1
|
Chua CLL, Morales RF, Chia PY, Yeo TW, Teo A. Neutrophils - an understudied bystander in dengue? Trends Microbiol 2024; 32:1132-1142. [PMID: 38749772 DOI: 10.1016/j.tim.2024.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 11/08/2024]
Abstract
Dengue is a mosquito-borne viral disease which causes significant morbidity and mortality each year. Previous research has proposed several mechanisms of pathogenicity that mainly involve the dengue virus and host humoral immunity. However, innate immune cells, such as neutrophils, may also play an important role in dengue, albeit a much less defined role. In this review, we discuss the emerging roles of neutrophils in dengue and their involvement in pathologies associated with severe dengue. We also describe the potential use of several neutrophil proteins as biomarkers for severe dengue. These studies suggest that neutrophils are important players in dengue, and a better understanding of neutrophil-dengue biology is urgently needed.
Collapse
Affiliation(s)
- Caroline Lin Lin Chua
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor's University, Subang Jaya, Malaysia
| | | | - Po Ying Chia
- National Centre for Infectious Diseases, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tsin Wen Yeo
- National Centre for Infectious Diseases, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Andrew Teo
- National Centre for Infectious Diseases, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
2
|
Plaça DR, Fonseca DLM, Marques AHC, Zaki Pour S, Usuda JN, Baiocchi GC, Prado CADS, Salgado RC, Filgueiras IS, Freire PP, Rocha V, Camara NOS, Catar R, Moll G, Jurisica I, Calich VLG, Giil LM, Rivino L, Ochs HD, Cabral-Miranda G, Schimke LF, Cabral-Marques O. Immunological signatures unveiled by integrative systems vaccinology characterization of dengue vaccination trials and natural infection. Front Immunol 2024; 15:1282754. [PMID: 38444851 PMCID: PMC10912564 DOI: 10.3389/fimmu.2024.1282754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Introduction Dengue virus infection is a global health problem lacking specific therapy, requiring an improved understanding of DENV immunity and vaccine responses. Considering the recent emerging of new dengue vaccines, here we performed an integrative systems vaccinology characterization of molecular signatures triggered by the natural DENV infection (NDI) and attenuated dengue virus infection models (DVTs). Methods and results We analyzed 955 samples of transcriptomic datasets of patients with NDI and attenuated dengue virus infection trials (DVT1, DVT2, and DVT3) using a systems vaccinology approach. Differential expression analysis identified 237 common differentially expressed genes (DEGs) between DVTs and NDI. Among them, 28 and 60 DEGs were up or downregulated by dengue vaccination during DVT2 and DVT3, respectively, with 20 DEGs intersecting across all three DVTs. Enriched biological processes of these genes included type I/II interferon signaling, cytokine regulation, apoptosis, and T-cell differentiation. Principal component analysis based on 20 common DEGs (overlapping between DVTs and our NDI validation dataset) distinguished dengue patients by disease severity, particularly in the late acute phase. Machine learning analysis ranked the ten most critical predictors of disease severity in NDI, crucial for the anti-viral immune response. Conclusion This work provides insights into the NDI and vaccine-induced overlapping immune response and suggests molecular markers (e.g., IFIT5, ISG15, and HERC5) for anti-dengue-specific therapies and effective vaccination development.
Collapse
Affiliation(s)
- Desirée Rodrigues Plaça
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Dennyson Leandro M. Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Alexandre H. C. Marques
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Shahab Zaki Pour
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Júlia Nakanishi Usuda
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Gabriela Crispim Baiocchi
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Caroline Aliane de Souza Prado
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Ranieri Coelho Salgado
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Igor Salerno Filgueiras
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Paula Paccielli Freire
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Vanderson Rocha
- Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology and Cell Therapy, Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
- Instituto D’Or de Ensino e Pesquisa, São Paulo, Brazil
- Fundação Pró-Sangue-Hemocentro de São Paulo, São Paulo, Brazil
- Department of Hematology, Churchill Hospital, University of Oxford, Oxford, United Kingdom
| | - Niels Olsen Saraiva Camara
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT) and Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Vera Lúcia Garcia Calich
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lasse M. Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Laura Rivino
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- Emerging Infectious Diseases, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Hans D. Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children’s Research Institute, Seattle, WA, United States
| | - Gustavo Cabral-Miranda
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lena F. Schimke
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, Berlin, Germany
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, SP, Brazil
| | - Otavio Cabral-Marques
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Instituto D’Or de Ensino e Pesquisa, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, Berlin, Germany
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, SP, Brazil
| |
Collapse
|
3
|
Natali EN, Horst A, Meier P, Greiff V, Nuvolone M, Babrak LM, Fink K, Miho E. The dengue-specific immune response and antibody identification with machine learning. NPJ Vaccines 2024; 9:16. [PMID: 38245547 PMCID: PMC10799860 DOI: 10.1038/s41541-023-00788-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/07/2023] [Indexed: 01/22/2024] Open
Abstract
Dengue virus poses a serious threat to global health and there is no specific therapeutic for it. Broadly neutralizing antibodies recognizing all serotypes may be an effective treatment. High-throughput adaptive immune receptor repertoire sequencing (AIRR-seq) and bioinformatic analysis enable in-depth understanding of the B-cell immune response. Here, we investigate the dengue antibody response with these technologies and apply machine learning to identify rare and underrepresented broadly neutralizing antibody sequences. Dengue immunization elicited the following signatures on the antibody repertoire: (i) an increase of CDR3 and germline gene diversity; (ii) a change in the antibody repertoire architecture by eliciting power-law network distributions and CDR3 enrichment in polar amino acids; (iii) an increase in the expression of JNK/Fos transcription factors and ribosomal proteins. Furthermore, we demonstrate the applicability of computational methods and machine learning to AIRR-seq datasets for neutralizing antibody candidate sequence identification. Antibody expression and functional assays have validated the obtained results.
Collapse
Affiliation(s)
- Eriberto Noel Natali
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Alexander Horst
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Patrick Meier
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Victor Greiff
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
| | - Mario Nuvolone
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Lmar Marie Babrak
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | | | - Enkelejda Miho
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- aiNET GmbH, Basel, Switzerland.
| |
Collapse
|
4
|
Shrivastava G, Valenzuela-Leon PC, Botello K, Calvo E. Aedes aegypti saliva modulates inflammasome activation and facilitates flavivirus infection in vitro. iScience 2024; 27:108620. [PMID: 38188518 PMCID: PMC10770497 DOI: 10.1016/j.isci.2023.108620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/16/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Mosquito borne flaviviruses such as dengue and Zika represent a major public health problem due to globalization and propagation of susceptible vectors worldwide. Vertebrate host responses to dengue and Zika infections include the processing and release of pro-inflammatory cytokines through the activation of inflammasomes, resulting in disease severity and fatality. Mosquito saliva can facilitate pathogen infection by downregulating the host's immune response. However, the role of mosquito saliva in modulating host innate immune responses remains largely unknown. Here, we show that mosquito salivary gland extract (SGE) inhibits dengue and Zika virus-induced inflammasome activation by reducing NLRP3 expression, Caspase-1 activation, and 1L-1β secretion in cultured human and mice macrophages. As a result, we observe that SGE inhibits virus detection in the early phase of infection. This study provides important insights into how mosquito saliva modulates host innate immunity during viral infection.
Collapse
Affiliation(s)
- Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| | - Paola Carolina Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| | - Karina Botello
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| |
Collapse
|
5
|
Rawat S, Kumar S, Duggal S, Banerjee A. Phenotypic alteration by dengue virus serotype 2 delays neutrophil apoptosis and stimulates the release of prosurvival secretome with immunomodulatory functions. J Leukoc Biol 2024; 115:276-292. [PMID: 37890093 DOI: 10.1093/jleuko/qiad133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Neutrophils are the most abundant granuloytes, are phenotypically heterogeneous, and exert detrimental or protective roles during antiviral response. Dengue virus has been reported to activate neutrophils. However, the effect of the dengue virus on the neutrophil phenotypes, survival, and release of inflammatory secretome is yet to be understood. Herein, we investigated the effect of dengue virus serotype 2 (DV-2) on effector functions of naïve neutrophils and studied the impact of its secretome on different immune cells. We found that DV-2 activates purified human neutrophils and causes a significant shift toward the CD16bright/CD62Ldim subtype in a multiplicity of infection and time-dependent manner. These phenotypically altered neutrophils show delayed apoptosis through nuclear factor κB and PI3K pathways and have decreased phagocytic capacity. Treatment of neutrophils with myeloperoxidase and PAD4 inhibitor before DV-2 incubation significantly reduced DV-2-induced double-stranded DNA release, suggesting that myeloperoxidase and PAD4 were involved at early stages for the neutrophil activation and double-stranded DNA release. We also report that DV-2-stimulated neutrophil secretome had a significant effect on viral infection, platelet activation, and naïve neutrophil survival via binding of tumor necrosis factor α to tumor necrosis factor receptor 1/2 receptors. Furthermore, incubation of endothelial cells with the DV-2-stimulated neutrophil secretome potentially inhibits proliferation and wound healing capacity and induces endothelial cell death, which can contribute to endothelial barrier dysfunction. In conclusion, the neutrophil-DV-2 interaction modulates the phenotype of neutrophils and the release of prosurvival and antiviral secretome that may act as a double-edged sword during dengue pathogenesis.
Collapse
Affiliation(s)
- Surender Rawat
- Laboratory of Virology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, 121001, Haryana, India
| | - Shubham Kumar
- Laboratory of Virology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, 121001, Haryana, India
| | - Shweta Duggal
- Laboratory of Virology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, 121001, Haryana, India
| | - Arup Banerjee
- Laboratory of Virology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad, 121001, Haryana, India
| |
Collapse
|
6
|
Usuda JN, Plaça DR, Fonseca DLM, Marques AHC, Filgueiras IS, Chaves VGB, Adri AS, Torrentes-Carvalho A, Hirata MH, Freire PP, Catar R, Cabral-Miranda G, Schimke LF, Moll G, Cabral-Marques O. Interferome signature dynamics during the anti-dengue immune response: a systems biology characterization. Front Immunol 2023; 14:1243516. [PMID: 37638052 PMCID: PMC10449254 DOI: 10.3389/fimmu.2023.1243516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Dengue virus (DENV) infection manifests as a febrile illness with three distinct phases: early acute, late acute, and convalescent. Dengue can result in clinical manifestations with different degrees of severity, dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. Interferons (IFNs) are antiviral cytokines central to the anti-DENV immune response. Notably, the distinct global signature of type I, II, and III interferon-regulated genes (the interferome) remains uncharacterized in dengue patients to date. Therefore, we performed an in-depth cross-study for the integrative analysis of transcriptome data related to DENV infection. Our systems biology analysis shows that the anti-dengue immune response is characterized by the modulation of numerous interferon-regulated genes (IRGs) enriching, for instance, cytokine-mediated signaling (e.g., type I and II IFNs) and chemotaxis, which is then followed by a transcriptional wave of genes associated with cell cycle, also regulated by the IFN cascade. The adjunct analysis of disease stratification potential, followed by a transcriptional meta-analysis of the interferome, indicated genes such as IFI27, ISG15, and CYBRD1 as potential suitable biomarkers of disease severity. Thus, this study characterizes the landscape of the interferome signature in DENV infection, indicating that interferome dynamics are a crucial and central part of the anti-dengue immune response.
Collapse
Affiliation(s)
- Júlia Nakanishi Usuda
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Desirée Rodrigues Plaça
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Dennyson Leandro M. Fonseca
- Interunit PostGraduate Program on Bioinformatics, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
| | - Alexandre H. C. Marques
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Igor Salerno Filgueiras
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Victor Gabriel Bastos Chaves
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Anny Silva Adri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paula Paccielli Freire
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rusan Catar
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Gustavo Cabral-Miranda
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F. Schimke
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
- Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Guido Moll
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Otavio Cabral-Marques
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Interunit PostGraduate Program on Bioinformatics, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
- Departament of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
- Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, SP, Brazil
| |
Collapse
|
7
|
Sarkar MMH, Rahman MS, Islam MR, Rahman A, Islam MS, Banu TA, Akter S, Goswami B, Jahan I, Habib MA, Uddin MM, Mia MZ, Miah MI, Shaikh AA, Khan MS. Comparative phylogenetic analysis and transcriptomic profiling of Dengue (DENV-3 genotype I) outbreak in 2021 in Bangladesh. Virol J 2023; 20:127. [PMID: 37337232 PMCID: PMC10278332 DOI: 10.1186/s12985-023-02030-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 04/04/2023] [Indexed: 06/21/2023] Open
Abstract
Background The next-generation sequencing (NGS) technology facilitates in-depth study of host-pathogen metatranscriptome. We, therefore, implicated phylodynamic and transcriptomic approaches through NGS technology to know/understand the dengue virus (DENV) origin and host response with dengue fever. Methods In this study, blood serum RNA was extracted from 21 dengue patients and 3 healthy individuals. Total transcriptomic data were analyzed for phylogenetic, phylodynamic, differential express gene (DEG), and gene ontology (GO) using respective bioinformatics tools. Results The viral genome sequence revealed dengue viral genome size ranges 10647 to 10707 nucleotide. Phylogenetic and phylodynamic analysis showed that the 2021 epidemic isolates were DENV-3 genotype-I and maintained as a new clade in compared to 2019 epidemic. Transcriptome analysis showed a total of 2686 genes were DEG in dengue patients compared to control with a q-value < 0.05. DESeq2 plot counts function of the top 24 genes with the smallest q-values of differential gene expression of RNA-seq data showed that 11 genes were upregulated, whereas 13 genes were downregulated. GO analysis showed a significant upregulation (p = < 0.001) in a process of multicellular organismal, nervous system, sensory perception of chemical stimulus, and G protein-coupled receptor signaling pathways in the dengue patients. However, there were a significant downregulation (p = < 0.001) of intracellular component, cellular anatomical entity, and protein-containing complex in dengue patients. Most importantly, there was a significant increase of a class of immunoregulatory proteins in dengue patients in compared to the controls, with increased GO of immune system process. In addition, upregulation of toll receptor (TLR) signaling pathways were found in dengue patients. These TLR pathways were particularly involved for the activation of innate system coupled with adaptive immune system that probably involved the rapid elimination of dengue virus infected cells. These differentially expressed genes could be further investigated for target based prophylactic interventions for dengue. Conclusion This is a first report describing DENV complete genomic features and differentially expressed genes in patients in Bangladesh. These genes may have diagnostic and therapeutic values for dengue infection. Continual genomic surveillance is required to further investigate the shift in dominant genotypes in relation to viral pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-023-02030-1.
Collapse
Affiliation(s)
| | - M Shaminur Rahman
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - M Rafiul Islam
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - Arafat Rahman
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Tanjina Akhtar Banu
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Shahina Akter
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Barna Goswami
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Iffat Jahan
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Md Ahashan Habib
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Mohammad Mohi Uddin
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Md Zakaria Mia
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Md Ibrahim Miah
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - Aftab Ali Shaikh
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Md Salim Khan
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh.
| |
Collapse
|
8
|
Ioannidis LJ, Studniberg SI, Eriksson EM, Suwarto S, Denis D, Liao Y, Shi W, Garnham AL, Sasmono RT, Hansen DS. Integrated systems immunology approach identifies impaired effector T cell memory responses as a feature of progression to severe dengue fever. J Biomed Sci 2023; 30:24. [PMID: 37055751 PMCID: PMC10103532 DOI: 10.1186/s12929-023-00916-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/02/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Typical symptoms of uncomplicated dengue fever (DF) include headache, muscle pains, rash, cough, and vomiting. A proportion of cases progress to severe dengue hemorrhagic fever (DHF), associated with increased vascular permeability, thrombocytopenia, and hemorrhages. Progression to severe dengue is difficult to diagnose at the onset of fever, which complicates patient triage, posing a socio-economic burden on health systems. METHODS To identify parameters associated with protection and susceptibility to DHF, we pursued a systems immunology approach integrating plasma chemokine profiling, high-dimensional mass cytometry and peripheral blood mononuclear cell (PBMC) transcriptomic analysis at the onset of fever in a prospective study conducted in Indonesia. RESULTS After a secondary infection, progression to uncomplicated dengue featured transcriptional profiles associated with increased cell proliferation and metabolism, and an expansion of ICOS+CD4+ and CD8+ effector memory T cells. These responses were virtually absent in cases progressing to severe DHF, that instead mounted an innate-like response, characterised by inflammatory transcriptional profiles, high circulating levels of inflammatory chemokines and with high frequencies of CD4low non-classical monocytes predicting increased odds of severe disease. CONCLUSIONS Our results suggests that effector memory T cell activation might play an important role ameliorating severe disease symptoms during a secondary dengue infection, and in the absence of that response, a strong innate inflammatory response is required to control viral replication. Our research also identified discrete cell populations predicting increased odds of severe disease, with potential diagnostic value.
Collapse
Affiliation(s)
- Lisa J Ioannidis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Stephanie I Studniberg
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Emily M Eriksson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Suhendro Suwarto
- Division of Tropical and Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo National Hospital (RSCM), Jakarta, Indonesia
| | - Dionisius Denis
- Eijkman Research Center for Molecular Biology, Jakarta, Indonesia
| | - Yang Liao
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - R Tedjo Sasmono
- Eijkman Research Center for Molecular Biology, Jakarta, Indonesia
| | - Diana S Hansen
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
9
|
Garishah FM, Boahen CK, Vadaq N, Pramudo SG, Tunjungputri RN, Riswari SF, van Rij RP, Alisjahbana B, Gasem MH, van der Ven AJAM, de Mast Q. Longitudinal proteomic profiling of the inflammatory response in dengue patients. PLoS Negl Trop Dis 2023; 17:e0011041. [PMID: 36595532 PMCID: PMC9838874 DOI: 10.1371/journal.pntd.0011041] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 01/13/2023] [Accepted: 12/20/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The immunopathogenesis of dengue virus (DENV) infection remains incompletely understood. To increase our understanding of inflammatory response in non-severe dengue, we assessed longitudinal changes in the inflammatory proteome in patients with an acute DENV infection. METHODS Using a multiplex proximity extension assay (PEA), we measured relative levels of 368 inflammatory markers in plasma samples from hospitalized patients with non-severe DENV infection in the acute (n = 43) and convalescence (n = 35) phase of the infection and samples of healthy controls (n = 10). RESULTS We identified 203 upregulated and 39 downregulated proteins in acute versus convalescent plasma samples. The upregulated proteins had a strong representation of interferon (IFN) and IFN-inducible effector proteins, cytokines (e.g. IL-10, IL-33) and cytokine receptors, chemokines, pro-apoptotic proteins (e.g. granzymes) and endothelial markers. A number of differentially expressed proteins (DEPs) have not been reported in previous studies. Functional network analysis highlighted a central role for IFNγ, IL-10, IL-33 and chemokines. We identified different novel associations between inflammatory proteins and circulating concentrations of the endothelial glycocalyx disruption surrogate marker syndecan-1. Conclusion: This unbiased proteome analysis provides a comprehensive insight in the inflammatory response in DENV infection and its association with glycocalyx disruption.
Collapse
Affiliation(s)
- Fadel Muhammad Garishah
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Collins K. Boahen
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nadira Vadaq
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Setyo G. Pramudo
- Department of Internal Medicine, Diponegoro National University Hospital, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
- Department of Internal Medicine, William Booth Hospital, Semarang, Indonesia
| | - Rahajeng N. Tunjungputri
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Silvita Fitri Riswari
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Research Center for Care and Control of Infectious Disease (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Ronald P. van Rij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bachti Alisjahbana
- Research Center for Care and Control of Infectious Disease (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhammad Hussein Gasem
- Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
- Department of Internal Medicine, Diponegoro National University Hospital, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - André J. A. M. van der Ven
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
10
|
Igiri BE, Okoduwa SIR, Munirat SA, Otu-Bassey IB, Bashir A, Onyiyioza OM, Enang IA, Okoduwa UJ. Diversity in Enteric Fever Diagnostic Protocols and Recommendation for Composite Reference Standard. IRANIAN JOURNAL OF MEDICAL MICROBIOLOGY 2023; 17:22-38. [DOI: 10.30699/ijmm.17.1.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
11
|
Srivastava S, Chaudhary N, Dhembla C, Sundd M, Gupta S, Patel AK. STAT3 inhibition mediated upregulation of multiple immune response pathways in dengue infection. Virology 2023; 578:81-91. [PMID: 36473280 DOI: 10.1016/j.virol.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022]
Abstract
Dengue infection is a world-wide public health threat infecting millions of people annually. Till date no specific antiviral or vaccine is available against dengue virus. Recent evidence indicates that targeting host STAT3 could prove to be an effective antiviral therapy against dengue infection. To explore the potential of STAT3 inhibition as an antiviral strategy, we utilized a STAT3 inhibitor stattic as antiviral agent and performed whole proteome analysis of mammalian cells by mass spectrometry. Differentially expressed proteins among the infected and stattic treated groups were sorted based on their fold change expression and their functional annotation studies were carried out to establish their biological networks. The results presented in the current study indicated that treatment with stattic induces several antiviral pathways to counteract dengue infection. Together with this, we also observed that treatment with stattic downregulates pathways involved in viral transcription and translation thus establishing STAT3 as a suitable target for the development of antiviral interventions. This study establishes the role of STAT3 inhibition as an alternative strategy to counteract DENV pathogenesis. Targeting STAT3 by stattic or similar molecules may help in identifying novel therapeutic interventions against DENV and probably other flaviviruses.
Collapse
Affiliation(s)
- Shikha Srivastava
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Nidhi Chaudhary
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chetna Dhembla
- Department of Biochemistry, University of Delhi, South Campus, Benito Juarez Marg, New Delhi, 110021, India
| | - Monica Sundd
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India
| | - Sunny Gupta
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ashok Kumar Patel
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
12
|
Xu G, Gao Y, Pan T, Li S, Zhang Y, Guo J, Tian Z, Xu J, Li Y, Li X. Dynamic immune ecosystem of dengue infection revealed by single-cell sequencing. J Leukoc Biol 2022; 112:1621-1631. [PMID: 35766188 DOI: 10.1002/jlb.6ma0622-738rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/06/2022] [Indexed: 01/04/2023] Open
Abstract
Dengue is the most common human arboviral disease worldwide, which can result in severe complications. A dysfunctional immune response in dengue infective patients is a recurrent theme impacting symptoms and mortality, but the heterogeneity and dynamics of immune infiltrates during dengue infection remain poorly characterized. Here, we identified the immune cell types in scRNA-seq data from 13127 cells of 10 dengue infective patients and discovered the dynamic immune ecosystems of dengue infection. Notably, genes that exhibited higher expression in specific cell types play important roles in response to virus infection in a module manner. Transcription factors (TFs) are the major regulators (i.e., PAX5, IRF7, KLF4, and IRF8) that can potentially regulate infection-related genes. We demonstrated that the dynamic rewired regulatory network during dengue infection. Moreover, our data revealed the complex cell-cell communications from control to fever and severe dengue patients and prevalent cell-cell communication rewiring was observed. We further identified the IFN-II and CXCL signaling pathways that medicated the communications and play important roles in dengue infection. Together, our comprehensive analysis of dynamic immune ecosystem of dengue infection provided novel insights for understanding the pathogenesis of and developing effective therapeutic strategies for dengue infection.
Collapse
Affiliation(s)
- Gang Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Yueying Gao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Tao Pan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Si Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Ya Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Jing Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Zhanyu Tian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yongsheng Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China
| | - Xia Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, China.,College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
13
|
Chia PY, Teo A, Yeo TW. Association of Neutrophil Mediators With Dengue Disease Severity and Cardiac Impairment in Adults. J Infect Dis 2022; 226:1974-1984. [PMID: 36208158 DOI: 10.1093/infdis/jiac383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/19/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cardiac impairment contributes to hypotension in severe dengue (SD). However, studies examining pathogenic factors affecting dengue-associated cardiac impairment are lacking. We examined the role of neutrophil mediators on cardiac impairment in clinical dengue. METHODS We prospectively enrolled adult patients with dengue and controls. Cardiac parameters were measured using a bioimpedance device. Neutrophils mediators were measured, including myeloperoxidase (MPO) and citrullinated histone H3. RESULTS We recruited 107 dengue patients and 30 controls. Patients with dengue were classified according to World Health Organization 2009 guidelines (44 with dengue fever [DF], 51 with DF with warning signs, and 12 with SD). During critical phase, stroke index (P < .001), cardiac index (P = .03), and Granov-Goor index (P < .001) were significantly lower in patients with dengue than in controls. During critical phase, MPO was significantly higher in patients with dengue than in controls (P < .001) and also significantly higher in patients with SD than in those with DF. In addition, MPO was inversely associated with the stroke, cardiac, and Granov-Goor indexes, during the critical phase, and longitudinally as well. CONCLUSIONS Cardiac function was decreased, and MPO increased, during with critical phase in patients SD compared with those with DF and controls. MPO may mediate dengue-associated cardiac impairment.
Collapse
Affiliation(s)
- Po Ying Chia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Department of Medicine, The Doherty Institute, University of Melbourne, Victoria, Australia
| | - Tsin Wen Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| |
Collapse
|
14
|
Niranjan R, Murugasamy V, Sunilkumar A, Manoj H, Ganesh K, Vidhyapriya P, Sankari T, Muthukumaravel S, Kumar A. Atorvastatin attenuates NS1 (Non-structural protein-1) of dengue type-2 serotype-induced expressions of matrix metalloproteinases in HL-60 cells, differentiated to neutrophils: Implications for the immunopathogenesis of dengue viral disease. Int Immunopharmacol 2022; 112:109082. [PMID: 36108401 DOI: 10.1016/j.intimp.2022.109082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND The dengue is a vector borne viral infection in humans. Bite of mosquito infected with a dengue virus transmits the disease. The neutrophils support more to the innate immune response by switching to infected tissues and triggering immunomodulatory mechanisms including the release of proteases and host defence peptides. METHODS Cell viability by MTT and trypan blue dye exclusion assay, bright field microscopy for assessment of cell morphology, cytokines measurements by ELISA, estimation of protein by Bradford assay were done. Assessments of matrix metalloproteinase genes mRNA expressions were done using real-time PCR. RESULTS In the present study, we have for the first time unveiled that, NS1 antigen of dengue type-2 serotype, induce and stimulate the neutrophils cells to express high levels of matrix metalloproteases. NS1 exposure of HL-60 cells differentiated to neutrophils affected cell morphology and in 24 h of exposure. We have demonstrated that, the NS1 antigen has induced MMP-2, MMP-14 and MMP-9 expressions in neutrophils in a 24hrs exposure time. NS1 exposure has also further upregulated MMP-1, MMP-13, and MMP-8 expressions in neutrophils in a 24hrs exposure time. Notably, treatment with atorvastatin concentrations downregulated the expression profile of the all matrix metalloprotease significantly. Importantly, NS1 antigen has significantly increased the IL-6, IL-13 release by the HL,60 cells which was reversed by atorvastatin. On the other hand, NS1 exposure enhanced the mRNA expressions of VEGF-A and VEGF-D which was reversed by atorvastatin. However, we found that, NS1 exposure reduced the mRNA expressions profile of VEGF-C, which was reversed by atorvastatin. CONCLUSION In conclusion, we report that, neutrophils associated matrix metalloprotease are involved in the pathogenesis of dengue viral disease. VEGF growth factors may also be released by the neutrophils which may subsequently participate in the endothelial dysfunctions leading to dengue shock syndrome.
Collapse
Affiliation(s)
- Rituraj Niranjan
- Immunology Laboratory, Division of Microbiology and Immunology, ICMR-Vector Control Research Centre, Puducherry 605006, India.
| | - Vyshali Murugasamy
- Immunology Laboratory, Division of Microbiology and Immunology, ICMR-Vector Control Research Centre, Puducherry 605006, India
| | - Anupama Sunilkumar
- Immunology Laboratory, Division of Microbiology and Immunology, ICMR-Vector Control Research Centre, Puducherry 605006, India
| | - H Manoj
- Immunology Laboratory, Division of Microbiology and Immunology, ICMR-Vector Control Research Centre, Puducherry 605006, India
| | - Khashpatika Ganesh
- Immunology Laboratory, Division of Microbiology and Immunology, ICMR-Vector Control Research Centre, Puducherry 605006, India
| | - Pitchavel Vidhyapriya
- Immunology Laboratory, Division of Microbiology and Immunology, ICMR-Vector Control Research Centre, Puducherry 605006, India
| | - T Sankari
- Division of Omics, ICMR-Vector Control Research Centre, Puducherry 605006, India
| | | | - Ashwani Kumar
- ICMR-Vector Control Research Centre, Puducherry 605006, India
| |
Collapse
|
15
|
Innate Immune Response to Dengue Virus: Toll-like Receptors and Antiviral Response. Viruses 2022; 14:v14050992. [PMID: 35632732 PMCID: PMC9147118 DOI: 10.3390/v14050992] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
Dengue is a mosquito-borne viral disease caused by the dengue virus (DENV1-4). The clinical manifestations range from asymptomatic to life-threatening dengue hemorrhagic fever (DHF) and/or Dengue Shock Syndrome (DSS). Viral and host factors are related to the clinical outcome of dengue, although the disease pathogenesis remains uncertain. The innate antiviral response to DENV is implemented by a variety of immune cells and inflammatory mediators. Blood monocytes, dendritic cells (DCs) and tissue macrophages are the main target cells of DENV infection. These cells recognize pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs). Pathogen recognition is a critical step in eliciting the innate immune response. Toll-like receptors (TLRs) are responsible for the innate recognition of pathogens and represent an essential component of the innate and adaptive immune response. Ten different TLRs are described in humans, which are expressed in many different immune cells. The engagement of TLRs with viral PAMPs triggers downstream signaling pathways leading to the production of inflammatory cytokines, interferons (IFNs) and other molecules essential for the prevention of viral replication. Here, we summarize the crucial TLRs’ roles in the antiviral innate immune response to DENV and their association with viral pathogenesis.
Collapse
|
16
|
Liu YE, Saul S, Rao AM, Robinson ML, Agudelo Rojas OL, Sanz AM, Verghese M, Solis D, Sibai M, Huang CH, Sahoo MK, Gelvez RM, Bueno N, Estupiñan Cardenas MI, Villar Centeno LA, Rojas Garrido EM, Rosso F, Donato M, Pinsky BA, Einav S, Khatri P. An 8-gene machine learning model improves clinical prediction of severe dengue progression. Genome Med 2022; 14:33. [PMID: 35346346 PMCID: PMC8959795 DOI: 10.1186/s13073-022-01034-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Each year 3-6 million people develop life-threatening severe dengue (SD). Clinical warning signs for SD manifest late in the disease course and are nonspecific, leading to missed cases and excess hospital burden. Better SD prognostics are urgently needed. METHODS We integrated 11 public datasets profiling the blood transcriptome of 365 dengue patients of all ages and from seven countries, encompassing biological, clinical, and technical heterogeneity. We performed an iterative multi-cohort analysis to identify differentially expressed genes (DEGs) between non-severe patients and SD progressors. Using only these DEGs, we trained an XGBoost machine learning model on public data to predict progression to SD. All model parameters were "locked" prior to validation in an independent, prospectively enrolled cohort of 377 dengue patients in Colombia. We measured expression of the DEGs in whole blood samples collected upon presentation, prior to SD progression. We then compared the accuracy of the locked XGBoost model and clinical warning signs in predicting SD. RESULTS We identified eight SD-associated DEGs in the public datasets and built an 8-gene XGBoost model that accurately predicted SD progression in the independent validation cohort with 86.4% (95% CI 68.2-100) sensitivity and 79.7% (95% CI 75.5-83.9) specificity. Given the 5.8% proportion of SD cases in this cohort, the 8-gene model had a positive and negative predictive value (PPV and NPV) of 20.9% (95% CI 16.7-25.6) and 99.0% (95% CI 97.7-100.0), respectively. Compared to clinical warning signs at presentation, which had 77.3% (95% CI 58.3-94.1) sensitivity and 39.7% (95% CI 34.7-44.9) specificity, the 8-gene model led to an 80% reduction in the number needed to predict (NNP) from 25.4 to 5.0. Importantly, the 8-gene model accurately predicted subsequent SD in the first three days post-fever onset and up to three days prior to SD progression. CONCLUSIONS The 8-gene XGBoost model, trained on heterogeneous public datasets, accurately predicted progression to SD in a large, independent, prospective cohort, including during the early febrile stage when SD prediction remains clinically difficult. The model has potential to be translated to a point-of-care prognostic assay to reduce dengue morbidity and mortality without overwhelming limited healthcare resources.
Collapse
Affiliation(s)
- Yiran E. Liu
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Cancer Biology Graduate Program, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, CA Stanford, USA
| | - Sirle Saul
- grid.168010.e0000000419368956Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, CA Stanford, USA
| | - Aditya Manohar Rao
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Immunology Graduate Program, School of Medicine, Stanford University, CA Stanford, USA
| | - Makeda Lucretia Robinson
- grid.168010.e0000000419368956Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, CA Stanford, USA
| | | | - Ana Maria Sanz
- grid.477264.4Clinical Research Center, Fundación Valle del Lili, Cali, Colombia
| | - Michelle Verghese
- grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, CA Stanford, USA
| | - Daniel Solis
- grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, CA Stanford, USA
| | - Mamdouh Sibai
- grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, CA Stanford, USA
| | - Chun Hong Huang
- grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, CA Stanford, USA
| | - Malaya Kumar Sahoo
- grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, CA Stanford, USA
| | - Rosa Margarita Gelvez
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Bucaramanga, Colombia
| | - Nathalia Bueno
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Bucaramanga, Colombia
| | | | | | | | - Fernando Rosso
- grid.477264.4Clinical Research Center, Fundación Valle del Lili, Cali, Colombia ,grid.477264.4Division of Infectious Diseases, Department of Internal Medicine, Fundación Valle del Lili, Cali, Colombia
| | - Michele Donato
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Center for Biomedical Informatics Research, Department of Medicine, School of Medicine, Stanford University, CA Stanford, USA
| | - Benjamin A. Pinsky
- grid.168010.e0000000419368956Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, CA Stanford, USA
| | - Shirit Einav
- grid.168010.e0000000419368956Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Department of Microbiology and Immunology, School of Medicine, Stanford University, CA Stanford, USA
| | - Purvesh Khatri
- grid.168010.e0000000419368956Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, CA Stanford, USA ,grid.168010.e0000000419368956Center for Biomedical Informatics Research, Department of Medicine, School of Medicine, Stanford University, CA Stanford, USA
| |
Collapse
|
17
|
Hung SJ, Tsai HP, Wang YF, Ko WC, Wang JR, Huang SW. Assessment of the Risk of Severe Dengue Using Intrahost Viral Population in Dengue Virus Serotype 2 Patients via Machine Learning. Front Cell Infect Microbiol 2022; 12:831281. [PMID: 35223554 PMCID: PMC8866709 DOI: 10.3389/fcimb.2022.831281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Dengue virus, a positive-sense single-stranded RNA virus, continuously threatens human health. Although several criteria for evaluation of severe dengue have been recently established, the ability to prognose the risk of severe outcomes for dengue patients remains limited. Mutant spectra of RNA viruses, including single nucleotide variants (SNVs) and defective virus genomes (DVGs), contribute to viral virulence and growth. Here, we determine the potency of intrahost viral population in dengue patients with primary infection that progresses into severe dengue. A total of 65 dengue virus serotype 2 infected patients in primary infection including 17 severe cases were enrolled. We utilized deep sequencing to directly define the frequency of SNVs and detection times of DVGs in sera of dengue patients and analyzed their associations with severe dengue. Among the detected SNVs and DVGs, the frequencies of 9 SNVs and the detection time of 1 DVG exhibited statistically significant differences between patients with dengue fever and those with severe dengue. By utilizing the detected frequencies/times of the selected SNVs/DVG as features, the machine learning model showed high average with a value of area under the receiver operating characteristic curve (AUROC, 0.966 ± 0.064). The elevation of the frequency of SNVs at E (nucleotide position 995 and 2216), NS2A (nucleotide position 4105), NS3 (nucleotide position 4536, 4606), and NS5 protein (nucleotide position 7643 and 10067) and the detection times of the selected DVG that had a deletion junction in the E protein region (nucleotide positions of the junction: between 969 and 1022) increased the possibility of dengue patients for severe dengue. In summary, we demonstrated the detected frequencies/times of SNVs/DVG in dengue patients associated with severe disease and successfully utilized them to discriminate severe patients using machine learning algorithm. The identified SNVs and DVGs that are associated with severe dengue will expand our understanding of intrahost viral population in dengue pathogenesis.
Collapse
Affiliation(s)
- Su-Jhen Hung
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| | - Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Fang Wang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Ren Wang
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Wen Huang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
- *Correspondence: Sheng-Wen Huang,
| |
Collapse
|
18
|
Affiliation(s)
- Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, Australia
- * E-mail: (AT); (TWY)
| | - Caroline Lin Lin Chua
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Po Ying Chia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Tsin Wen Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- * E-mail: (AT); (TWY)
| |
Collapse
|
19
|
Thach TQ, Eisa HG, Hmeda AB, Faraj H, Thuan TM, Abdelrahman MM, Awadallah MG, Ha NX, Noeske M, Abdul Aziz JM, Nam NH, Nile ME, Dumre SP, Huy NT, Hirayama K. Predictive markers for the early prognosis of dengue severity: A systematic review and meta-analysis. PLoS Negl Trop Dis 2021; 15:e0009808. [PMID: 34610027 PMCID: PMC8519480 DOI: 10.1371/journal.pntd.0009808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 10/15/2021] [Accepted: 09/10/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Predictive markers represent a solution for the proactive management of severe dengue. Despite the low mortality rate resulting from severe cases, dengue requires constant examination and round-the-clock nursing care due to the unpredictable progression of complications, posing a burden on clinical triage and material resources. Accordingly, identifying markers that allow for predicting disease prognosis from the initial diagnosis is needed. Given the improved pathogenesis understanding, myriad candidates have been proposed to be associated with severe dengue progression. Thus, we aim to review the relationship between the available biomarkers and severe dengue. METHODOLOGY We performed a systematic review and meta-analysis to compare the differences in host data collected within 72 hours of fever onset amongst the different disease severity levels. We searched nine bibliographic databases without restrictive criteria of language and publication date. We assessed risk of bias and graded robustness of evidence using NHLBI quality assessments and GRADE, respectively. This study protocol is registered in PROSPERO (CRD42018104495). PRINCIPAL FINDINGS Of 4000 records found, 40 studies for qualitative synthesis, 19 for meta-analysis. We identified 108 host and viral markers collected within 72 hours of fever onset from 6160 laboratory-confirmed dengue cases, including hematopoietic parameters, biochemical substances, clinical symptoms, immune mediators, viral particles, and host genes. Overall, inconsistent case classifications explained substantial heterogeneity, and meta-analyses lacked statistical power. Still, moderate-certainty evidence indicated significantly lower platelet counts (SMD -0.65, 95% CI -0.97 to -0.32) and higher AST levels (SMD 0.87, 95% CI 0.36 to 1.38) in severe cases when compared to non-severe dengue during this time window. CONCLUSION The findings suggest that alterations of platelet count and AST level-in the first 72 hours of fever onset-are independent markers predicting the development of severe dengue.
Collapse
Affiliation(s)
- Tran Quang Thach
- Department of Immunogenetics, Nagasaki University, Nagasaki, Japan
| | - Heba Gamal Eisa
- Faculty of Medicine, Menoufia University, Shebin El-Koum, Egypt
| | | | - Hazem Faraj
- Faculty of Medicine, University of Tripoli, Tripoli, Libya
| | - Tieu Minh Thuan
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | | | - Nam Xuan Ha
- Hue University of Medicine and Pharmacy, Hue, Vietnam
| | - Michael Noeske
- American University of the Caribbean School of Medicine, Cupecoy, Sint Maarten
| | | | - Nguyen Hai Nam
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | - Nguyen Tien Huy
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Nagasaki University, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
20
|
Fontoura MA, Rocha RF, Marques RE. Neutrophil Recruitment and Participation in Severe Diseases Caused by Flavivirus Infection. Life (Basel) 2021; 11:717. [PMID: 34357089 PMCID: PMC8304117 DOI: 10.3390/life11070717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are first-line responders to infections and are recruited to target tissues through the action of chemoattractant molecules, such as chemokines. Neutrophils are crucial for the control of bacterial and fungal infections, but their role in the context of viral infections has been understudied. Flaviviruses are important human viral pathogens transmitted by arthropods. Infection with a flavivirus may result in a variety of complex disease manifestations, including hemorrhagic fever, encephalitis or congenital malformations. Our understanding of flaviviral diseases is incomplete, and so is the role of neutrophils in such diseases. Here we present a comprehensive overview on the participation of neutrophils in severe disease forms evolving from flavivirus infection, focusing on the role of chemokines and their receptors as main drivers of neutrophil function. Neutrophil activation during viral infection was shown to interfere in viral replication through effector functions, but the resulting inflammation is significant and may be detrimental to the host. For congenital infections in humans, neutrophil recruitment mediated by CXCL8 would be catastrophic. Evidence suggests that control of neutrophil recruitment to flavivirus-infected tissues may reduce immunopathology in experimental models and patients, with minimal loss to viral clearance. Further investigation on the roles of neutrophils in flaviviral infections may reveal unappreciated functions of this leukocyte population while increasing our understanding of flaviviral disease pathogenesis in its multiple forms.
Collapse
Affiliation(s)
- Marina Alves Fontoura
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
- Cellular and Structural Biology Graduate Program, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-865, Brazil
| | - Rebeca Fróes Rocha
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
- Genetics and Molecular Biology Graduate Program, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-970, Brazil
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
| |
Collapse
|
21
|
Rawat S, Vrati S, Banerjee A. Neutrophils at the crossroads of acute viral infections and severity. Mol Aspects Med 2021; 81:100996. [PMID: 34284874 PMCID: PMC8286244 DOI: 10.1016/j.mam.2021.100996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022]
Abstract
Neutrophils are versatile immune effector cells essential for mounting a first-line defense against invading pathogens. However, uncontrolled activation can lead to severe life-threatening complications. Neutrophils exist as a heterogeneous population, and their interaction with pathogens and other immune cells may shape the outcome of the host immune response. Diverse classes of viruses, including the recently identified novel SARS-CoV-2, have shown to alter the various aspects of neutrophil biology, offering possibilities for selective intervention. Here, we review heterogeneity within the neutrophil population, highlighting the functional consequences of circulating phenotypes and their critical involvement in exaggerating protective and pathological immune responses against the viruses. We discuss the recent findings of neutrophil extracellular traps (NETs) in COVID-19 pathology and cover other viruses, where neutrophil biology and NETs are crucial for developing disease severity. In the end, we have also pointed out the areas where neutrophil-mediated responses can be finely tuned to outline opportunities for therapeutic manipulation in controlling inflammation against viral infection.
Collapse
Affiliation(s)
- Surender Rawat
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Sudhanshu Vrati
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Arup Banerjee
- Regional Centre for Biotechnology, Faridabad, Haryana, India.
| |
Collapse
|
22
|
Sangkaew S, Ming D, Boonyasiri A, Honeyford K, Kalayanarooj S, Yacoub S, Dorigatti I, Holmes A. Risk predictors of progression to severe disease during the febrile phase of dengue: a systematic review and meta-analysis. THE LANCET. INFECTIOUS DISEASES 2021; 21:1014-1026. [PMID: 33640077 PMCID: PMC8240557 DOI: 10.1016/s1473-3099(20)30601-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 05/01/2020] [Accepted: 06/30/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND The ability to accurately predict early progression of dengue to severe disease is crucial for patient triage and clinical management. Previous systematic reviews and meta-analyses have found significant heterogeneity in predictors of severe disease due to large variation in these factors during the time course of the illness. We aimed to identify factors associated with progression to severe dengue disease that are detectable specifically in the febrile phase. METHODS We did a systematic review and meta-analysis to identify predictors identifiable during the febrile phase associated with progression to severe disease defined according to WHO criteria. Eight medical databases were searched for studies published from Jan 1, 1997, to Jan 31, 2020. Original clinical studies in English assessing the association of factors detected during the febrile phase with progression to severe dengue were selected and assessed by three reviewers, with discrepancies resolved by consensus. Meta-analyses were done using random-effects models to estimate pooled effect sizes. Only predictors reported in at least four studies were included in the meta-analyses. Heterogeneity was assessed using the Cochrane Q and I2 statistics, and publication bias was assessed by Egger's test. We did subgroup analyses of studies with children and adults. The study is registered with PROSPERO, CRD42018093363. FINDINGS Of 6643 studies identified, 150 articles were included in the systematic review, and 122 articles comprising 25 potential predictors were included in the meta-analyses. Female patients had a higher risk of severe dengue than male patients in the main analysis (2674 [16·2%] of 16 481 vs 3052 [10·5%] of 29 142; odds ratio [OR] 1·13 [95% CI 1·01-1·26) but not in the subgroup analysis of studies with children. Pre-existing comorbidities associated with severe disease were diabetes (135 [31·3%] of 431 with vs 868 [16·0%] of 5421 without; crude OR 4·38 [2·58-7·43]), hypertension (240 [35·0%] of 685 vs 763 [20·6%] of 3695; 2·19 [1·36-3·53]), renal disease (44 [45·8%] of 96 vs 271 [16·0%] of 1690; 4·67 [2·21-9·88]), and cardiovascular disease (nine [23·1%] of 39 vs 155 [8·6%] of 1793; 2·79 [1·04-7·50]). Clinical features during the febrile phase associated with progression to severe disease were vomiting (329 [13·5%] of 2432 with vs 258 [6·8%] of 3797 without; 2·25 [1·87-2·71]), abdominal pain and tenderness (321 [17·7%] of 1814 vs 435 [8·1%] of 5357; 1·92 [1·35-2·74]), spontaneous or mucosal bleeding (147 [17·9%] of 822 vs 676 [10·8%] of 6235; 1·57 [1·13-2·19]), and the presence of clinical fluid accumulation (40 [42·1%] of 95 vs 212 [14·9%] of 1425; 4·61 [2·29-9·26]). During the first 4 days of illness, platelet count was lower (standardised mean difference -0·34 [95% CI -0·54 to -0·15]), serum albumin was lower (-0·5 [-0·86 to -0·15]), and aminotransferase concentrations were higher (aspartate aminotransferase [AST] 1·06 [0·54 to 1·57] and alanine aminotransferase [ALT] 0·73 [0·36 to 1·09]) among individuals who progressed to severe disease. Dengue virus serotype 2 was associated with severe disease in children. Secondary infections (vs primary infections) were also associated with severe disease (1682 [11·8%] of 14 252 with vs 507 [5·2%] of 9660 without; OR 2·26 [95% CI 1·65-3·09]). Although the included studies had a moderate to high risk of bias in terms of study confounding, the risk of bias was low to moderate in other domains. Heterogeneity of the pooled results varied from low to high on different factors. INTERPRETATION This analysis supports monitoring of the warning signs described in the 2009 WHO guidelines on dengue. In addition, testing for infecting serotype and monitoring platelet count and serum albumin, AST, and ALT concentrations during the febrile phase of illness could improve the early prediction of severe dengue. FUNDING Wellcome Trust, National Institute for Health Research, Collaborative Project to Increase Production of Rural Doctors, and Royal Thai Government.
Collapse
Affiliation(s)
- Sorawat Sangkaew
- Section of Adult Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK; Department of Social Medicine, Hatyai Hospital, Songkhla, Thailand.
| | - Damien Ming
- Section of Adult Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Adhiratha Boonyasiri
- Section of Adult Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Kate Honeyford
- Global Digital Health Unit, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Siripen Kalayanarooj
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Sophie Yacoub
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Ho Chi Minh City, Vietnam; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Ilaria Dorigatti
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Alison Holmes
- Section of Adult Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK; Antimicrobial Resistance Collaborative, Imperial College London, London, UK
| |
Collapse
|
23
|
Complex Roles of Neutrophils during Arboviral Infections. Cells 2021; 10:cells10061324. [PMID: 34073501 PMCID: PMC8227388 DOI: 10.3390/cells10061324] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Arboviruses are known to cause large-scale epidemics in many parts of the world. These arthropod-borne viruses are a large group consisting of viruses from a wide range of families. The ability of their vector to enhance viral pathogenesis and transmission makes the development of treatments against these viruses challenging. Neutrophils are generally the first leukocytes to be recruited to a site of infection, playing a major role in regulating inflammation and, as a result, viral replication and dissemination. However, the underlying mechanisms through which neutrophils control the progression of inflammation and disease remain to be fully understood. In this review, we highlight the major findings from recent years regarding the role of neutrophils during arboviral infections. We discuss the complex nature of neutrophils in mediating not only protection, but also augmenting disease pathology. Better understanding of neutrophil pathways involved in effective protection against arboviral infections can help identify potential targets for therapeutics.
Collapse
|
24
|
Hanley JP, Tu HA, Dragon JA, Dickson DM, Rio-Guerra RD, Tighe SW, Eckstrom KM, Selig N, Scarpino SV, Whitehead SS, Durbin AP, Pierce KK, Kirkpatrick BD, Rizzo DM, Frietze S, Diehl SA. Immunotranscriptomic profiling the acute and clearance phases of a human challenge dengue virus serotype 2 infection model. Nat Commun 2021; 12:3054. [PMID: 34031380 PMCID: PMC8144425 DOI: 10.1038/s41467-021-22930-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
About 20-25% of dengue virus (DENV) infections become symptomatic ranging from self-limiting fever to shock. Immune gene expression changes during progression to severe dengue have been documented in hospitalized patients; however, baseline or kinetic information is difficult to standardize in natural infection. Here we profile the host immunotranscriptome response in humans before, during, and after infection with a partially attenuated rDEN2Δ30 challenge virus (ClinicalTrials.gov NCT02021968). Inflammatory genes including type I interferon and viral restriction pathways are induced during DENV2 viremia and return to baseline after viral clearance, while others including myeloid, migratory, humoral, and growth factor immune regulation factors pathways are found at non-baseline levels post-viremia. Furthermore, pre-infection baseline gene expression is useful to predict rDEN2Δ30-induced immune responses and the development of rash. Our results suggest a distinct immunological profile for mild rDEN2Δ30 infection and offer new potential biomarkers for characterizing primary DENV infection.
Collapse
Affiliation(s)
- John P Hanley
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Huy A Tu
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Cellular and Molecular Biomedical Sciences Program, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Julie A Dragon
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Integrated Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Dorothy M Dickson
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Roxana Del Rio-Guerra
- Flow Cytometry and Cell Sorting Facility, Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Scott W Tighe
- Vermont Integrated Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Korin M Eckstrom
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Integrated Genomics Resource, University of Vermont, Burlington, VT, USA
| | - Nicholas Selig
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Stephen S Whitehead
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Anna P Durbin
- Center for Immunization Research, Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen K Pierce
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Beth D Kirkpatrick
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Donna M Rizzo
- Department of Civil and Environmental Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT, USA
| | - Seth Frietze
- Cellular and Molecular Biomedical Sciences Program, University of Vermont, Burlington, VT, USA
- Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Sean A Diehl
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- Translational Global Infectious Disease Research Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- Cellular and Molecular Biomedical Sciences Program, University of Vermont, Burlington, VT, USA.
- Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
25
|
Poonpanichakul T, Chan-In W, Opasawatchai A, Loison F, Matangkasombut O, Charoensawan V, Matangkasombut P. Innate Lymphoid Cells Activation and Transcriptomic Changes in Response to Human Dengue Infection. Front Immunol 2021; 12:599805. [PMID: 34079535 PMCID: PMC8165392 DOI: 10.3389/fimmu.2021.599805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 04/29/2021] [Indexed: 12/19/2022] Open
Abstract
Background Dengue virus (DENV) infection has a global impact on public health. The clinical outcomes (of DENV) can vary from a flu-like illness called dengue fever (DF), to a more severe form, known as dengue hemorrhagic fever (DHF). The underlying innate immune mechanisms leading to protective or detrimental outcomes have not been fully elucidated. Helper innate lymphoid cells (hILCs), an innate lymphocyte recently discovered, functionally resemble T-helper cells and are important in inflammation and homeostasis. However, the role of hILCs in DENV infection had been unexplored. Methods We performed flow cytometry to investigate the frequency and phenotype of hILCs in peripheral blood mononuclear cells from DENV-infected patients of different disease severities (DF and DHF), and at different phases (febrile and convalescence) of infection. Intracellular cytokine staining of hILCs from DF and DHF were also evaluated by flow cytometry after ex vivo stimulation. Further, the hILCs were sorted and subjected to transcriptome analysis using RNA sequencing. Differential gene expression analysis was performed to compare the febrile and convalescent phase samples in DF and DHF. Selected differentially expressed genes were then validated by quantitative PCR. Results Phenotypic analysis showed marked activation of all three hILC subsets during the febrile phase as shown by higher CD69 expression when compared to paired convalescent samples, although the frequency of hILCs remained unchanged. Upon ex vivo stimulation, hILCs from febrile phase DHF produced significantly higher IFN-γ and IL-4 when compared to those of DF. Transcriptomic analysis showed unique hILCs gene expression in DF and DHF, suggesting that divergent functions of hILCs may be associated with different disease severities. Differential gene expression analysis indicated that hILCs function both in cytokine secretion and cytotoxicity during the febrile phase of DENV infection. Conclusions Helper ILCs are activated in the febrile phase of DENV infection and display unique transcriptomic changes as well as cytokine production that correlate with severity. Targeting hILCs during early innate response to DENV might help shape subsequent immune responses and potentially lessen the disease severity in the future.
Collapse
Affiliation(s)
- Tiraput Poonpanichakul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand.,Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Wilawan Chan-In
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Anunya Opasawatchai
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Fabien Loison
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Oranart Matangkasombut
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Varodom Charoensawan
- Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand.,Integrative Computational BioScience Center (ICBS), Mahidol University, Nakhon Pathom, Thailand
| | - Ponpan Matangkasombut
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
26
|
Hoang LT, Jain P, Pillay TD, Tolosa-Wright M, Niazi U, Takwoingi Y, Halliday A, Berrocal-Almanza LC, Deeks JJ, Beverley P, Kon OM, Lalvani A. Transcriptomic signatures for diagnosing tuberculosis in clinical practice: a prospective, multicentre cohort study. THE LANCET. INFECTIOUS DISEASES 2021; 21:366-375. [PMID: 33508221 PMCID: PMC7907671 DOI: 10.1016/s1473-3099(20)30928-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/15/2020] [Accepted: 11/23/2020] [Indexed: 12/23/2022]
Abstract
Background Blood transcriptomic signatures for diagnosis of tuberculosis have shown promise in case-control studies, but none have been prospectively designed or validated in adults presenting with the full clinical spectrum of suspected tuberculosis, including extrapulmonary tuberculosis and common differential diagnoses that clinically resemble tuberculosis. We aimed to evaluate the diagnostic accuracy of transcriptomic signatures in patients presenting with clinically suspected tuberculosis in routine practice. Methods The Validation of New Technologies for Diagnostic Evaluation of Tuberculosis (VANTDET) study was nested within a prospective, multicentre cohort study in secondary care in England (IDEA 11/H0722/8). Patients (aged ≥16 years) suspected of having tuberculosis in the routine clinical inpatient and outpatient setting were recruited at ten National Health Service hospitals in England for IDEA and were included in VANTDET if they provided consent for genomic analysis. Patients had whole blood taken for microarray analysis to measure abundance of transcripts and were followed up for 6–12 months to determine final diagnoses on the basis of predefined diagnostic criteria. The diagnostic accuracy of six signatures derived from the cohort and three previously published transcriptomic signatures with potentially high diagnostic performance were assessed by calculating area under the receiver-operating characteristic curves (AUC-ROCs), sensitivities, and specificities. Findings Between Nov 25, 2011, and Dec 31, 2013, 1162 participants were enrolled. 628 participants (aged ≥16 years) were included in the analysis, of whom 212 (34%) had culture-confirmed tuberculosis, 89 (14%) had highly probable tuberculosis, and 327 (52%) had tuberculosis excluded. The novel signature with highest performance for identifying all active tuberculosis gave an AUC-ROC of 0·87 (95% CI 0·81–0·92), sensitivity of 77% (66–87), and specificity of 84% (74–91). The best-performing published signature gave an AUC-ROC of 0·83 (0·80–0·86), sensitivity of 78% (73–83), and specificity of 76% (70–80). For detecting highly probable tuberculosis, the best novel signature yielded results of 0·86 (0·71–0·95), 77% (56–94%), and 77% (57–95%). None of the relevant cohort-derived or previously published signatures achieved the WHO-defined targets of paired sensitivity and specificity for a non-sputum-based diagnostic test. Interpretation In a clinically representative cohort in routine practice in a low-incidence setting, transcriptomic signatures did not have adequate accuracy for diagnosis of tuberculosis, including in patients with highly probable tuberculosis where the unmet need is greatest. These findings suggest that transcriptomic signatures have little clinical utility for diagnostic assessment of suspected tuberculosis. Funding National Institute for Health Research.
Collapse
Affiliation(s)
- Long T Hoang
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Pooja Jain
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Timesh D Pillay
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Mica Tolosa-Wright
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Umar Niazi
- Guy's and St Thomas' National Health Service Foundation Trust and King's College London National Institute for Health Research Biomedical Research Centre Translational Bioinformatics Platform, Guy's Hospital, London, UK
| | - Yemisi Takwoingi
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK; National Institute of Health Research Birmingham Biomedical Research Centre, University Hospitals Birmingham National Health Service Foundation Trust and University of Birmingham, Birmingham, UK
| | - Alice Halliday
- Bristol Children's Vaccine Centre, University of Bristol, UK
| | - Luis C Berrocal-Almanza
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jonathan J Deeks
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK; National Institute of Health Research Birmingham Biomedical Research Centre, University Hospitals Birmingham National Health Service Foundation Trust and University of Birmingham, Birmingham, UK
| | - Peter Beverley
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Onn Min Kon
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK; Tuberculosis Service, Imperial College Healthcare National Health Service Trust, London, UK
| | - Ajit Lalvani
- Tuberculosis Research Centre, National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
27
|
Kim D, Song J, Lee S, Jung J, Jang W. An Integrative Transcriptomic Analysis of Systemic Juvenile Idiopathic Arthritis for Identifying Potential Genetic Markers and Drug Candidates. Int J Mol Sci 2021; 22:ijms22020712. [PMID: 33445803 PMCID: PMC7828236 DOI: 10.3390/ijms22020712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/02/2021] [Accepted: 01/07/2021] [Indexed: 12/20/2022] Open
Abstract
Systemic juvenile idiopathic arthritis (sJIA) is a rare subtype of juvenile idiopathic arthritis, whose clinical features are systemic fever and rash accompanied by painful joints and inflammation. Even though sJIA has been reported to be an autoinflammatory disorder, its exact pathogenesis remains unclear. In this study, we integrated a meta-analysis with a weighted gene co-expression network analysis (WGCNA) using 5 microarray datasets and an RNA sequencing dataset to understand the interconnection of susceptibility genes for sJIA. Using the integrative analysis, we identified a robust sJIA signature that consisted of 2 co-expressed gene sets comprising 103 up-regulated genes and 25 down-regulated genes in sJIA patients compared with healthy controls. Among the 128 sJIA signature genes, we identified an up-regulated cluster of 11 genes and a down-regulated cluster of 4 genes, which may play key roles in the pathogenesis of sJIA. We then detected 10 bioactive molecules targeting the significant gene clusters as potential novel drug candidates for sJIA using an in silico drug repositioning analysis. These findings suggest that the gene clusters may be potential genetic markers of sJIA and 10 drug candidates can contribute to the development of new therapeutic options for sJIA.
Collapse
|
28
|
Assessing the risk of dengue severity using demographic information and laboratory test results with machine learning. PLoS Negl Trop Dis 2020; 14:e0008960. [PMID: 33362244 PMCID: PMC7757819 DOI: 10.1371/journal.pntd.0008960] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dengue virus causes a wide spectrum of disease, which ranges from subclinical disease to severe dengue shock syndrome. However, estimating the risk of severe outcomes using clinical presentation or laboratory test results for rapid patient triage remains a challenge. Here, we aimed to develop prognostic models for severe dengue using machine learning, according to demographic information and clinical laboratory data of patients with dengue. METHODOLOGY/PRINCIPAL FINDINGS Out of 1,581 patients in the National Cheng Kung University Hospital with suspected dengue infections and subjected to NS1 antigen, IgM and IgG, and qRT-PCR tests, 798 patients including 138 severe cases were enrolled in the study. The primary target outcome was severe dengue. Machine learning models were trained and tested using the patient dataset that included demographic information and qualitative laboratory test results collected on day 1 when they sought medical advice. To develop prognostic models, we applied various machine learning methods, including logistic regression, random forest, gradient boosting machine, support vector classifier, and artificial neural network, and compared the performance of the methods. The artificial neural network showed the highest average discrimination area under the receiver operating characteristic curve (0.8324 ± 0.0268) and balance accuracy (0.7523 ± 0.0273). According to the model explainer that analyzed the contributions/co-contributions of the different factors, patient age and dengue NS1 antigenemia were the two most important risk factors associated with severe dengue. Additionally, co-existence of anti-dengue IgM and IgG in patients with dengue increased the probability of severe dengue. CONCLUSIONS/SIGNIFICANCE We developed prognostic models for the prediction of dengue severity in patients, using machine learning. The discriminative ability of the artificial neural network exhibited good performance for severe dengue prognosis. This model could help clinicians obtain a rapid prognosis during dengue outbreaks. However, the model requires further validation using external cohorts in future studies.
Collapse
|
29
|
Rajput R, Sharma J, Nair MT, Khanna M, Arora P, Sood V. Regulation of Host Innate Immunity by Non-Coding RNAs During Dengue Virus Infection. Front Cell Infect Microbiol 2020; 10:588168. [PMID: 33330133 PMCID: PMC7734804 DOI: 10.3389/fcimb.2020.588168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
An estimated 3.9 billion individuals in 128 nations (about 40% of global population) are at risk of acquiring dengue virus infection. About 390 million cases of dengue are reported each year with higher prevalence in the developing world. A recent modeling-based report suggested that half of the population across the globe is at risk of dengue virus infection. In any given dengue outbreak, a percentage of infected population develops severe clinical manifestations, and this remains one of the “unsolved conundrums in dengue pathogenesis”. Although, host immunity and virus serotypes are known to modulate the infection, there are still certain underlying factors that play important roles in modulating dengue pathogenesis. Advanced genomics-based technologies have led to identification of regulatory roles of non-coding RNAs. Accumulating evidence strongly suggests that viruses and their hosts employ non-coding RNAs to modulate the outcome of infection in their own favor. The foremost ones seem to be the cellular microRNAs (miRNAs). Being the post-transcriptional regulators, miRNAs can be regarded as direct switches capable of turning “on” or “off” the viral replication process. Recently, role of long non-coding RNAs (lncRNAs) in modulating viral infections via interferon dependent or independent signaling has been recognized. Hence, we attempt to identify the “under-dog”, the non-coding RNA regulators of dengue virus infection. Such essential knowledge will enhance the understanding of dengue virus infection in holistic manner, by exposing the specific molecular targets for development of novel prophylactic, therapeutic or diagnostic strategies.
Collapse
Affiliation(s)
- Roopali Rajput
- Department of Microbiology (Virology Unit), Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India.,Department of Molecular Medicine, National Institute of Tuberculosis and Respiratory Diseases, New Delhi, India
| | - Jitender Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Mahima T Nair
- Department of Zoology, Hansraj College, University of Delhi, Delhi, India
| | - Madhu Khanna
- Department of Microbiology (Virology Unit), Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Pooja Arora
- Department of Zoology, Hansraj College, University of Delhi, Delhi, India
| | - Vikas Sood
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
30
|
King CA, Wegman AD, Endy TP. Mobilization and Activation of the Innate Immune Response to Dengue Virus. Front Cell Infect Microbiol 2020; 10:574417. [PMID: 33224897 PMCID: PMC7670994 DOI: 10.3389/fcimb.2020.574417] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022] Open
Abstract
Dengue virus is an important human pathogen, infecting an estimated 400 million individuals per year and causing symptomatic disease in a subset of approximately 100 million. Much of the effort to date describing the host response to dengue has focused on the adaptive immune response, in part because of the well-established roles of antibody-dependent enhancement and T cell original sin as drivers of severe dengue upon heterotypic secondary infection. However, the innate immune system is a crucial factor in the host response to dengue, as it both governs the fate and vigor of the adaptive immune response, and mediates the acute inflammatory response in tissues. In this review, we discuss the innate inflammatory response to dengue infection, focusing on the role of evolutionarily conserved innate immune cells, their effector functions, and clinical course.
Collapse
Affiliation(s)
- Christine A. King
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | | | | |
Collapse
|
31
|
Baker S, Blohmke CJ, Maes M, Johnston PI, Darton TC. The Current Status of Enteric Fever Diagnostics and Implications for Disease Control. Clin Infect Dis 2020; 71:S64-S70. [PMID: 32725220 PMCID: PMC7388712 DOI: 10.1093/cid/ciaa503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Enteric (typhoid) fever remains a problem in low- and middle-income countries that lack the infrastructure to maintain sanitation and where inadequate diagnostic methods have restricted our ability to identify and control the disease more effectively. As we move into a period of potential disease elimination through the introduction of typhoid conjugate vaccine (TCV), we again need to reconsider the role of typhoid diagnostics in how they can aid in facilitating disease control. Recent technological advances, including serology, transcriptomics, and metabolomics, have provided new insights into how we can detect signatures of invasive Salmonella organisms interacting with the host during infection. Many of these new techniques exhibit potential that could be further explored with the aim of creating a new enteric fever diagnostic to work in conjunction with TCV. We need a sustained effort within the enteric fever field to accelerate, validate, and ultimately introduce 1 (or more) of these methods to facilitate the disease control initiative. The window of opportunity is still open, but we need to recognize the need for communication with other research areas and commercial organizations to assist in the progression of these diagnostic approaches. The elimination of enteric fever is now becoming a real possibility, but new diagnostics need to be part of the equation and factored into future calculations for disease control.
Collapse
Affiliation(s)
- Stephen Baker
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Mailis Maes
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Peter I Johnston
- Florey Institute for Host-Pathogen Interactions, Department for Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Thomas C Darton
- Florey Institute for Host-Pathogen Interactions, Department for Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
32
|
Risk factors and biomarkers of severe dengue. Curr Opin Virol 2020; 43:1-8. [PMID: 32688269 DOI: 10.1016/j.coviro.2020.06.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022]
Abstract
Dengue virus infects several million people each year. Although usually a self-limiting disease, some patients can develop life-threatening severe complications, characterized by plasma leakage, hemorrhaging, and shock. The signs and symptoms of severe disease usually arise late in the disease course when patients are recovering and fever has subsided, making it difficult to predict. Efforts are underway to identify risk factors and biomarkers that can accurately predict disease severity in the acute febrile phase of the disease, facilitating early intervention and treatment strategies for those at greatest risk. In this review we discuss recent advancements in identifying risk factors and biomarkers for the prognosis of severe dengue.
Collapse
|
33
|
Axelrod T, Eltzov E, Marks RS. Capture-Layer Lateral Flow Immunoassay: A New Platform Validated in the Detection and Quantification of Dengue NS1. ACS OMEGA 2020; 5:10433-10440. [PMID: 32426600 PMCID: PMC7226885 DOI: 10.1021/acsomega.0c00367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 03/31/2020] [Indexed: 05/07/2023]
Abstract
The lateral flow immunoassay (LFIA) is the most successful point-of-care testing (POCT) method to date. In the case of clinical biomarkers that require quantification, it remains a challenge to quantitate those biomarkers using the lateral flow immunoassay remains a challenge due to the cost of the reader and possibly the type of marker used. In the present work, a new concept of a platform LFIA device configuration is proposed in which different, aligned membrane components, some already existing in the classical lateral flow immunoassay, and the others created with special new functions in the present device. As the sample containing the target analyte passes through the aforementioned membranes, the target analyte will initially interact with a target-specific antibody-conjugated to horseradish peroxidase (HRP). Thereafter, the newly formed immunocomplex will diffuse through a proprietary capture membrane (that ensures that the nontarget-bound antibodies do not continue further and thus remain "captured" to that specific area). This is done by having the target molecules (or components thereof) immobilized onto the said capture layer. The target-bound immunocomplexes will then be allowed by the system configuration to continue further to the last layer, where the signal will be generated and quantified. Thus, in the absence of the target analyte in the sample, the free antibodies will be filtered at the capture layer by preimmobilized analyte molecules, thus preventing a false positive signal to occur. We validated the concept in the detection of dengue NS1 protein in view of making a triage test. The sample containing NS1 will first meet HRP-conjugated NS1-specific antibodies and become attached, thus producing an NS1-specific antibody-HRP immunocomplex. The sample then flows through the blocking layer, where the immunocomplex is unchallenged and thus allowed to reach the last "absorbent" pad, incorporating the substrate for the HRP marker. In the case of a positive test, a signal is generated, that is proportional to the amount of immunocomplexes (and therefore the NS1 concentration), and then analyzed and measured at the absorbent pad. Any unbound anti-NS1 antibody will be stopped at the blocking matrix by preimmobilized NS1, so there will be no false positive. As this study is the initial study of a novel configuration, much of the work comprised of optimization steps, such as determining the required NS1 membrane-immobilization concentration and the required target-specific capture antibody concentration. Our immunoassay was tested with spiked buffer and serum samples to mimic the clinical conditions, with a range of NS1 concentrations, and was found, at this time, to be fivefold more sensitive than a gold standard enzyme-linked immunosorbent assay (ELISA) test (5 ng mL-1) performed in our laboratory. This method shows another form of LFIA that has the potential to be quantitative (at least semiquantitative), albeit not solving the reader cost; however, unlike the regular LFIA, we do not use nanobeads but instead enzymes, allowing, in theory, greater sensitivity, while retaining the one-step procedure. The test is accurate and has low production costs.
Collapse
Affiliation(s)
- Tim Axelrod
- Department
of Biotechnology Engineering, Faculty of Engineering Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Evgeni Eltzov
- Department
of Postharvest Science, Institute of Postharvest and Food Sciences, The Volcani Center, Agricultural Research Organization, Bet Dagan 50250, Israel
| | - Robert S. Marks
- Department
of Biotechnology Engineering, Faculty of Engineering Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- National
Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The
Ilse Katz Center for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
34
|
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: 2.0] [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
|
35
|
Grifoni A, Tian Y, Sette A, Weiskopf D. Transcriptomic immune profiles of human flavivirus-specific T-cell responses. Immunology 2020; 160:3-9. [PMID: 31778581 DOI: 10.1111/imm.13161] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/06/2019] [Accepted: 11/23/2019] [Indexed: 12/14/2022] Open
Abstract
The Flavivirus genus of viruses includes dengue (DENV), Zika (ZIKV), yellow fever (YFV), Japanese encephalitis (JEV), and West Nile (WNV) viruses. Infections with these species combined are prevalent in tropical and sub-tropical areas, affecting millions of people and ranging from asymptomatic to severe forms of the disease. They therefore pose a serious threat to global public health. Several studies imply a role for T cells in the protection but also pathogenesis against the different flavivirus species. Identifying flavivirus-specific T-cell immune profiles and determining how pre-exposure of one species might affect the immune response against subsequent infections from other species is important to further define the role of T cells in the immune response against infection. Understanding the immune profiles of the flavivirus-specific T-cell response in natural infection is important to understand the T-cell response in the context of vaccination. In this review, we summarize the current knowledge on human immune profiles of flavivirus-specific T-cell reactivity, comparing natural infection with the acute form of the disease and vaccination in different flavivirus infections.
Collapse
Affiliation(s)
- Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Yuan Tian
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| |
Collapse
|
36
|
Blohmke CJ, Muller J, Gibani MM, Dobinson H, Shrestha S, Perinparajah S, Jin C, Hughes H, Blackwell L, Dongol S, Karkey A, Schreiber F, Pickard D, Basnyat B, Dougan G, Baker S, Pollard AJ, Darton TC. Diagnostic host gene signature for distinguishing enteric fever from other febrile diseases. EMBO Mol Med 2019; 11:e10431. [PMID: 31468702 PMCID: PMC6783646 DOI: 10.15252/emmm.201910431] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/30/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022] Open
Abstract
Misdiagnosis of enteric fever is a major global health problem, resulting in patient mismanagement, antimicrobial misuse and inaccurate disease burden estimates. Applying a machine learning algorithm to host gene expression profiles, we identified a diagnostic signature, which could distinguish culture-confirmed enteric fever cases from other febrile illnesses (area under receiver operating characteristic curve > 95%). Applying this signature to a culture-negative suspected enteric fever cohort in Nepal identified a further 12.6% as likely true cases. Our analysis highlights the power of data-driven approaches to identify host response patterns for the diagnosis of febrile illnesses. Expression signatures were validated using qPCR, highlighting their utility as PCR-based diagnostics for use in endemic settings.
Collapse
Affiliation(s)
- Christoph J Blohmke
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | | | - Malick M Gibani
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Hazel Dobinson
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Sonu Shrestha
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Soumya Perinparajah
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Celina Jin
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Harri Hughes
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Luke Blackwell
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Sabina Dongol
- Patan Academy of Healthy SciencesOxford University Clinical Research UnitKathmanduNepal
| | - Abhilasha Karkey
- Patan Academy of Healthy SciencesOxford University Clinical Research UnitKathmanduNepal
| | | | - Derek Pickard
- Infection Genomics ProgramThe Wellcome Trust Sanger InstituteHinxtonUK
| | - Buddha Basnyat
- Patan Academy of Healthy SciencesOxford University Clinical Research UnitKathmanduNepal
| | - Gordon Dougan
- Infection Genomics ProgramThe Wellcome Trust Sanger InstituteHinxtonUK
| | - Stephen Baker
- The Hospital for Tropical DiseasesWellcome Trust Major Overseas ProgrammeOxford University Clinical Research UnitHo Chi Minh CityVietnam
| | - Andrew J Pollard
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
| | - Thomas C Darton
- Department of PaediatricsCentre for Clinical Vaccinology and Tropical MedicineOxford Vaccine GroupOxfordUK
- Oxford National Institute of Health Research Biomedical CentreUniversity of OxfordOxfordUK
- The Hospital for Tropical DiseasesWellcome Trust Major Overseas ProgrammeOxford University Clinical Research UnitHo Chi Minh CityVietnam
- Department of Infection, Immunity and Cardiovascular DiseaseUniversity of SheffieldSheffieldUK
| |
Collapse
|
37
|
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
|
38
|
Balakrishna Pillai A, Cherupanakkal C, Immanuel J, Saravanan E, Eswar Kumar V, A A, Kadhiravan T, Rajendiran S. Expression Pattern of Selected Toll-like Receptors (TLR's) in the PBMC's of Severe and Non-severe Dengue Cases. Immunol Invest 2019; 49:443-452. [PMID: 31475595 DOI: 10.1080/08820139.2019.1653908] [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: 02/08/2023]
Abstract
Objective: The role of TLR's in the pathogenesis of dengue is not explored well. Differential expression of TLR2 and TLR4 was reported in dengue cases. In the present study in order to understand the expression pattern of various TLR's, including TLR2, TLR3, TLR4 and TLR9, mRNA levels were determined in various dengue study groups compared to control groups, at the time of admission and around defervescence using quantitative real-time PCR (RT-PCR).Methods: A total of 88 dengue cases with 32 severe and 56 non-severe cases were involved in the study. Gene expression pattern of the study groups was compared with 31 other febrile illness (OFI) cases and 63 healthy controls. Transcript levels of the target genes were estimated from the peripheral blood mononuclear cells (PBMC) samples collected from cases and controls using quantitative real-time PCR.Results: We have noted a significant alteration in the levels of all TLR's in dengue and OFI cases compared to healthy controls at the time of admission. Interestingly we have noted a significant alteration in the levels of TLR9 in severe and non-severe cases during defervescence. The same was not detected in the OFI group.Conclusion: The present study found a change in TLR's during dengue infection. This suggests us to explore the TLR's as therapeutic candidate for anti-dengue virus strategies. However, in order to ascertain the involvement of TLR's in the disease pathology and its role as biomarkers for prognosis, a complete dynamics of TLR's expression needs to be studied.
Collapse
Affiliation(s)
- Agieshkumar Balakrishna Pillai
- Central Inter-Disciplinary Research Facility (CIDRF), Sri Balaji Vidyapeeth (SBV) Deemed To Be University, Puducherry, India
| | - Cleetus Cherupanakkal
- Department of Biochemistry, Believers Church Medical College Hospital, Thiruvalla, Kerala, India
| | - Jeffrey Immanuel
- Central Inter-Disciplinary Research Facility (CIDRF), Sri Balaji Vidyapeeth (SBV) Deemed To Be University, Puducherry, India
| | - Elanthiraiyan Saravanan
- Central Inter-Disciplinary Research Facility (CIDRF), Sri Balaji Vidyapeeth (SBV) Deemed To Be University, Puducherry, India
| | - Vignewswari Eswar Kumar
- Central Inter-Disciplinary Research Facility (CIDRF), Sri Balaji Vidyapeeth (SBV) Deemed To Be University, Puducherry, India
| | - Akshayavardhini A
- Central Inter-Disciplinary Research Facility (CIDRF), Sri Balaji Vidyapeeth (SBV) Deemed To Be University, Puducherry, India
| | - Tamilarasu Kadhiravan
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Soundravally Rajendiran
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
39
|
Robinson M, Sweeney TE, Barouch-Bentov R, Sahoo MK, Kalesinskas L, Vallania F, Sanz AM, Ortiz-Lasso E, Albornoz LL, Rosso F, Montoya JG, Pinsky BA, Khatri P, Einav S. A 20-Gene Set Predictive of Progression to Severe Dengue. Cell Rep 2019; 26:1104-1111.e4. [PMID: 30699342 PMCID: PMC6352713 DOI: 10.1016/j.celrep.2019.01.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/01/2018] [Accepted: 01/09/2019] [Indexed: 12/19/2022] Open
Abstract
There is a need to identify biomarkers predictive of severe dengue. Single-cohort transcriptomics has not yielded generalizable results or parsimonious, predictive gene sets. We analyzed blood samples of dengue patients from seven gene expression datasets (446 samples, five countries) using an integrated multi-cohort analysis framework and identified a 20-gene set that predicts progression to severe dengue. We validated the predictive power of this 20-gene set in three retrospective dengue datasets (84 samples, three countries) and a prospective Colombia cohort (34 patients), with an area under the receiver operating characteristic curve of 0.89, 100% sensitivity, and 76% specificity. The 20-gene dengue severity scores declined during the disease course, suggesting an infection-triggered host response. This 20-gene set is strongly associated with the progression to severe dengue and represents a predictive signature, generalizable across ages, host genetic factors, and virus strains, with potential implications for the development of a host response-based dengue prognostic assay.
Collapse
Affiliation(s)
- Makeda Robinson
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Timothy E Sweeney
- Institute for Immunity, Transplantation, and Infection, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University, Stanford, CA, USA
| | - Rina Barouch-Bentov
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Malaya Kumar Sahoo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Larry Kalesinskas
- Institute for Immunity, Transplantation, and Infection, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University, Stanford, CA, USA
| | - Francesco Vallania
- Institute for Immunity, Transplantation, and Infection, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University, Stanford, CA, USA
| | - Ana Maria Sanz
- Clinical Research Center, Fundación Valle del Lili, Cali, Colombia
| | - Eliana Ortiz-Lasso
- Pathology and Laboratory Department, Fundación Valle del Lili, Cali, Colombia
| | | | - Fernando Rosso
- Clinical Research Center, Fundación Valle del Lili, Cali, Colombia; Department of Internal Medicine, Division of Infectious Diseases, Fundación Valle del Lili, Cali, Colombia
| | - Jose G Montoya
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Benjamin A Pinsky
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation, and Infection, Department of Medicine, Stanford University, Stanford, CA, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University, Stanford, CA, USA.
| | - Shirit Einav
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
40
|
Opasawatchai A, Amornsupawat P, Jiravejchakul N, Chan-In W, Spoerk NJ, Manopwisedjaroen K, Singhasivanon P, Yingtaweesak T, Suraamornkul S, Mongkolsapaya J, Sakuntabhai A, Matangkasombut P, Loison F. Neutrophil Activation and Early Features of NET Formation Are Associated With Dengue Virus Infection in Human. Front Immunol 2019; 9:3007. [PMID: 30687301 PMCID: PMC6336714 DOI: 10.3389/fimmu.2018.03007] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/05/2018] [Indexed: 12/26/2022] Open
Abstract
The involvement of the immune system in the protection and pathology of natural dengue virus (DENV) has been extensively studied. However, despite studies that have referred to activation of neutrophils in DENV infections, the exact roles of neutrophils remain elusive. Here, we explored the phenotypic and functional responses of neutrophils in a cohort of adult dengue patients. Results indicated that during an acute DENV infection, neutrophils up-regulate CD66b expression, and produce a more robust respiratory response as compared with that in convalescent or healthy individuals; this confirmed in vivo neutrophil activation during DENV infection. Spontaneous decondensation of nuclei, an early event of neutrophil extracellular trap (NET) formation, was also markedly increased in cells isolated from DENV-infected patients during the acute phase of the infection. In vitro incubation of NETs with DENV-2 virus significantly decreased DENV infectivity. Interestingly, increased levels of NET components were found in the serum of patients with more severe disease form-dengue hemorrhagic fever (DHF), but not uncomplicated dengue fever, during the acute phase of the infection. Levels of pro-inflammatory cytokines IL-8 and TNFα were also increased in DHF patients as compared with those in healthy and DF subjects. This suggested that NETs may play dual roles during DENV infection. The increased ability for NET formation during acute DENV infection appeared to be independent of PAD4-mediated histone H3 hyper-citrullination. Our study suggests that neutrophils are involved in immunological responses to DENV infection.
Collapse
Affiliation(s)
- Anunya Opasawatchai
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Faculty of Dentistry, Mahidol University, Bangkok, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Panicha Amornsupawat
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Wilawan Chan-In
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Nicholas J Spoerk
- Department of Bacteriology, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Pratap Singhasivanon
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | | | - Juthathip Mongkolsapaya
- Department of Medicine, Imperial College London, London, United Kingdom.,Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Anavaj Sakuntabhai
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France.,Centre National de la Recherche Scientifique (CNRS), URA3012, Paris, France
| | - Ponpan Matangkasombut
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Fabien Loison
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
41
|
Kumar VE, Cherupanakkal C, Catherine M, Kadhiravan T, Parameswaran N, Rajendiran S, Pillai AB. Endogenous gene selection for relative quantification PCR and IL6 transcript levels in the PBMC's of severe and non-severe dengue cases. BMC Res Notes 2018; 11:550. [PMID: 30071880 PMCID: PMC6071388 DOI: 10.1186/s13104-018-3620-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023] Open
Abstract
Objectives Dengue viral infection ranges from dengue fever to dengue haemorrhagic fever and lethal dengue shock syndrome. Currently no means are available to monitor the progression of disease. Real time PCR based gene expression analyses are used to find potential molecular markers for effective prediction of dengue clinical outcome. The accuracy of qPCR analysis is strongly dependent on transcript normalization using stably expressed endogenous genes, which if selected imprecisely can lead to misinterpreted results. We aimed to determine the best fit for endogenous gene among six genes namely COX, ACTB, GAPDH, HMBS, HPRT and B2M for dengue viral infection cases. Gene stability was inferred from qPCR data by normalizing with two algorithms geNorm and Normfinder and the rankings generated were validated by gene expression analysis against target gene IL-6. Results Both the algorithms showed ACTB, HPRT, GAPDH as most stable genes. Normalizing with the stable genes revealed a significant fold change (p < .05) in IL-6 levels of .32, .52, .69, and .62 in non-dengue febrile illness, non severe, severe and All Dengue groups respectively compared to healthy controls. based on our study, we suggest ACTB with HPRT/GAPDH combination for normalization in qPCR for precise quantification of transcripts in dengue infected studies. Electronic supplementary material The online version of this article (10.1186/s13104-018-3620-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vigneshwari Easwar Kumar
- Central Inter-Disciplinary Research Facility, Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607402, India
| | - Cleetus Cherupanakkal
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Minna Catherine
- Central Inter-Disciplinary Research Facility, Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607402, India
| | - Tamilarasu Kadhiravan
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Narayanan Parameswaran
- Department of Pediatrics, Jawaharlal Institute of, Postgraduate Medical Education and Research, Puducherry, India
| | - Soundravally Rajendiran
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India.
| | - Agieshkumar Balakrishna Pillai
- Central Inter-Disciplinary Research Facility, Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607402, India.
| |
Collapse
|
42
|
Nikolayeva I, Bost P, Casademont I, Duong V, Koeth F, Prot M, Czerwinska U, Ly S, Bleakley K, Cantaert T, Dussart P, Buchy P, Simon-Lorière E, Sakuntabhai A, Schwikowski B. A Blood RNA Signature Detecting Severe Disease in Young Dengue Patients at Hospital Arrival. J Infect Dis 2018; 217:1690-1698. [PMID: 29490079 PMCID: PMC5946912 DOI: 10.1093/infdis/jiy086] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/13/2018] [Indexed: 12/23/2022] Open
Abstract
Background Early detection of severe dengue can improve patient care and survival. To date, no reliable single-gene biomarker exists. We hypothesized that robust multigene signatures exist. Methods We performed a prospective study on Cambodian dengue patients aged 4 to 22 years. Peripheral blood mononuclear cells (PBMCs) were obtained at hospital admission. We analyzed 42 transcriptomic profiles of patients with secondary dengue infected with dengue serotype 1. Our novel signature discovery approach controls the number of included genes and captures nonlinear relationships between transcript concentrations and severity. We evaluated the signature on secondary cases infected with different serotypes using 2 datasets: 22 PBMC samples from additional patients in our cohort and 32 whole blood samples from an independent cohort. Results We identified an 18-gene signature for detecting severe dengue in patients with secondary infection upon hospital admission with a sensitivity of 0.93 (95% confidence interval [CI], .82-.98), specificity of 0.67 (95% CI, .53-.80), and area under the receiver operating characteristic curve (AUC) of 0.86 (95% CI, .75-.97). At validation, the signature had empirical AUCs of 0.85 (95% CI, .69-1.00) and 0.83 (95% CI, .68-.98) for the PBMCs and whole blood datasets, respectively. Conclusions The signature could detect severe dengue in secondary-infected patients upon hospital admission. Its genes offer new insights into the pathogenesis of severe dengue.
Collapse
Affiliation(s)
- Iryna Nikolayeva
- Systems Biology Lab, Center for Bioinformatics, Biostatistics, and Integrative Biology (C3BI), USR 3756 - Institut Pasteur and CNRS
| | - Pierre Bost
- Systems Biology Lab, Center for Bioinformatics, Biostatistics, and Integrative Biology (C3BI), USR 3756 - Institut Pasteur and CNRS.,Graduate School of Life Sciences ED515, Sorbonne Universités UPMC Paris VI
| | - Isabelle Casademont
- Unité de Génétique fonctionnelle des maladies infectieuses, Institut Pasteur, Paris, France.,CNRS UMR2000: Génomique évolutive, modélisation et santé, Institut Pasteur, Paris, France
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Fanny Koeth
- Unité de Génétique fonctionnelle des maladies infectieuses, Institut Pasteur, Paris, France.,CNRS UMR2000: Génomique évolutive, modélisation et santé, Institut Pasteur, Paris, France
| | - Matthieu Prot
- Unité de Génétique fonctionnelle des maladies infectieuses, Institut Pasteur, Paris, France.,CNRS UMR2000: Génomique évolutive, modélisation et santé, Institut Pasteur, Paris, France
| | | | - Sowath Ly
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Kevin Bleakley
- INRIA Saclay, Palaiseau.,Département de Mathématiques d'Orsay, Orsay, France
| | - Tineke Cantaert
- Immunology Group, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Philippe Buchy
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.,GSK vaccines R&D, Singapore
| | - Etienne Simon-Lorière
- Unité de Génétique fonctionnelle des maladies infectieuses, Institut Pasteur, Paris, France.,CNRS UMR2000: Génomique évolutive, modélisation et santé, Institut Pasteur, Paris, France
| | - Anavaj Sakuntabhai
- Unité de Génétique fonctionnelle des maladies infectieuses, Institut Pasteur, Paris, France.,CNRS UMR2000: Génomique évolutive, modélisation et santé, Institut Pasteur, Paris, France
| | - Benno Schwikowski
- Systems Biology Lab, Center for Bioinformatics, Biostatistics, and Integrative Biology (C3BI), USR 3756 - Institut Pasteur and CNRS
| |
Collapse
|
43
|
Oliveira M, Lert-itthiporn W, Cavadas B, Fernandes V, Chuansumrit A, Anunciação O, Casademont I, Koeth F, Penova M, Tangnararatchakit K, Khor CC, Paul R, Malasit P, Matsuda F, Simon-Lorière E, Suriyaphol P, Pereira L, Sakuntabhai A. Joint ancestry and association test indicate two distinct pathogenic pathways involved in classical dengue fever and dengue shock syndrome. PLoS Negl Trop Dis 2018; 12:e0006202. [PMID: 29447178 PMCID: PMC5813895 DOI: 10.1371/journal.pntd.0006202] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/02/2018] [Indexed: 11/18/2022] Open
Abstract
Ethnic diversity has been long considered as one of the factors explaining why the severe forms of dengue are more prevalent in Southeast Asia than anywhere else. Here we take advantage of the admixed profile of Southeast Asians to perform coupled association-admixture analyses in Thai cohorts. For dengue shock syndrome (DSS), the significant haplotypes are located in genes coding for phospholipase C members (PLCB4 added to previously reported PLCE1), related to inflammation of blood vessels. For dengue fever (DF), we found evidence of significant association with CHST10, AHRR, PPP2R5E and GRIP1 genes, which participate in the xenobiotic metabolism signaling pathway. We conducted functional analyses for PPP2R5E, revealing by immunofluorescence imaging that the coded protein co-localizes with both DENV1 and DENV2 NS5 proteins. Interestingly, only DENV2-NS5 migrated to the nucleus, and a deletion of the predicted top-linking motif in NS5 abolished the nuclear transfer. These observations support the existence of differences between serotypes in their cellular dynamics, which may contribute to differential infection outcome risk. The contribution of the identified genes to the genetic risk render Southeast and Northeast Asian populations more susceptible to both phenotypes, while African populations are best protected against DSS and intermediately protected against DF, and Europeans the best protected against DF but the most susceptible against DSS.
Collapse
Affiliation(s)
- Marisa Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France
| | - Worachart Lert-itthiporn
- Bioinformatics and Data Management for Research, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Bruno Cavadas
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Verónica Fernandes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
| | - Ampaiwan Chuansumrit
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Orlando Anunciação
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
| | - Isabelle Casademont
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France
- Pasteur Kyoto International Joint Research Unit for Integrative Vaccinomics, Kyoto, Japan
| | - Fanny Koeth
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France
- Pasteur Kyoto International Joint Research Unit for Integrative Vaccinomics, Kyoto, Japan
| | - Marina Penova
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France
- Pasteur Kyoto International Joint Research Unit for Integrative Vaccinomics, Kyoto, Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kanchana Tangnararatchakit
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chiea Chuen Khor
- Genome Institute of Singapore, A-STAR, Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
| | - Richard Paul
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France
- Pasteur Kyoto International Joint Research Unit for Integrative Vaccinomics, Kyoto, Japan
- CNRS, Unité de Recherche Associée 3012, Paris, France
| | - Prida Malasit
- Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol University, Bangkok, Thailand
- Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Fumihiko Matsuda
- Pasteur Kyoto International Joint Research Unit for Integrative Vaccinomics, Kyoto, Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Etienne Simon-Lorière
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France
- Pasteur Kyoto International Joint Research Unit for Integrative Vaccinomics, Kyoto, Japan
- CNRS, Unité de Recherche Associée 3012, Paris, France
| | - Prapat Suriyaphol
- Bioinformatics and Data Management for Research, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Luisa Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
- * E-mail: (LP); (AS)
| | - Anavaj Sakuntabhai
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, Paris, France
- Pasteur Kyoto International Joint Research Unit for Integrative Vaccinomics, Kyoto, Japan
- CNRS, Unité de Recherche Associée 3012, Paris, France
- * E-mail: (LP); (AS)
| |
Collapse
|
44
|
Xavier-Carvalho C, Cardoso CC, de Souza Kehdy F, Pacheco AG, Moraes MO. Host genetics and dengue fever. INFECTION GENETICS AND EVOLUTION 2017; 56:99-110. [PMID: 29133029 DOI: 10.1016/j.meegid.2017.11.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022]
Abstract
Dengue is a major worldwide problem in tropical and subtropical areas; it is caused by four different viral serotypes, and it can manifest as asymptomatic, mild, or severe. Many factors interact to determine the severity of the disease, including the genetic profile of the infected patient. However, the mechanisms that lead to severe disease and eventually death have not been determined, and a great challenge is the early identification of patients who are more likely to progress to a worse health condition. Studies performed in regions with cyclic outbreaks such as Cuba, Brazil, and Colombia have demonstrated that African ancestry confers protection against severe dengue. Highlighting the host genetics as an important factor in infectious diseases, a large number of association studies between genetic polymorphisms and dengue outcomes have been published in the last two decades. The most widely used approach involves case-control studies with candidate genes, such as the HLA locus and genes for receptors, cytokines, and other immune mediators. Additionally, a Genome-Wide Association Study (GWAS) identified SNPs associated with African ethnicity that had not previously been identified in case-control studies. Despite the increasing number of publications in America, Africa, and Asia, the results are quite controversial, and a meta-analysis is needed to assess the consensus among the studies. SNPs in the MICB, TNF, CD209, FcγRIIA, TPSAB1, CLEC5A, IL10 and PLCE1 genes are associated with the risk or protection of severe dengue, and the findings have been replicated in different populations. A thorough understanding of the viral, human genetic, and immunological mechanisms of dengue and how they interact is essential for effectively preventing dengue, but also managing and treating patients.
Collapse
Affiliation(s)
| | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | | | | | - Milton Ozório Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
45
|
Yacoub S, Lam PK, Huynh TT, Nguyen Ho HH, Dong Thi HT, Van NT, Lien LT, Ha QNT, Le DHT, Mongkolspaya J, Culshaw A, Yeo TW, Wertheim H, Simmons C, Screaton G, Wills B. Endothelial Nitric Oxide Pathways in the Pathophysiology of Dengue: A Prospective Observational Study. Clin Infect Dis 2017; 65:1453-1461. [PMID: 28673038 PMCID: PMC5850435 DOI: 10.1093/cid/cix567] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/20/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Dengue can cause increased vascular permeability that may lead to hypovolemic shock. Endothelial dysfunction may underlie this; however, the association of endothelial nitric oxide (NO) pathways with disease severity is unknown. METHODS We performed a prospective observational study in 2 Vietnamese hospitals, assessing patients presenting early (<72 hours of fever) and patients hospitalized with warning signs or severe dengue. The reactive hyperemic index (RHI), which measures endothelium-dependent vasodilation and is a surrogate marker of endothelial function and NO bioavailability, was evaluated using peripheral artery tonometry (EndoPAT), and plasma levels of l-arginine, arginase-1, and asymmetric dimethylarginine were measured at serial time-points. The main outcome of interest was plasma leakage severity. RESULTS Three hundred fourteen patients were enrolled; median age of the participants was 21(interquartile range, 13-30) years. No difference was found in the endothelial parameters between dengue and other febrile illness. Considering dengue patients, the RHI was significantly lower for patients with severe plasma leakage compared to those with no leakage (1.46 vs 2.00; P < .001), over acute time-points, apparent already in the early febrile phase (1.29 vs 1.75; P = .012). RHI correlated negatively with arginase-1 and positively with l-arginine (P = .001). CONCLUSIONS Endothelial dysfunction/NO bioavailability is associated with worse plasma leakage, occurs early in dengue illness and correlates with hypoargininemia and high arginase-1 levels.
Collapse
Affiliation(s)
- Sophie Yacoub
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
- Department of Medicine, Imperial College London, United Kingdom
| | - Phung Khanh Lam
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
| | | | | | - Hoai Tam Dong Thi
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
- Hospital for Tropical Diseases, Ho Chi Minh City, and
| | - Nguyen Thu Van
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
| | - Le Thi Lien
- National Hospital for Tropical Diseases, Hanoi, Vietnam
| | - Quyen Nguyen Than Ha
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
| | - Duyen Huynh Thi Le
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
| | | | - Abigail Culshaw
- Department of Medicine, Imperial College London, United Kingdom
| | - Tsin Wen Yeo
- Menzies School of Health Research, Darwin, Australia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Heiman Wertheim
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
- Nuffield Department of Medicine, University of Oxford, United Kingdom; and
| | - Cameron Simmons
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
- Department of Microbiology and Immunology, University of Melbourne, Australia
| | - Gavin Screaton
- Department of Medicine, Imperial College London, United Kingdom
| | - Bridget Wills
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Hanoi and Ho Chi Minh City, Vietnam
- Nuffield Department of Medicine, University of Oxford, United Kingdom; and
| |
Collapse
|
46
|
Schwameis M, Buchtele N, Wadowski PP, Schoergenhofer C, Jilma B. Chikungunya vaccines in development. Hum Vaccin Immunother 2017; 12:716-31. [PMID: 26554522 PMCID: PMC4964651 DOI: 10.1080/21645515.2015.1101197] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chikungunya virus has become a global health threat, spreading to the industrial world of Europe and the Americas; no treatment or prophylactic vaccine is available. Since the late 1960s much effort has been put into the development of a vaccine, and several heterogeneous strategies have already been explored. Only two candidates have recently qualified to enter clinical phase II trials, a chikungunya virus-like particle-based vaccine and a recombinant live attenuated measles virus-vectored vaccine. This review focuses on the current status of vaccine development against chikungunya virus in humans and discusses the diversity of immunization strategies, results of recent human trials and promising vaccine candidates.
Collapse
Affiliation(s)
- Michael Schwameis
- a Departments of Clinical Pharmacology and Internal Medicine I , Medical University of Vienna , Vienna , Austria
| | - Nina Buchtele
- a Departments of Clinical Pharmacology and Internal Medicine I , Medical University of Vienna , Vienna , Austria
| | - Patricia Pia Wadowski
- a Departments of Clinical Pharmacology and Internal Medicine I , Medical University of Vienna , Vienna , Austria
| | | | - Bernd Jilma
- a Departments of Clinical Pharmacology and Internal Medicine I , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
47
|
Transcriptional Profiling Confirms the Therapeutic Effects of Mast Cell Stabilization in a Dengue Disease Model. J Virol 2017; 91:JVI.00617-17. [PMID: 28659489 PMCID: PMC5571258 DOI: 10.1128/jvi.00617-17] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/22/2017] [Indexed: 12/28/2022] Open
Abstract
There are no approved therapeutics for the treatment of dengue disease despite the global prevalence of dengue virus (DENV) and its mosquito vectors. DENV infections can lead to vascular complications, hemorrhage, and shock due to the ability of DENV to infect a variety of immune and nonimmune cell populations. Increasingly, studies have implicated the host response as a major contributor to severe disease. Inflammatory products of various cell types, including responding T cells, mast cells (MCs), and infected monocytes, can contribute to immune pathology. In this study, we show that the host response to DENV infection in immunocompetent mice recapitulates transcriptional changes that have been described in human studies. We found that DENV infection strongly induced metabolic dysregulation, complement signaling, and inflammation. DENV also affected the immune cell content of the spleen and liver, enhancing NK, NKT, and CD8+ T cell activation. The MC-stabilizing drug ketotifen reversed many of these responses without suppressing memory T cell formation and induced additional changes in the transcriptome and immune cell composition of the spleen, consistent with reduced inflammation. This study provides a global transcriptional map of immune activation in DENV target organs of an immunocompetent host and supports the further development of targeted immunomodulatory strategies to treat DENV disease.IMPORTANCE Dengue virus (DENV), which causes febrile illness, is transmitted by mosquito vectors throughout tropical and subtropical regions of the world. Symptoms of DENV infection involve damage to blood vessels and, in rare cases, hemorrhage and shock. Currently, there are no targeted therapies to treat DENV infection, but it is thought that drugs that target the host immune response may be effective in limiting symptoms that result from excessive inflammation. In this study, we measured the host transcriptional response to infection in multiple DENV target organs using a mouse model of disease. We found that DENV infection induced metabolic dysregulation and inflammatory responses and affected the immune cell content of the spleen and liver. The use of the mast cell stabilization drug ketotifen reversed many of these responses and induced additional changes in the transcriptome and immune cell repertoire that contribute to decreased dengue disease.
Collapse
|
48
|
Banerjee A, Shukla S, Pandey AD, Goswami S, Bandyopadhyay B, Ramachandran V, Das S, Malhotra A, Agarwal A, Adhikari S, Rahman M, Chatterjee S, Bhattacharya N, Basu N, Pandey P, Sood V, Vrati S. RNA-Seq analysis of peripheral blood mononuclear cells reveals unique transcriptional signatures associated with disease progression in dengue patients. Transl Res 2017; 186:62-78.e9. [PMID: 28683259 DOI: 10.1016/j.trsl.2017.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/06/2017] [Accepted: 06/08/2017] [Indexed: 12/21/2022]
Abstract
Patients infected with Dengue virus usually present a mild, self-limiting febrile dengue infection (DI) that occasionally leads to a potentially lethal complication, called the severe dengue (DS). The ability to identify the prognostic markers of DS could allow an improved disease intervention and management. To identify the transcriptional signatures associated with the dengue disease progression, we carried out the high-throughput sequencing of the RNA isolated from the peripheral blood mononuclear cells (PBMCs) of the dengue patients of varying severity and compared with that in the patients with other febrile illnesses (OFIs) or the healthy controls. The transcriptional signatures that discriminated the DS patients from OFI and DI patients were broadly related to the pathways involving glycine, serine, and threonine metabolisms, extracellular matrix organization, ubiquitination, and cytokines and inflammatory response. Several upregulated genes in the inflammatory process (MPO, DEFA4, ELANE, AUZ1, CTSG, OLFM4, SLC16A14, and CRISP3) that were associated with the dengue disease progression are known to facilitate leukocyte-mediated migration, and neutrophil activation and degranulation process. High activity of MPO and ELANE in the plasma samples of the follow-up and recovered dengue patients, as well as and the presence of a larger amount of cell-free dsDNA in the DS patients, suggested an association of neutrophil-mediated immunity with dengue disease progression. Careful monitoring of some of these gene transcripts, and control of the activity of proteins encoded by them, may have a great translational significance for the prognosis and management of the dengue patients.
Collapse
Affiliation(s)
- Arup Banerjee
- Vaccine and Infectious Disease Research Center (VIDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India.
| | - Shweta Shukla
- University College of Medical Sciences (UCMS) & Guru Teg Bahadur (GTB) Hospital, Delhi, Delhi, India
| | - Abhay Deep Pandey
- Vaccine and Infectious Disease Research Center (VIDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Saptamita Goswami
- Virology Unit, Department of Microbiology, Calcutta School of Tropical Medicine (STM), Kolkata, West Bengal, India
| | - Bhaswati Bandyopadhyay
- Virology Unit, Department of Microbiology, Calcutta School of Tropical Medicine (STM), Kolkata, West Bengal, India
| | | | - Shukla Das
- University College of Medical Sciences (UCMS) & Guru Teg Bahadur (GTB) Hospital, Delhi, Delhi, India
| | - Arjun Malhotra
- Vaccine and Infectious Disease Research Center (VIDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Amitesh Agarwal
- University College of Medical Sciences (UCMS) & Guru Teg Bahadur (GTB) Hospital, Delhi, Delhi, India
| | - Srima Adhikari
- Virology Unit, Department of Microbiology, Calcutta School of Tropical Medicine (STM), Kolkata, West Bengal, India
| | - Mehebubar Rahman
- Virology Unit, Department of Microbiology, Calcutta School of Tropical Medicine (STM), Kolkata, West Bengal, India
| | | | - Nemai Bhattacharya
- Virology Unit, Department of Microbiology, Calcutta School of Tropical Medicine (STM), Kolkata, West Bengal, India
| | - Nandita Basu
- Virology Unit, Department of Microbiology, Calcutta School of Tropical Medicine (STM), Kolkata, West Bengal, India
| | - Priyanka Pandey
- National Institute of Biomedical Genomics (NIBMG), Kalyani, West Bengal, India
| | - Vikas Sood
- Vaccine and Infectious Disease Research Center (VIDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Sudhanshu Vrati
- Vaccine and Infectious Disease Research Center (VIDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India; Regional Center for Biotechnology (RCB), Faridabad, Haryana, India.
| |
Collapse
|
49
|
Taguchi YH. Principal Components Analysis Based Unsupervised Feature Extraction Applied to Gene Expression Analysis of Blood from Dengue Haemorrhagic Fever Patients. Sci Rep 2017; 7:44016. [PMID: 28276456 PMCID: PMC5343617 DOI: 10.1038/srep44016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 02/02/2017] [Indexed: 12/12/2022] Open
Abstract
Dengue haemorrhagic fever (DHF) sometimes occurs after recovery from the disease caused by Dengue virus (DENV), and is often fatal. However, the mechanism of DHF has not been determined, possibly because no suitable methodologies are available to analyse this disease. Therefore, more innovative methods are required to analyse the gene expression profiles of DENV-infected patients. Principal components analysis (PCA)-based unsupervised feature extraction (FE) was applied to the gene expression profiles of DENV-infected patients, and an integrated analysis of two independent data sets identified 46 genes as critical for DHF progression. PCA using only these 46 genes rendered the two data sets highly consistent. The application of PCA to the 46 genes of an independent third data set successfully predicted the progression of DHF. A fourth in vitro data set confirmed the identification of the 46 genes. These 46 genes included interferon- and heme-biosynthesis-related genes. The former are enriched in binding sites for STAT1, STAT2, and IRF1, which are associated with DHF-promoting antibody-dependent enhancement, whereas the latter are considered to be related to the dysfunction of spliceosomes, which may mediate haemorrhage. These results are outcomes that other type of bioinformatic analysis could hardly achieve.
Collapse
Affiliation(s)
- Y-H Taguchi
- Department of Physics, Chuo University, Tokyo, 112-8551, Japan
| |
Collapse
|
50
|
Alphonse MP, Duong TT, Shumitzu C, Hoang TL, McCrindle BW, Franco A, Schurmans S, Philpott DJ, Hibberd ML, Burns J, Kuijpers TW, Yeung RSM. Inositol-Triphosphate 3-Kinase C Mediates Inflammasome Activation and Treatment Response in Kawasaki Disease. THE JOURNAL OF IMMUNOLOGY 2016; 197:3481-3489. [PMID: 27694492 DOI: 10.4049/jimmunol.1600388] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/04/2016] [Indexed: 01/05/2023]
Abstract
Kawasaki disease (KD) is a multisystem vasculitis that predominantly targets the coronary arteries in children. Phenotypic similarities between KD and recurrent fever syndromes point to the potential role of inflammasome activation in KD. Mutations in NLRP3 are associated with recurrent fever/autoinflammatory syndromes. We show that the KD-associated genetic polymorphism in inositol-triphosphate 3-kinase C (ITPKC) (rs28493229) has important functional consequences, governing ITPKC protein levels and thereby intracellular calcium, which in turn regulates NLRP3 expression and production of IL-1β and IL-18. Analysis of transcript abundance, protein levels, and cellular response profiles from matched, serial biospecimens from a cohort of genotyped KD subjects points to the critical role of ITPKC in mediating NLRP3 inflammasome activation. Treatment failure in those with the high-risk ITPKC genotype was associated with the highest basal and stimulated intracellular calcium levels and with increased cellular production of IL-1β and IL-18 and higher circulating levels of both cytokines. Mechanistic studies using Itpkc-deficient mice in a disease model support the genomic, cellular, and clinical findings in affected children. Our findings provide the mechanism behind the observed efficacy of rescue therapy with IL-1 blockade in recalcitrant KD, and we identify that regulation of calcium mobilization is fundamental to the underlying immunobiology in KD.
Collapse
Affiliation(s)
- Martin Prince Alphonse
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Cell Biology Research Program, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Trang T Duong
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Chisato Shumitzu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | | | - Brian W McCrindle
- Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Alessandra Franco
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA-Research Center, University of Liege, 4000 Liege, Belgium; and
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | - Jane Burns
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | - Taco W Kuijpers
- Department of Pediatrics, Sanquin Blood Bank, 1066 Amsterdam, the Netherlands
| | - Rae S M Yeung
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; .,Cell Biology Research Program, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada.,Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|