to climate change. We find that climate change will likely lead to increases in suitability for dengue transmission and will increase the periods of the year in which introductions may lead to outbreaks, particularly in cities that typically have mild winters and warm summers, such as New Orleans, Louisiana, and El Paso, Texas. We discuss our results in the context of temperature heterogeneity within and across cities and how these differences may impact the potential for dengue emergence given present day and predicted future temperatures.
Collapse
Affiliation(s)
- Michael A Robert
- Department of Biology, University of New Mexico, Albuquerque, NM, United States; Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States; Department of Mathematics, Physics, and Statistics, University of the Sciences, Philadelphia, PA, United States.
| | - Rebecca C Christofferson
- Department of Pathobiology, Louisiana State University, Baton Rouge, LA, United States; Center for Computation and Technology, Louisiana State University, Baton Rouge, LA, United States
| | - Paula D Weber
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States
| | - Helen J Wearing
- Department of Biology, University of New Mexico, Albuquerque, NM, United States; Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
245
|
Islam M, Kalita T, Saikia AK, Begum A, Baruah V, Singh N, Borkotoky R, Bose S. Significance of RANTES-CCR5 axis and linked downstream immunomodulation in Dengue pathogenesis: A study from Guwahati, India. J Med Virol 2019; 91:2066-2073. [PMID: 31368534 DOI: 10.1002/jmv.25561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 07/27/2019] [Indexed: 11/09/2022]
Abstract
We aimed to evaluate the significance of the RANTES-CCR5 axis and resulting immunomodulatory status in Dengue pathogenesis involving a Guwahati, India based population where Dengue cases have increased alarmingly. An increased CC-chemokine receptor type 5 (CCR5) messenger RNA expression and CCR5 positive cell count profile was observed in Dengue cases, the highest being in severe cases. CCR5 ligand RANTES expression was significantly decreased in Dengue cases and inversely correlated with Dengue viremia fold change in severe cases. Monocytes are involved in Dengue virus homing and replication. Its levels and activation profile were higher in Dengue cases. A hyper Th1-biased immunomodulatory profile with upregulated tumor necrosis factor-α levels, and downregulated expression of antiviral cytokine interferon-γ and key regulatory Th2 anti-inflammatory cytokine interleukin 10 was observed in severe Dengue cases compared with mild Dengue cases and controls. The results, therefore, suggest the significance of RANTES-CCR5 axis deregulation and resulting altered immunomodulation in Dengue pathogenesis, and holds prognostic and therapeutic significance.
Collapse
Affiliation(s)
- Mafidul Islam
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Trishna Kalita
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Anjan K Saikia
- Gastroenterology and Hepatology, GNRC Hospital, Guwahati, Assam, India
| | - Anjuma Begum
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Vargab Baruah
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Nidhi Singh
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Raktim Borkotoky
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Sujoy Bose
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| |
Collapse
|
246
|
Arodes ES, Dewi BE, Sudiro TM. Horseradish peroxidase-labeled rabbit anti-non-structural protein 1 of dengue virus-2 for the diagnosis of dengue virus infections. MEDICAL JOURNAL OF INDONESIA 2019. [DOI: 10.13181/mji.v28i2.1951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Early diagnosis of dengue virus (DENV) infection is essential for patient management and disease control. Detection of the antigen non-structural protein 1 (NS1) has been proven to provide early diagnosis of DENV infection. Thus, commercial NS1 antigen detection assays have been increasingly used and are becoming thetool of choice among clinicians to confirm DENV infection in Indonesia.
METHODS To obtain anti-NS1 DENV antibody, NS1 protein (90 µg/ml) from the collection of the Department of Microbiology, Faculty of Medicine, Universitas Indonesia was injected into a rabbit. The anti-NS1 antibody from the rabbit was then labeled with horseradish peroxidase (HRP) using the periodate oxidation method. Sera were tested by enzyme-linked immunosorbent assay (ELISA) to detect NS1 from DENV-infected patients.
RESULTS Serially diluted antibody labeled with HRP tested using the direct ELISA method showed the highest absorbance value at a 1:100 dilution (Mean [SD] = 1.35 [0.35]); even at a dilution as high as 1:3,200 (0.22 [0.15]), antibody labeled with HRP was able to detect the NS1 protein, although the absorbance value did not differ greatly from that of the negative control (0.13 [0.01]).
CONCLUSIONS In an attempt to develop an NS1-based diagnostic test, polyclonal anti-NS1 DENV antibody was successfully produced as a diagnostic assay to determine the presence of DENV NS1 antigen in patients’ sera.
Collapse
|
247
|
Abstract
This is a selective review of recent publications on dengue clinical features, epidemiology, pathogenesis, and vaccine development placed in a context of observations made over the past half century. Four dengue viruses (DENVs) are transmitted by urban cycle mosquitoes causing diseases whose nature and severity are influenced by interacting factors such as virus, age, immune status of the host, and human genetic variability. A phenomenon that controls the kinetics of DENV infection, antibody-dependent enhancement, best explains the correlation of the vascular permeability syndrome with second heterotypic DENV infections and infection in the presence of passively acquired antibodies. Based on growing evidence in vivo and in vitro, the tissue-damaging DENV non-structural protein 1 (NS1) is responsible for most of the pathophysiological features of severe dengue. This review considers the contribution of hemophagocytic histiocytosis syndrome to cases of severe dengue, the role of movement of humans in dengue epidemiology, and modeling and planning control programs and describes a country-wide survey for dengue infections in Bangladesh and efforts to learn what controls the clinical outcome of dengue infections. Progress and problems with three tetravalent live-attenuated vaccines are reviewed. Several research mysteries remain: why is the risk of severe disease during second heterotypic DENV infection so low, why is the onset of vascular permeability correlated with defervescence, and what are the crucial components of protective immunity?
Collapse
Affiliation(s)
- Scott Halstead
- Emeritus Professor, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| |
Collapse
|
248
|
Gonçalves BDS, Nogueira RMR, Bispo de Filippis AM, Horta MAP. Factors predicting the severity of dengue in patients with warning signs in Rio de Janeiro, Brazil (1986–2012). Trans R Soc Trop Med Hyg 2019; 113:670-677. [DOI: 10.1093/trstmh/trz066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/29/2019] [Accepted: 06/25/2019] [Indexed: 01/15/2023] Open
Abstract
AbstractBackgroundSince 1981, >12 million cases of dengue have been reported in Brazil. Early prediction of severe dengue with no warning signs is crucial to avoid progression to severe dengue. Here we aimed to identify early markers of dengue severity and characterize dengue infection in patients in Rio de Janeiro.MethodsWe evaluated early severity markers, serotypes, infection status, number of days of illness and viral loads associated with dengue fever in patients from Rio de Janeiro, Brazil through an observational retrospective study (1986–2012). We compared dengue without warning signs and dengue with warning signs/severe dengue (DWWS/SD). Infection status was classified by enzyme-linked immunosorbent assay and viraemia was quantified by quantitative real-time reverse transcription polymerase chain reaction.ResultsThe presence of DWWS/ SD was significantly associated with younger age; patients 13–19 y of age had a significantly greater chance of presenting warning signs. Dengue virus type 3 (DENV3) was more likely to induce DWWS/SD, which was more frequent on days 4–5 of illness.ConclusionsDENV3, 4–5 d of illness and 13–19 y of age were early biomarkers of dengue severity. To our knowledge, this was the first study to analyse the characteristics of dengue severity in the state of Rio de Janeiro over 27 y of epidemics since the introduction of DENV.
Collapse
Affiliation(s)
- Bianca De Santis Gonçalves
- Flavivirus Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Rita Maria Ribeiro Nogueira
- Flavivirus Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Ana Maria Bispo de Filippis
- Flavivirus Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| | - Marco Aurélio Pereira Horta
- Flavivirus Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
249
|
Nguyen NM, Duong BT, Azam M, Phuong TT, Park H, Thuy PTB, Yeo SJ. Diagnostic Performance of Dengue Virus Envelope Domain III in Acute Dengue Infection. Int J Mol Sci 2019; 20:ijms20143464. [PMID: 31311082 PMCID: PMC6679088 DOI: 10.3390/ijms20143464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
Dengue, one of the most prevalent illnesses caused by dengue viruses that are members of the genus Flavivirus, is a significant global health problem. However, similar clinical symptoms and high antigenic homologies with other Flaviviruses in the endemic area pose difficulties for differential diagnosis of dengue from other arbovirus infections. Here, we investigated four types of recombinant envelope protein domain III (DV-rED III) derived from four dengue virus (DENV) serotypes for diagnostic potential in detecting IgM in acute phase (mainly 2–3 days after onset of fever). Each independent DV-1, -3, and -4-rED III-ELISA showed less than 60% sensitivity, but the combined results of DV-1, -3, and -4-rED III-ELISA led to sensitivity of 81.82% (18/22) (95% CI, 59.72 to 94.81) and 100% specificity (46/46) (95% CI, 92.29 to 100.00) as each antigen compensated the other antigen-derived negative result. In conclusion, the independent combination of data derived from each recombinant antigen (DV1-, DV3-, and DV4-rED III) showed comparable efficacy for the detection of IgM in patients with acute-phase dengue infection.
Collapse
Affiliation(s)
- Ngoc Minh Nguyen
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea
| | - Bao Tuan Duong
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea
| | - Mudsser Azam
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea
| | | | - Hyun Park
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea
| | - Phung Thi Bich Thuy
- Department of Research of Biomolecular for Infectious Disease, National Children's Hospital, Hanoi 100000, Vietnam.
| | - Seon-Ju Yeo
- Zoonosis Research Center, Department of Infection Biology, School of Medicine, Wonkwang University, Iksan 570-749, Korea.
| |
Collapse
|
250
|
Yamanaka A, Konishi E. Intraperitoneal injection with dengue virus type 1-infected K562 cells results in complete fatality among immunocompetent mice. Antiviral Res 2019; 170:104560. [PMID: 31310782 DOI: 10.1016/j.antiviral.2019.104560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/09/2019] [Accepted: 07/13/2019] [Indexed: 11/20/2022]
Abstract
Dengue is one of the most important mosquito-borne viral diseases. Over half of the world's population is living in dengue endemic countries, where 100 million cases are estimated to occur annually. Although one dengue vaccine is currently available commercially, unfortunately its safety and efficacy has not been demonstrated for seronegative populations. Therefore, other vaccine candidates as well as antivirals are urgently required to control dengue diseases. To contribute to the development of preventative measures, in the present study we established an immunocompetent-mouse infection model using dengue virus type 1 Mochizuki strain. Following intraperitoneal injection with K562 cells infected with Mochizuki strain, all mice injected with ≥1 × 106 cells were killed within 7-11 days. Mice injected with ≥1 × 107 cells showed viremia (~104-105 FFU/ml) within 24 h of injection. Since a higher infective titer was detected in the mouse brain, this suggested that viruses were transmitted from the blood circulation into the brain. In further experiments, mice immunized with two types of DNA vaccines were challenged with virus. In contrast to the non-immunized control mice, all vaccinated mice survived after challenge. This immunocompetent-mouse infection model using dengue virus type 1 Mochizuki strain may be a useful tool to evaluate vaccines and preventive medicines against dengue virus.
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|