1
|
Cherie TJJ, Choong CSH, Abid MB, Weber MW, Yap ES, Seneviratne SL, Abeysuriya V, de Mel S. Immuno-Haematologic Aspects of Dengue Infection: Biologic Insights and Clinical Implications. Viruses 2024; 16:1090. [PMID: 39066252 PMCID: PMC11281699 DOI: 10.3390/v16071090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Dengue infection is caused by the dengue virus (DENV) and is transmitted to humans by infected female Aedes aegypti and Aedes albopictus mosquitoes. There are nearly 100 million new dengue cases yearly in more than 120 countries, with a five-fold increase in incidence over the past four decades. While many patients experience a mild illness, a subset suffer from severe disease, which can be fatal. Dysregulated immune responses are central to the pathogenesis of dengue, and haematologic manifestations are a prominent feature of severe disease. While thrombocytopaenia and coagulopathy are major causes of bleeding in severe dengue, leucocyte abnormalities are emerging as important markers of prognosis. In this review, we provide our perspective on the clinical aspects and pathophysiology of haematologic manifestations in dengue. We also discuss the key gaps in our current practice and areas to be addressed by future research.
Collapse
Affiliation(s)
- Tan Jiao Jie Cherie
- Department of Medicine, National University Health System, Singapore 119228, Singapore;
| | - Clarice Shi Hui Choong
- Department of Haematology Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore;
| | - Muhammad Bilal Abid
- Division of Haematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.B.A.); (M.W.W.)
| | - Matthew W. Weber
- Division of Haematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.B.A.); (M.W.W.)
| | - Eng Soo Yap
- Department of Laboratory Medicine, National University Health System, Singapore 119228, Singapore;
| | - Suranjith L. Seneviratne
- Institute of Immunity and Transplantation, Royal Free Hospital and University College London, London NW3 2PP, UK
- Nawaloka Hospital Research and Educational Foundation, Nawaloka Hospitals PLC, Colombo 00200, Sri Lanka
| | - Visula Abeysuriya
- Department of Immunology, Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 00300, Sri Lanka;
| | - Sanjay de Mel
- Department of Haematology Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore;
| |
Collapse
|
2
|
Vo HTM, Upasani V, Auerswald H, Lay S, Sann S, Vanderlinden A, Ken S, Sorn S, Ly S, Duong V, Dussart P, Cantaert T. Temporal patterns of functional anti-dengue antibodies in dengue infected individuals with different disease outcome or infection history. Sci Rep 2022; 12:17863. [PMID: 36284116 PMCID: PMC9596418 DOI: 10.1038/s41598-022-21722-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 09/30/2022] [Indexed: 01/20/2023] Open
Abstract
Heterotypic secondary dengue virus (DENV) infection is a risk factor for the development of severe disease. To assess the contribution of the developing polyclonal humoral immune response to the course of acute infection, we have determined anti-DENV IgG titers, neutralizing antibodies, percentages of antibodies binding to DENV-infected cells and antibody‑dependent enhancement (ADE) to the infecting serotype in DENV-infected Cambodian children (n = 58), ranging from asymptomatic dengue to severe disease. The results showed that ADE titers are highest against the infecting serotype during heterotypic secondary DENV-2 infection. Moreover, IgG titers, neutralizing antibodies and ADE titers against the infecting serotype peak at D10 and are maintained until D60 after laboratory-confirmed secondary DENV infection. Anti-DENV IgG titers and the magnitude of the functional antibody response were higher in secondary DENV-infected patients compared to primary infected patients. No differences in antibody titers, neutralizing or enhancing antibodies could be observed between asymptomatic or hospitalized patients between 6 and 8 days after laboratory-confirmed DENV-1 infection. However, at this time point, the level of IgG bound to DENV-infected cells was associated with disease severity in hospitalized patients. Taken together, our data offer insights for more comprehensive interpretation of antibody response profile to natural infection and its correlation to disease outcome.
Collapse
Affiliation(s)
- Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
- Centre for Tropical Medicine, Oxford University Clinical Research Unit, Ho Chi Minh, Vietnam
| | - Vinit Upasani
- Immunology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Axelle Vanderlinden
- Immunology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Sreymom Ken
- Virology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Sopheak Sorn
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia
- The Pasteur Network, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, The Pasteur Network, Phnom Penh, Cambodia.
| |
Collapse
|
3
|
Maheshwari D, Saini K, Singh P, Singla M, Nayak K, Aggarwal C, Chawla YM, Bajpai P, Kaur M, Gunisetty S, Eberhardt CS, Nyodu R, Moore K, Suthar MS, Medigeshi GR, Anderson E, Lodha R, Kabra SK, Ahmed R, Chandele A, Murali-Krishna K. Contrasting behavior between the three human monocyte subsets in dengue pathophysiology. iScience 2022; 25:104384. [PMID: 35620424 PMCID: PMC9127603 DOI: 10.1016/j.isci.2022.104384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/09/2022] [Accepted: 05/05/2022] [Indexed: 10/26/2022] Open
|
4
|
Li YR, Dunn ZS, Garcia G, Carmona C, Zhou Y, Lee D, Yu J, Huang J, Kim JT, Arumugaswami V, Wang P, Yang L. Development of off-the-shelf hematopoietic stem cell-engineered invariant natural killer T cells for COVID-19 therapeutic intervention. Stem Cell Res Ther 2022; 13:112. [PMID: 35313965 PMCID: PMC8935266 DOI: 10.1186/s13287-022-02787-2] [Citation(s) in RCA: 6] [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: 08/23/2021] [Accepted: 10/16/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND New COVID-19 treatments are desperately needed as case numbers continue to rise and emergent strains threaten vaccine efficacy. Cell therapy has revolutionized cancer treatment and holds much promise in combatting infectious disease, including COVID-19. Invariant natural killer T (iNKT) cells are a rare subset of T cells with potent antiviral and immunoregulatory functions and an excellent safety profile. Current iNKT cell strategies are hindered by the extremely low presence of iNKT cells, and we have developed a platform to overcome this critical limitation. METHODS We produced allogeneic HSC-engineered iNKT (AlloHSC-iNKT) cells through TCR engineering of human cord blood CD34+ hematopoietic stem cells (HSCs) and differentiation of these HSCs into iNKT cells in an Ex Vivo HSC-Derived iNKT Cell Culture. We then established in vitro SARS-CoV-2 infection assays to assess AlloHSC-iNKT cell antiviral and anti-hyperinflammation functions. Lastly, using in vitro and in vivo preclinical models, we evaluated AlloHSC-iNKT cell safety and immunogenicity for off-the-shelf application. RESULTS We reliably generated AlloHSC-iNKT cells at high-yield and of high-purity; these resulting cells closely resembled endogenous human iNKT cells in phenotypes and functionalities. In cell culture, AlloHSC-iNKT cells directly killed SARS-CoV-2 infected cells and also selectively eliminated SARS-CoV-2 infection-stimulated inflammatory monocytes. In an in vitro mixed lymphocyte reaction (MLR) assay and an NSG mouse xenograft model, AlloHSC-iNKT cells were resistant to T cell-mediated alloreaction and did not cause GvHD. CONCLUSIONS Here, we report a method to robustly produce therapeutic levels of AlloHSC-iNKT cells. Preclinical studies showed that these AlloHSC-iNKT cells closely resembled endogenous human iNKT cells, could reduce SARS-CoV-2 virus infection load and mitigate virus infection-induced hyperinflammation, and meanwhile were free of GvHD-risk and resistant to T cell-mediated allorejection. These results support the development of AlloHSC-iNKT cells as a promising off-the-shelf cell product for treating COVID-19; such a cell product has the potential to target the new emerging SARS-CoV-2 variants as well as the future new emerging viruses.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zachary Spencer Dunn
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, Los Angeles, CA, 90089, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Camille Carmona
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Derek Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jie Huang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jocelyn T Kim
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, Los Angeles, CA, 90089, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
5
|
Ajmeriya S, Kumar A, Karmakar S, Rana S, Singh H. Neutralizing Antibodies and Antibody-Dependent Enhancement in COVID-19: A Perspective. J Indian Inst Sci 2022; 102:671-687. [PMID: 35136306 PMCID: PMC8814804 DOI: 10.1007/s41745-021-00268-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Antibody-dependent enhancement (ADE) is an alternative route of viral entry in the susceptible host cell. In this process, antiviral antibodies enhance the entry access of virus in the cells via interaction with the complement or Fc receptors leading to the worsening of infection. SARS-CoV-2 variants pose a general concern for the efficacy of neutralizing antibodies that may fail to neutralize infection, raising the possibility of a more severe form of COVID-19. Data from various studies on respiratory viruses raise the speculation that antibodies elicited against SARS-CoV-2 and during COVID-19 recovery could potentially exacerbate the infection through ADE at sub-neutralizing concentrations; this may contribute to disease pathogenesis. It is, therefore, of utmost importance to study the effectiveness of the anti-SARS-CoV-2 antibodies in COVID-19-infected subjects. Theoretically, ADE remains a general concern for the efficacy of antibodies elicited during infection, most notably in convalescent plasma therapy and in response to vaccines where it could be counterproductive.
Collapse
Affiliation(s)
- Swati Ajmeriya
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Amit Kumar
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, Room no 3020, Ansari Nagar, New Delhi, 110029 India
| | - Shweta Rana
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Harpreet Singh
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| |
Collapse
|
6
|
Sun P, Jani V, Johnson A, Cheng Y, Nagabhushana N, Williams M, Morrison BJ, Defang G. T cell and memory B cell responses in tetravalent DNA, tetravalent inactivated and tetravalent live-attenuated prime-boost dengue vaccines in rhesus macaques. Vaccine 2021; 39:7510-7520. [PMID: 34823910 DOI: 10.1016/j.vaccine.2021.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 11/17/2022]
Abstract
We previously reported the efficacy of prime-boost vaccination using three tetravalent (T) dengue vaccines, DNA (TDNA), purified inactivated vaccine (TPIV), and live attenuated vaccine (TLAV). We demonstrated that the TPIV/TLAV prime-boost vaccination yielded the highest and most durable neutralizing antibodies and 100% protection to all 4 serotypes of dengue virus in rhesus macaques. This study compares gene transcription, T and B cell responses elicited by these prime-boost combinations in rhesus macaques. This study shows that the TLAV vaccine increased the expression of the innate immune genes, DDX58 and TLR7, IL1A, IL1B, TNF, CXCL8, CXCL10, IRF1, IRF7, and IFNB, more robustly as compared to TDNA and TPIV vaccines. Overall, two doses of TDNA and one dose of TLAV efficiently elicited a T cell IFNγ response to PrM/E with a comparable magnitude. Compared to TDNA vaccine, the TLAV vaccine elicited additional IFNγ response to C, NS1, NS3, and NS5. The TPIV vaccine alone produced poor IFNγ response; however, the TLAV significantly boosted its IFNγ response. The T cell response repertoire associated with TPIV/TLAV prime-boost was to both the structural C/PrM/E and NS proteins, and the T cells were multifunctional as the CD4+ T cells produced IFNγ, TNF α, and IL2 and the CD8+ cells produced TNF α and IFNγ. Opposite to the pattern of CMI, the TPIV vaccine alone elicited the highest BMem compared to the other two vaccines, which continuously remained as the highest after boosting. In summary, the TDNA and TLAV vaccines elicited a strong T cell response whereas the TPIV vaccine elicited a superior BMem. The T cell response of the TPIV vaccine was significantly boosted by the TLAV vaccine. The elevated T cell response may have provided T cell help for a sustained antibody response for TPIV/TLAV vaccines, which is required for a protective immunity against a live virus challenge.
Collapse
Affiliation(s)
- Peifang Sun
- Naval Medical Research Center, Silver Spring, MD, United States.
| | - Vihasi Jani
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Alison Johnson
- Boehringer Ingelheim Pharmaceuticals, Inc, CT, United States
| | | | - Nishith Nagabhushana
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Maya Williams
- Chemistry Division, US Naval Research Laboratory, DC, United States
| | | | - Gabriel Defang
- Naval Medical Research Center, Silver Spring, MD, United States
| |
Collapse
|
7
|
Lertjuthaporn S, Keawvichit R, Polsrila K, Sukapirom K, Chuansumrit A, Chokephaibulkit K, Ansari AA, Khowawisetsut L, Pattanapanyasat K. Kinetic Changes of Peripheral Blood Monocyte Subsets and Expression of Co-Stimulatory Molecules during Acute Dengue Virus Infection. Pathogens 2021; 10:pathogens10111458. [PMID: 34832614 PMCID: PMC8625762 DOI: 10.3390/pathogens10111458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/31/2021] [Accepted: 11/09/2021] [Indexed: 11/21/2022] Open
Abstract
Monocytes, one of the main target cells for dengue virus (DENV) infection, contribute to the resolution of viremia and to pathogenesis. We performed a longitudinal study by a detailed phenotypic comparison of classical (CD14++CD16−, non-classical (CD14+CD16++) and intermediate (CD14++CD16+) monocyte subsets in blood samples from dengue fever (DF) to the severe dengue hemorrhagic fever (DHF) and healthy individuals. Various costimulatory molecules of CD40, CD80, CD86 and inducible costimulatory ligand (ICOSL) expressed on these three monocyte subsets were also analyzed. DENV-infected patients showed an increase in the frequency of intermediate monocytes and a decrease in the classical monocytes when compared to healthy individuals. Although these differences did not correlate with disease severity, changes during the early phase of infection gradually returned to normal in the defervescence phase. Moreover, decreased frequency of classical monocytes was associated with a significant up-regulation of co-stimulatory molecules CD40, CD86 and ICOSL. Kinetics of these co-stimulatory molecule-expressing classical monocytes showed different patterns throughout the sampling times of acute DENV infection. Different distribution of monocyte subsets and their co-stimulatory molecules in the peripheral blood during acute infection might exacerbate immune responses like cytokine storms and ADE, and future studies on intracellular molecular pathways utilized by these monocyte linages are warranted.
Collapse
Affiliation(s)
- Sakaorat Lertjuthaporn
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand;
| | - Rassamon Keawvichit
- Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok 10300, Thailand;
| | - Korakot Polsrila
- Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Kasama Sukapirom
- Center of Excellence for Microparticle and Exosome in Diseases, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Ampaiwan Chuansumrit
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Kulkanya Chokephaibulkit
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Aftab A. Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Correspondence: (L.K.); (K.P.)
| | - Kovit Pattanapanyasat
- Center of Excellence for Microparticle and Exosome in Diseases, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
- Correspondence: (L.K.); (K.P.)
| |
Collapse
|
8
|
Pradeep SP, Hoovina Venkatesh P, Manchala NR, Vayal Veedu A, Basavaraju RK, Selvasundari L, Ramakrishna M, Chandrakiran Y, Krishnamurthy V, Holigi S, Thomas T, Ross CR, Dias M, Satchidanandam V. Innate Immune Cytokine Profiling and Biomarker Identification for Outcome in Dengue Patients. Front Immunol 2021; 12:677874. [PMID: 34335578 PMCID: PMC8318829 DOI: 10.3389/fimmu.2021.677874] [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: 03/08/2021] [Accepted: 06/22/2021] [Indexed: 11/21/2022] Open
Abstract
Background Early biomarkers of progression to severe dengue are urgently required to enable effective patient management and control treatment costs. Innate immune cells, which comprise the earliest responders to infection and along with the cytokines and chemokines they secrete, play a vital role in orchestrating the subsequent adaptive immune response and have been implicated in the enhancement of infection and “cytokine storm” associated with dengue severity. We investigated the early innate immune cytokine profile of dengue patients during acute phase of disease in a prospective blinded study that included subjects with acute dengue and febrile controls from four major hospitals in Bengaluru, India along with healthy controls. We used intracellular cytokine staining and flow cytometry to identify innate immune biomarkers that can predict progression to severe dengue. Results Dengue infection resulted in enhanced secretion of multiple cytokines by all queried innate immune cell subsets, dominated by TNF-α from CD56+CD3+ NKT cells, monocyte subsets, and granulocytes along with IFN-γ from CD56+CD3+ NKT cells. Of note, significantly higher proportions of TNF-α secreting granulocytes and monocyte subsets at admission were associated with mild dengue and minimal symptoms. Dengue NS1 antigenemia used as a surrogate of viral load directly correlated with proportion of cytokine-secreting innate immune cells and was significantly higher in those who went on to recover with minimal symptoms. In patients with secondary dengue or those with bleeding or elevated liver enzymes who revealed predisposition to severe outcomes, early activation as well as efficient downregulation of innate responses were compromised. Conclusion Our findings suggested that faulty/delayed kinetics of innate immune activation and downregulation was a driver of disease severity. We identified IFN-γ+CD56+CD3+ NKT cells and IL-6+ granulocytes at admission as novel early biomarkers that can predict the risk of progression to severity (composite AUC = 0.85–0.9). Strong correlations among multiple cytokine-secreting innate cell subsets revealed that coordinated early activation of the entire innate immune system in response to dengue virus infection contributed to resolution of infection and speedy recovery.
Collapse
Affiliation(s)
- Sai Pallavi Pradeep
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | | | - Nageswar R Manchala
- Division of Infectious Diseases Unit, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Arjun Vayal Veedu
- Division of Infectious Diseases Unit, St. John's Research Institute, St. John's Medical College, Bengaluru, India
| | - Rajani K Basavaraju
- Department of Medicine, Kempegowda Institute of Medical Sciences and Research Centre, Bengaluru, India
| | | | - Manikanta Ramakrishna
- Department of Medicine, Bengaluru Medical College and Research Institute, Bengaluru, India
| | - Yogitha Chandrakiran
- Department of Medicine, Kempegowda Institute of Medical Sciences and Research Centre, Bengaluru, India
| | | | - Shivaranjani Holigi
- Department of Medicine, Bengaluru Medical College and Research Institute, Bengaluru, India
| | - Tinku Thomas
- Department of Biostatistics, St. John's Medical College, Bengaluru, India
| | - Cecil R Ross
- Department of Medicine, St. John's Medical College, Bengaluru, India
| | - Mary Dias
- Department of Microbiology, St. John's Medical College, Bengaluru, India
| | - Vijaya Satchidanandam
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
9
|
Increased TNF- α Initiates Cytoplasmic Vacuolization in Whole Blood Coculture with Dengue Virus. J Immunol Res 2021; 2021:6654617. [PMID: 34041302 PMCID: PMC8121593 DOI: 10.1155/2021/6654617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/09/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022] Open
Abstract
During the acute febrile phase of dengue virus (DENV) infection, viremia can cause severe systemic immune responses accompanied by hematologic disorders. This study investigated the potential induction and mechanism of the cytopathic effects of DENV on peripheral blood cells ex vivo. At one day postinfection, there was viral nonstructural protein NS1 but no further virus replication measured in the whole blood culture. Notably, DENV exposure caused significant vacuolization in monocytic phagocytes. With a minor change in the complete blood cell count, except for a minor increase in neutrophils and a significant decrease in monocytes, the immune profiling assay identified several changes, particularly a significant reduction in CD14-positive monocytes as well as CD11c-positive dendritic cells. Abnormal production of TNF-α was highly associated with the induction of vacuolization. Manipulating TNF-α expression resulted in cytopathogenic effects. These results demonstrate the potential hematological damage caused by ex vivo DENV-induced TNF-α.
Collapse
|
10
|
Upasani V, Vo HTM, Auerswald H, Laurent D, Heng S, Duong V, Rodenhuis-Zybert IA, Dussart P, Cantaert T. Direct Infection of B Cells by Dengue Virus Modulates B Cell Responses in a Cambodian Pediatric Cohort. Front Immunol 2021; 11:594813. [PMID: 33643283 PMCID: PMC7907177 DOI: 10.3389/fimmu.2020.594813] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Dengue is an acute viral disease caused by dengue virus (DENV), which is transmitted by Aedes mosquitoes. Symptoms of DENV infection range from inapparent to severe and can be life-threatening. DENV replicates in primary immune cells such as dendritic cells and macrophages, which contribute to the dissemination of the virus. Susceptibility of other immune cells such as B cells to direct infection by DENV and their subsequent response to infection is not well defined. In a cohort of 60 Cambodian children, we showed that B cells are susceptible to DENV infection. Moreover, we show that B cells can support viral replication of laboratory adapted and patient-derived DENV strains. B cells were permissive to DENV infection albeit low titers of infectious virions were released in cell supernatants CD300a, a phosphatidylserine receptor, was identified as a potential attachment factor or receptor for entry of DENV into B cells. In spite of expressing Fcγ-receptors, antibody-mediated enhancement of DENV infection was not observed in B cells in an in vitro model. Direct infection by DENV induced proliferation of B cells in dengue patients in vivo and plasmablast/plasma cell formation in vitro. To summarize, our results show that B cells are susceptible to direct infection by DENV via CD300a and the subsequent B cell responses could contribute to dengue pathogenesis.
Collapse
Affiliation(s)
- Vinit Upasani
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.,Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Denis Laurent
- Kantha Bopha Children Hospital, Phnom Penh, Cambodia
| | - Sothy Heng
- Kantha Bopha Children Hospital, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Izabela A Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| |
Collapse
|
11
|
Thibodeaux JJ, Nuñez D, Rivera A. A generalized within-host model of dengue infection with a non-constant monocyte production rate. JOURNAL OF BIOLOGICAL DYNAMICS 2020; 14:143-161. [PMID: 32122254 DOI: 10.1080/17513758.2020.1733678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 02/04/2020] [Indexed: 06/10/2023]
Abstract
In this paper, we generalize a previous model of within-host dengue infection with a nonconstant monocyte production rate. We establish the existence of three equilibria and give some local stability results. We then estimate three parameters in the model from clinical data for dengue virus serotype 2. It is then shown that the model can exhibit behaviours that are not possible under the assumption of constant monocyte production. Lastly, we perform a sensitivity analysis of the model in two contexts, antiviral treatment and immunostimulatory treatment. The results predict that antiviral treatments that reduce the viral replication rate in infected monocytes are the most effective, while immunostimulatory treatments that increase the rate at which infected monocytes are removed are best.
Collapse
Affiliation(s)
- Jeremy J Thibodeaux
- Department of Mathematics and Computer Science, Loyola University New Orleans, New Orleans, LA, USA
| | - Daniel Nuñez
- Department of Natural Sciences and Mathematics, Javeriana University Cali, Cali, Colombia
| | - Andres Rivera
- Department of Natural Sciences and Mathematics, Javeriana University Cali, Cali, Colombia
| |
Collapse
|
12
|
Cloutier M, Nandi M, Ihsan AU, Chamard HA, Ilangumaran S, Ramanathan S. ADE and hyperinflammation in SARS-CoV2 infection- comparison with dengue hemorrhagic fever and feline infectious peritonitis. Cytokine 2020; 136:155256. [PMID: 32866898 PMCID: PMC7439999 DOI: 10.1016/j.cyto.2020.155256] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic has rapidly spread around the world with significant morbidity and mortality in a subset of patients including the elderly. The poorer outcomes are associated with 'cytokine storm-like' immune responses, otherwise referred to as 'hyperinflammation'. While most of the infected individuals show minimal or no symptoms and recover spontaneously, a small proportion of the patients exhibit severe symptoms characterized by extreme dyspnea and low tissue oxygen levels, with extensive damage to the lungs referred to as acute respiratory distress symptom (ARDS). The consensus is that the hyperinflammatory response of the host is akin to the cytokine storm observed during sepsis and is the major cause of death. Uncertainties remain on the factors that lead to hyperinflammatory response in some but not all individuals. Hyperinflammation is a common feature in different viral infections such as dengue where existing low-titer antibodies to the virus enhances the infection in immune cells through a process called antibody-dependent enhancement or ADE. ADE has been reported following vaccination or secondary infections with other corona, Ebola and dengue virus. Detailed analysis has shown that antibodies to any viral epitope can induce ADE when present in sub-optimal titers or is of low affinity. In this review we will discuss ADE in the context of dengue and coronavirus infections including Covid-19.
Collapse
Affiliation(s)
- Maryse Cloutier
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Madhuparna Nandi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Awais Ullah Ihsan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugues Allard Chamard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche Clinique, Centre Hospitalier d'Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche Clinique, Centre Hospitalier d'Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche Clinique, Centre Hospitalier d'Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
13
|
Narayan R, Tripathi S. Intrinsic ADE: The Dark Side of Antibody Dependent Enhancement During Dengue Infection. Front Cell Infect Microbiol 2020; 10:580096. [PMID: 33123500 PMCID: PMC7573563 DOI: 10.3389/fcimb.2020.580096] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 08/31/2020] [Indexed: 01/14/2023] Open
Abstract
Dengue fever is an Aedes mosquito-borne illness caused by any one of the four different dengue virus (DENV) serotypes (1–4) and manifests in the form of symptoms ranging from mild or asymptomatic to severe disease with vascular leakage, leading to shock, and viral hemorrhagic syndrome. Increased risk of severe disease occurs during secondary infection with a virus serotype distinct from that of prior dengue infection. This occurs by antibody dependent enhancement (ADE) of infection, wherein sub-neutralizing antibodies against the virus particles opsonize dengue virus entry via formation of immune complexes that interact with fragment crystallizable gamma receptors (FcγR) on monocytes, dendritic cells, and macrophages. The ADE phenomenon has two components: Extrinsic and Intrinsic ADE. While extrinsic ADE contributes to enhanced virus entry, intrinsic ADE results in heightened virus production by inhibition of type1 interferon and activation of interleukin-10 biosynthesis, thereby favoring a Th2 type immune response. Intrinsic ADE has greater contribution in enhancing Dengue replication as compared to extrinsic ADE. Detailed elucidation of intrinsic ADE during secondary dengue infection can increase our understanding of DENV-pathogenesis and aid in the development of host-targeting antivirals. Here we review literature focusing on intrinsic factors contributing to severe dengue pathology and suggest possible avenues for further research.
Collapse
Affiliation(s)
- Rohan Narayan
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bengaluru, India.,Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
| | - Shashank Tripathi
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bengaluru, India.,Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
14
|
Valliyott L, Dungdung R, Pilankatta R. Semi-quantification of antibody-dependent enhancement (ADE) in the uptake of Adenovirus serotype 5 into THP-1 cells. Anal Biochem 2020; 591:113568. [PMID: 31881180 DOI: 10.1016/j.ab.2019.113568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/17/2022]
Abstract
Replication defective recombinant Ad5 vectors (rAdV5) are extensively explored for its applications in gene therapy and vaccine delivery. Ad5 enter into monocytes and macrophages through CAR independent route as an immune complex termed as antibody-dependent enhancement (ADE). We developed an effective method for estimating the ADE of rAdV5 encoding GFP (rAdV5-GFP) into THP-1 cells, using fluorimetric semi-quantification of GFP. Initially, twenty numbers of human sera samples were screened in HeLa cells for anti-Ad5 antibody titer using neutralization assay. Uptake of rAdV5-GFP in THP-1 cells was observed only after pre-incubation with the serially diluted human sera which are attributed to ADE. The optimal dilution which showed the maximum GFP expression as per the fluorescence microscopic analysis in THP-1 cells was used for further analysis. Fluorimetric analysis of the THP-1 cell lysate showed a maximum GFP intensity of 17058 RFU, which was equivalent to the 0.397 pmoles of Alexa Fluor 488 under the same experimental condition. Similarly, immunoblot analysis of GFP in THP-1 cell lysate and HeLa cell lysate confirmed the entry of rAdV5-GFP into the cells. The assay can serve as a platform for understanding the molecular events involved in ADE for the uptake of viruses into immune cells.
Collapse
Affiliation(s)
- Lathika Valliyott
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Krishna Block, Central University of Kerala, Periya, Kasargod, Kerala, 671316, India
| | - Ranjeet Dungdung
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Krishna Block, Central University of Kerala, Periya, Kasargod, Kerala, 671316, India
| | - Rajendra Pilankatta
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Krishna Block, Central University of Kerala, Periya, Kasargod, Kerala, 671316, India.
| |
Collapse
|
15
|
Kuczera D, Assolini JP, Tomiotto-Pellissier F, Pavanelli WR, Silveira GF. Highlights for Dengue Immunopathogenesis: Antibody-Dependent Enhancement, Cytokine Storm, and Beyond. J Interferon Cytokine Res 2019; 38:69-80. [PMID: 29443656 DOI: 10.1089/jir.2017.0037] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Infection with dengue virus (DENV) can lead to a wide spectrum of clinical presentations, ranging from asymptomatic infection to death. It is estimated that the disease manifests only in 90 million cases out of the total 390 million yearly infections. Even though research has not yet elucidated which are the precise pathophysiological mechanisms that trigger severe forms of dengue, the infection elicits a critical immune response significant for dengue pathogenesis development. Understanding how the immune response to DENV is established and how it can resolve the infection or turn into an immunopathology is of great importance in DENV research. Currently, studies have extensively debated 2 hypotheses involving immune response: antibody-dependent enhancement and cytokine storm. However, despite its undeniable importance in severe forms of the disease, these 2 hypotheses are based on a primed immune status resulting from previous heterologous infection, abstaining them from explaining the severe forms of dengue in naive immune subjects, for example. Thus, it seems that a more intricate arrangement of causes and conditions must be achieved to severe dengue to occur. Among them, the cytokine network signature elicited, in association with viral aspects deserves special attention regarding the establishment of infection and evolution to pathogenesis. In this work, we intend to shed light on how those elements contribute to severe dengue development.
Collapse
Affiliation(s)
- Diogo Kuczera
- 1 Laboratório de Virologia Molecular, Instituto Carlos Chagas , ICC/Fiocruz/PR, Curitiba, Brazil
| | - João Paulo Assolini
- 2 Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina, Londrina, Brazil
| | - Fernanda Tomiotto-Pellissier
- 2 Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina, Londrina, Brazil
| | - Wander Rogério Pavanelli
- 2 Departamento de Ciências Patológicas, Centro de Ciências Biológicas , Universidade Estadual de Londrina, Londrina, Brazil
| | | |
Collapse
|
16
|
Kamaladasa A, Gomes L, Wijesinghe A, Jeewandara C, Toh YX, Jayathilaka D, Ogg GS, Fink K, Malavige GN. Altered monocyte response to the dengue virus in those with varying severity of past dengue infection. Antiviral Res 2019; 169:104554. [PMID: 31288040 DOI: 10.1016/j.antiviral.2019.104554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVE We sought to investigate the differences in monocyte immune responses to the dengue virus (DENV) in those who previously had either severe disease (past SD) or non-severe dengue (past NSD) following a secondary dengue infection. METHOD Monocytes from healthy individuals who had either past SD (n = 6) or past NSD (n = 6) were infected at MOI one with all four DENV serotypes following incubation with autologous serum. 36-hours post infection, levels of inflammatory cytokines and viral loads were measured in the supernatant and expression of genes involved in viral sensing and interferon signaling was determined. RESULTS Monocytes of individuals with past SD produced significantly higher viral loads (p = 0.0426 and cytokines (IL-10 p = 0.008, IL-1β p = 0.008 and IL-6 p = 0.0411) when infected with DENV serotypes they were not immune to, compared to those who has past NSD. Monocytes of individuals with past SD also produced significantly higher viral loads (p = 0.022) and cytokines (IL-10 p < 0.0001, IL-1β < 0.0001 and IL-6 p < 0.0001) when infected with DENV serotypes they were previously exposed to, despite the monocytes being infected in the presence of autologous serum. A significant upregulation of NLRP3 (p = 0.005), RIG-I (0.0004) and IFNB-1 (0.01) genes were observed in those who had past SD compared to past NSD when infected with non-immune DENV serotypes. CONCLUSION Monocytes from those with past SD appear to show marked differences in viral loads, viral sensing and production of inflammatory mediators in response to the DENV, when compared to those who experienced past NSD, suggesting that initial innate immune responses may influence the disease outcome.
Collapse
Affiliation(s)
- Achala Kamaladasa
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | - Laksiri Gomes
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | - Ayesha Wijesinghe
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | | | - Ying Xiu Toh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Deshni Jayathilaka
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka
| | - Graham S Ogg
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre and University of Oxford, OX3 9DS, UK
| | - Katja Fink
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| | - G N Malavige
- Centre for Dengue Research, University of Sri Jayawardanapura, Sri Lanka; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre and University of Oxford, OX3 9DS, UK.
| |
Collapse
|
17
|
Jasso-Miranda C, Herrera-Camacho I, Flores-Mendoza LK, Dominguez F, Vallejo-Ruiz V, Sanchez-Burgos GG, Pando-Robles V, Santos-Lopez G, Reyes-Leyva J. Antiviral and immunomodulatory effects of polyphenols on macrophages infected with dengue virus serotypes 2 and 3 enhanced or not with antibodies. Infect Drug Resist 2019; 12:1833-1852. [PMID: 31303775 PMCID: PMC6611719 DOI: 10.2147/idr.s210890] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/26/2019] [Indexed: 12/30/2022] Open
Abstract
Background: There is a lack of specific antiviral therapy against dengue virus (DENV) in current use. Therefore, a great proportion of dengue cases progress to severe clinical forms due to a complex interplay between virus and host immune response. It has been hypothesized that heterotypic non-neutralizing antibodies enhance DENV infection in phagocytic cells, and this induces an inflammatory response that is involved in the pathogenesis of severe dengue. Purpose: To identify the antiviral and immunomodulatory effects of polyphenols on dengue virus infection. Methods: Human U937-DC-SIGN macrophages were infected with DENV serotypes 2 or 3 in the presence or not of enhancing antibody 4G2. Viral titers and the secretion of tumor necrosis factor-alpha, IL-6, IL-10 and interferon-alpha were analyzed timely. Results: DENV infection alone induced high production of IL-6 and TNF-α, but in the presence of 4G2 antibody, viral titers and TNF-α secretion were potentiated. Based on anti-inflammatory antecedents, the polyphenols curcumin, fisetin, resveratrol, apigenin, quercetin and rutin were tested for antiviral and immunomodulatory properties. Only quercetin and fisetin inhibited DENV-2 and DENV-3 infection in the absence or presence of enhancing antibody (>90%, p<0.001); they also inhibited TNF-α and IL-6 secretion (p<0.001). Conclusion: Quercetin and fisetin down-regulate the production of proinflammatory cytokines induced by DENV infection enhanced by antibodies a mechanism involved in severe dengue.
Collapse
Affiliation(s)
- Carolina Jasso-Miranda
- Laboratory of Immunology and Virology, East Biomedical Research Center, Mexican Institute of Social Security (IMSS), CP 74360 Metepec, Puebla, México.,Laboratory of Biochemistry and Molecular Biology, Center of Chemistry, Institute of Sciences, Meritorious Autonomous University of Puebla, CP 72570 San Manuel, Puebla, Mexico
| | - Irma Herrera-Camacho
- Laboratory of Biochemistry and Molecular Biology, Center of Chemistry, Institute of Sciences, Meritorious Autonomous University of Puebla, CP 72570 San Manuel, Puebla, Mexico
| | - Lilian Karem Flores-Mendoza
- Department of Chemical, Biologic and Agricultural Sciences, Science and Enginery Division, University of Sonora, CP 85880 Navojoa, Sonora, Mexico
| | - Fabiola Dominguez
- Laboratory of Immunology and Virology, East Biomedical Research Center, Mexican Institute of Social Security (IMSS), CP 74360 Metepec, Puebla, México
| | - Veronica Vallejo-Ruiz
- Laboratory of Immunology and Virology, East Biomedical Research Center, Mexican Institute of Social Security (IMSS), CP 74360 Metepec, Puebla, México
| | | | - Victoria Pando-Robles
- Infectious Disease Research Center, National Institute of Public Health, CP 62100 Cuernavaca, Morelos, Mexico
| | - Gerardo Santos-Lopez
- Laboratory of Immunology and Virology, East Biomedical Research Center, Mexican Institute of Social Security (IMSS), CP 74360 Metepec, Puebla, México
| | - Julio Reyes-Leyva
- Laboratory of Immunology and Virology, East Biomedical Research Center, Mexican Institute of Social Security (IMSS), CP 74360 Metepec, Puebla, México
| |
Collapse
|
18
|
Sun P, Williams M, Nagabhushana N, Jani V, Defang G, Morrison BJ. NK Cells Activated through Antibody-Dependent Cell Cytotoxicity and Armed with Degranulation/IFN-γ Production Suppress Antibody-dependent Enhancement of Dengue Viral Infection. Sci Rep 2019; 9:1109. [PMID: 30710094 PMCID: PMC6358599 DOI: 10.1038/s41598-018-36972-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/02/2018] [Indexed: 12/20/2022] Open
Abstract
Antibody (Ab)-dependent enhancement (ADE) is a hypothesized mechanism of increased disease severity during secondary dengue virus (DENV) infection. This study investigates Ab-dependent cell cytotoxicity (ADCC) in counteracting ADE. In our system, DENV and DENV-immune sera were added to peripheral blood mononuclear cells (PBMCs), and ADE and NK cell activation were simultaneously monitored. ADE was detected in monocytes and a concurrent activation of NK cells was observed. Activated NK cells expressed IFN-γ and CD107a. IFN-γ was detected at 24 hours (24 h) followed by a rapid decline; CD107a expression peaked at 48 h and persisted for >7 days. Optimal activation of NK cells required the presence of enhancement serum together with ADE-affected monocytes and soluble factors, suggesting the coexistence of the counteractive ADCC Abs, in the same ADE-serum, capable of strongly promoting NK cell activation. The function of NK cells against ADE was demonstrated using a depletion assay. NK cell-depleted PBMCs had increased ADE as compared to whole PBMCs. Conversely, adding activated NK cells back into the NK-depleted-PBMCs or to purified monocytes decreased ADE. Blocking IFN-γ expression also increased ADE. The study suggests that under ADE conditions, NK cells can be activated by ADCC Abs and can control the magnitude of ADE.
Collapse
Affiliation(s)
- Peifang Sun
- Henry Jackson Foundation, Bethesda, MD, USA.
| | - Maya Williams
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | | | | | - Gabriel Defang
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| | - Brian J Morrison
- Viral and Rickettsial Diseases Department, Infectious Diseases Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| |
Collapse
|
19
|
Castillo JA, Naranjo JS, Rojas M, Castaño D, Velilla PA. Role of Monocytes in the Pathogenesis of Dengue. Arch Immunol Ther Exp (Warsz) 2018; 67:27-40. [DOI: 10.1007/s00005-018-0525-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/03/2018] [Indexed: 11/29/2022]
|
20
|
Supramaniam A, Lui H, Bellette BM, Rudd PA, Herrero LJ. How myeloid cells contribute to the pathogenesis of prominent emerging zoonotic diseases. J Gen Virol 2018; 99:953-969. [DOI: 10.1099/jgv.0.001024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Aroon Supramaniam
- 1Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | - Hayman Lui
- 2School of Medicine, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | | | - Penny A. Rudd
- 1Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | - Lara J. Herrero
- 1Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia
- 2School of Medicine, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| |
Collapse
|
21
|
Plasma Neutrophil Elastase, α 1-Antitrypsin, α 2-Macroglobulin and Neutrophil Elastase-α 1-Antitrypsin Complex Levels in patients with Dengue Fever. Indian J Clin Biochem 2017; 33:218-221. [PMID: 29651215 DOI: 10.1007/s12291-017-0658-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/28/2017] [Indexed: 01/13/2023]
Abstract
Dengue fever (DF) is characterized by systemic inflammatory response including neutrophil activation leading to uncontrolled elastase activity. This study was aimed to measure the activity of plasma neutrophil elastase (NE), its endogenous inhibitors α1-antitrypsin (α1-AT) and α2-macroglobulin (α2-MG) and elastase in complex with α1-AT (NE-α1-AT complex) in DF. 50 dengue patients [39 DF and 11 dengue hemorrhagic fever (DHF)] and 52 healthy subjects were included in the study. NE was measured using N-succinyl-tri-alanine-p-nitroanilide as substrate. α1-AT, α2-MG and NE-α1-AT complex were estimated by ELISA. The result analysis indicated that the dengue patients had significantly higher elastase activity with significantly reduced inhibitor levels compared to controls. Between DF and DHF patients, DHF group had significantly higher elastase activity. In conclusion, significantly elevated NE and reduced inhibitors level in dengue fever indicate these parameters could be of significance in DF particularly for the assessment of progression of inflammatory processes.
Collapse
|
22
|
Sun P, Morrison BJ, Beckett CG, Liang Z, Nagabhushana N, Li A, Porter KR, Williams M. NK cell degranulation as a marker for measuring antibody-dependent cytotoxicity in neutralizing and non-neutralizing human sera from dengue patients. J Immunol Methods 2016; 441:24-30. [PMID: 27856192 DOI: 10.1016/j.jim.2016.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 10/26/2016] [Accepted: 11/12/2016] [Indexed: 11/30/2022]
Abstract
The study assessed antibody-dependent NK cell degranulation, a biomarker relevant to antibody-dependent cell cytotoxicity (ADCC), to analyze dengue immune sera. We first determined binding intensity of patient sera to the surface of DENV-infected cells and examined the types of antigens expressed on infected cells. Antigens from pre-membrane (PreM) and envelope (E), but not from NS proteins were detected on the surface of infected cells. After adding NK cells to infected target cells previously treated with patient sera, rapid NK cell degranulation was observed. Non-neutralizing patient sera generated comparable NK cell degranulation as that of neutralizing sera, suggesting ADCC may be a protective mechanism apart from Ab neutralization. The level of NK cell degranulation varied dramatically among human individuals and was associated with the level of CD16 expression on NK cells, informing on the complexity of ADCC among human population.
Collapse
Affiliation(s)
| | | | | | | | | | - An Li
- University of Maryland College Park, United States
| | | | | |
Collapse
|
23
|
Taylor A, Foo SS, Bruzzone R, Dinh LV, King NJC, Mahalingam S. Fc receptors in antibody-dependent enhancement of viral infections. Immunol Rev 2016; 268:340-64. [PMID: 26497532 PMCID: PMC7165974 DOI: 10.1111/imr.12367] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sensitization of the humoral immune response to invading viruses and production of antiviral antibodies forms part of the host antiviral repertoire. Paradoxically, for a number of viral pathogens, under certain conditions, antibodies provide an attractive means of enhanced virus entry and replication in a number of cell types. Known as antibody‐dependent enhancement (ADE) of infection, the phenomenon occurs when virus‐antibody immunocomplexes interact with cells bearing complement or Fc receptors, promoting internalization of the virus and increasing infection. Frequently associated with exacerbation of viral disease, ADE of infection presents a major obstacle to the prevention of viral disease by vaccination and is thought to be partly responsible for the adverse effects of novel antiviral therapeutics such as intravenous immunoglobulins. There is a growing body of work examining the intracellular signaling pathways and epitopes responsible for mediating ADE, with a view to aiding rational design of antiviral strategies. With in vitro studies also confirming ADE as a feature of infection for a growing number of viruses, challenges remain in understanding the multilayered molecular mechanisms of ADE and its effect on viral pathogenesis.
Collapse
Affiliation(s)
- Adam Taylor
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| | - Suan-Sin Foo
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong SAR, Hong Kong.,Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Luan Vu Dinh
- Discipline of Pathology, Bosch Institute, School of Medical Sciences, Sydney Medical School, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Nicholas J C King
- Discipline of Pathology, Bosch Institute, School of Medical Sciences, Sydney Medical School, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Suresh Mahalingam
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| |
Collapse
|
24
|
Findlay JS, Ulaeto D, D'Elia RV. Cytokines and viral hemorrhagic fever: potential for therapeutic intervention. Future Virol 2015. [DOI: 10.2217/fvl.15.5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT The recent Ebola outbreak in West Africa highlights the need to improve our understanding of why viral hemorrhagic fevers (VHFs) are so devastating. There is a requirement to generate effective prophylactics, such as vaccines, and therapies, especially those that are effective postsymptomatically. For a range of pathogens, it appears that overstimulation of pro-inflammatory cytokines, the ‘cytokine storm’, causes serious immunopathology in patients. In this review, we will focus on the cytokine response following infection by representatives of the viruses which can cause VHF: Ebola virus and Marburg virus, Crimean–Congo hemorrhagic fever virus, Dengue virus, Junin and Lassa virus. Specifically, the role of the cytokine storm in causing VHF and the use of therapeutic immunomodulatory compounds to help treat these fatal and debilitating diseases will be explored.
Collapse
Affiliation(s)
- James S Findlay
- Biomedical Sciences, Defence Science & Technology Laboratory (Dstl) Porton Down, Salisbury, SP4 0JQ, UK
| | - David Ulaeto
- Biomedical Sciences, Defence Science & Technology Laboratory (Dstl) Porton Down, Salisbury, SP4 0JQ, UK
| | - Riccardo V D'Elia
- Biomedical Sciences, Defence Science & Technology Laboratory (Dstl) Porton Down, Salisbury, SP4 0JQ, UK
| |
Collapse
|
25
|
Gu W, Guo L, Yu H, Niu J, Huang M, Luo X, Li R, Tian Z, Feng L, Wang Y. Involvement of CD16 in antibody-dependent enhancement of porcine reproductive and respiratory syndrome virus infection. J Gen Virol 2015; 96:1712-22. [PMID: 25752917 DOI: 10.1099/vir.0.000118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The immunological effect of porcine reproductive and respiratory syndrome disease virus (PRRSV) vaccines is thought to be influenced by a variety of host factors, in which antibody-dependent enhancement (ADE) of infection is one crucial factor. Here, we assessed the mechanism of ADE of PRRSV infection. First, we found that subneutralizing serum could induce ADE of PRRSV infection in porcine alveolar macrophages (PAMs). Quantitative PCR, Western blotting and flow cytometry revealed that CD16 is the most abundant Fcγ receptor (FcγR) expressed on the surface of PAMs; thus, the role of CD16 in ADE of PRRSV infection was examined in PAMs. By using functional blocking antibodies, we demonstrated that CD16 is involved in enhanced virus production in PRRSV-antibody immune complex-infected PAMs. Because PAMs co-express different FcγR isoforms, we evaluated the effects of CD16 in FcγR-non-bearing cells by transfection. Using these engineered cells, we found that CD16 could specifically bind to the PRRSV-antibody immune complex and subsequently mediate internalization of the virus, resulting in the generation of progeny virus. We also showed that efficient expression of CD16 required association of the FcR γ-chain. Together, our findings provide significant new insights into PRRSV infection, which can be enhanced by CD16-mediated PRRSV-antibody immune complexes. This CD16-mediated ADE may induce a shift in PRRSV tropism towards CD16-expressing cells, distributing virus to more organs during virus infection.
Collapse
Affiliation(s)
- Weihong Gu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Longjun Guo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Haidong Yu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Junwei Niu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Mingming Huang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Xiaolei Luo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Ren Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Zhijun Tian
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Li Feng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yue Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| |
Collapse
|
26
|
Sam SS, Teoh BT, Chinna K, AbuBakar S. High producing tumor necrosis factor alpha gene alleles in protection against severe manifestations of dengue. Int J Med Sci 2015; 12:177-86. [PMID: 25589894 PMCID: PMC4293183 DOI: 10.7150/ijms.8988] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 12/09/2014] [Indexed: 11/13/2022] Open
Abstract
UNLABELLED Dengue virus (DENV) infection usually presents with mild self-limiting dengue fever (DF). Few however, would present with the more severe form of the disease, dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). In the present study, the association between IL-12B, IL-10 and TNF-α gene polymorphisms and dengue severity was investigated. METHODS A case-control study was performed on a total of 120 unrelated controls, 86 DF patients and 196 DHF/DSS patients. The polymorphisms in IL-12B, IL-10 and TNF-α genes were genotyped using PCR-RFLP and PCR-sequencing methods. RESULTS A protective association of TNF-α -308A allele and -308GA genotype against DHF/DSS was observed, while TNF-α -238A allele and -238GA genotype were associated with DHF/DSS. A combination of TNF-α -308GA+AA genotype and IL-10 non-GCC haplotypes, IL-12B pro homozygotes (pro1/pro1, pro2/pro2) and IL-12B 3'UTR AC were significantly correlated with protective effects against DHF/DSS. An association between the cytokine gene polymorphisms and protection against the clinical features of severe dengue including thrombocytopenia and increased liver enzymes was observed in this study. CONCLUSION The overall findings of the study support the correlation of high-producer TNF-α genotypes combined with low-producer IL-10 haplotypes and IL-12B genotypes in reduced risk of DHF/DSS.
Collapse
Affiliation(s)
- Sing-Sin Sam
- 1. Tropical Infectious Diseases Research and Education Center (TIDREC), Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Boon-Teong Teoh
- 1. Tropical Infectious Diseases Research and Education Center (TIDREC), Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Karuthan Chinna
- 2. Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sazaly AbuBakar
- 1. Tropical Infectious Diseases Research and Education Center (TIDREC), Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Pierson TC, Diamond MS. A game of numbers: the stoichiometry of antibody-mediated neutralization of flavivirus infection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 129:141-66. [PMID: 25595803 DOI: 10.1016/bs.pmbts.2014.10.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The humoral response contributes to the protection against viral pathogens. Although antibodies have the potential to inhibit viral infections via several mechanisms, an ability to neutralize viruses directly may be particularly important. Neutralizing antibody titers are commonly used as predictors of protection from infection, especially in the context of vaccine responses and immunity. Despite the simplicity of the concept, how antibody binding results in virus inactivation is incompletely understood despite decades of research. Flaviviruses have been an attractive system in which to seek a structural and quantitative understanding of how antibody interactions with virions modulate infection because of the contribution of antibodies to both protection and pathogenesis. This review will present a stoichiometric model of antibody-mediated neutralization of flaviviruses and discuss how these concepts can inform the development of vaccines and antibody-based therapeutics.
Collapse
Affiliation(s)
- Theodore C Pierson
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
28
|
Salazar MI, del Angel RM, Lanz-Mendoza H, Ludert JE, Pando-Robles V. The role of cell proteins in dengue virus infection. J Proteomics 2014; 111:6-15. [DOI: 10.1016/j.jprot.2014.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/26/2014] [Accepted: 06/02/2014] [Indexed: 01/12/2023]
|
29
|
Fu Y, Yip A, Seah PG, Blasco F, Shi PY, Hervé M. Modulation of inflammation and pathology during dengue virus infection by p38 MAPK inhibitor SB203580. Antiviral Res 2014; 110:151-7. [PMID: 25131378 DOI: 10.1016/j.antiviral.2014.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 08/04/2014] [Accepted: 08/07/2014] [Indexed: 11/29/2022]
Abstract
Dengue virus (DENV) infection could lead to dengue fever (DF), dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS). The disease outcome is controlled by both viral and host factors. Inflammation mediators from DENV-infected cells could contribute to increased vascular permeability, leading to severe DHF/DSS. Therefore, suppression of inflammation could be a potential therapeutic approach for treatment of dengue patients. In this context, p38 MAPK (mitogen-activated protein kinase) is a key enzyme that modulates the initiation of stress and inflammatory responses. Here we show that SB203580, a p38 MAPK inhibitor, suppressed the over production of DENV-induced pro-inflammatory mediators such as TNF-α, IL-8, and RANTES from human PBMCs, monocytic THP-1, and granulocyte KU812 cell lines. Oral administration of SB203580 in DENV-infected AG129 mice prevented hematocrit rise and lymphopenia, limited the development of inflammation and pathology (including intestine leakage), and significantly improved survival. These results, for the first time, have provided experimental evidence to imply that a short term inhibition of p38 MAPK may be beneficial to reduce disease symptoms in dengue patients.
Collapse
Affiliation(s)
- Yilong Fu
- Novartis Institute for Tropical Diseases, Singapore 138670, Singapore
| | - Andy Yip
- Novartis Institute for Tropical Diseases, Singapore 138670, Singapore
| | - Peck Gee Seah
- Novartis Institute for Tropical Diseases, Singapore 138670, Singapore
| | - Francesca Blasco
- Novartis Institute for Tropical Diseases, Singapore 138670, Singapore
| | - Pei-Yong Shi
- Novartis Institute for Tropical Diseases, Singapore 138670, Singapore.
| | - Maxime Hervé
- Novartis Institute for Tropical Diseases, Singapore 138670, Singapore.
| |
Collapse
|
30
|
Marinho CF, Azeredo EL, Torrentes-Carvalho A, Marins-Dos-Santos A, Kubelka CF, de Souza LJ, Cunha RV, de-Oliveira-Pinto LM. Down-regulation of complement receptors on the surface of host monocyte even as in vitro complement pathway blocking interferes in dengue infection. PLoS One 2014; 9:e102014. [PMID: 25061945 PMCID: PMC4111305 DOI: 10.1371/journal.pone.0102014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/13/2014] [Indexed: 12/16/2022] Open
Abstract
In dengue virus (DENV) infection, complement system (CS) activation appears to have protective and pathogenic effects. In severe dengue fever (DF), the levels of DENV non-structural-1 protein and of the products of complement activation, including C3a, C5a and SC5b-9, are higher before vascular leakage occurs, supporting the hypothesis that complement activation contributes to unfavourable outcomes. The clinical manifestations of DF range from asymptomatic to severe and even fatal. Here, we aimed to characterise CS by their receptors or activation product, in vivo in DF patients and in vitro by DENV-2 stimulation on monocytes. In comparison with healthy controls, DF patients showed lower expression of CR3 (CD11b), CR4 (CD11c) and, CD59 on monocytes. The DF patients who were high producers of SC5b-9 were also those that showed more pronounced bleeding or vascular leakage. Those findings encouraged us to investigate the role of CS in vitro, using monocytes isolated from healthy subjects. Prior blocking with CR3 alone (CD11b) or CR3 (CD11b/CD18) reduced viral infection, as quantified by the levels of intracellular viral antigen expression and soluble DENV non-structural viral protein. However, we found that CR3 alone (CD11b) or CR3 (CD11b/CD18) blocking did not influence major histocompatibility complex presentation neither active caspase-1 on monocytes, thus probably ruling out inflammasome-related mechanisms. Although it did impair the secretion of tumour necrosis factor alpha and interferon alpha. Our data provide strategies of blocking CR3 (CD11b) pathways could have implications for the treatment of viral infection by antiviral-related mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rivaldo Venâncio Cunha
- Department of Clinical Medicine, Universidade Federal do Mato Grosso do Sul, Campo Grande, Brazil
| | | |
Collapse
|
31
|
Pierson TC, Diamond MS. Vaccine Development as a Means to Control Dengue Virus Pathogenesis: Do We Know Enough? Annu Rev Virol 2014; 1:375-98. [PMID: 26958727 DOI: 10.1146/annurev-virology-031413-085453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dengue virus (DENV) is a mosquito-transmitted RNA virus responsible for 390 million infections each year and significant morbidity and mortality throughout tropical and subtropical regions of the world. Efforts to develop a DENV vaccine span 70 years and include the work of luminaries of the virus vaccine field. Although vaccines have been used to reduce the global health burden of other flaviviruses, the unique requirement for a single vaccine to protect against four different groups of dengue viruses, and the link between secondary infections and DENV disease pathogenesis, has limited success to date. In this review, we discuss several promising DENV vaccine candidates in clinical trials and assess how recent advances in understanding of DENV biology and immunity may expedite efforts toward the development of safe and effective vaccines.
Collapse
Affiliation(s)
- Theodore C Pierson
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892;
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, and Pathology & Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri 63110;
| |
Collapse
|
32
|
Malavige GN, Ogg GS. T cell responses in dengue viral infections. J Clin Virol 2013; 58:605-11. [PMID: 24220605 DOI: 10.1016/j.jcv.2013.10.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 10/04/2013] [Accepted: 10/19/2013] [Indexed: 11/17/2022]
Abstract
Dengue viral infections are the commonest mosquito borne viral infection in the world, affecting more than 100 countries and 390 million individuals annually. Currently, there are no effective antiviral drugs or an effective vaccine to prevent infection. A main hurdle in developing a safe and effective vaccine has been our poor understanding of the complex nature of the protective immune response in acute dengue infection and the presence of four dengue virus (DV) serotypes that are highly homologous. The role of DV specific T cells in the pathogenesis of severe clinical disease in not clear. It has been speculated that highly cross reactive T cells for the previous infecting heterologous DV serotype, which produce pro-inflammatory cytokines, contribute to disease pathogenesis. These cross reactive T cells are believed to be suboptimal in clearing the infection with the current DV-serotype. However, other studies have shown that cross-reactive DV-specific T cells are absent or present in very low frequency during acute infection, appearing only during the convalescent period in the majority of patients. Furthermore, significant apoptosis of T cells occurs in severe acute clinical disease. Overall therefore, it is unclear what role T cells play in contributing to disease pathogenesis during acute dengue infection. Existing data have been complicated by cross-reactivity in T cells assays. These findings can now be re-evaluated in the light of novel technologies to identify serotype-specific T cell responses.
Collapse
Affiliation(s)
- Gathsaurie Neelika Malavige
- Centre for Dengue Research, Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayawardanapura, Sri Lanka; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre and University of Oxford, OX3 9DS, UK.
| | | |
Collapse
|
33
|
Malavige GN, Jeewandara C, Alles KML, Salimi M, Gomes L, Kamaladasa A, Jayaratne SD, Ogg GS. Suppression of virus specific immune responses by IL-10 in acute dengue infection. PLoS Negl Trop Dis 2013; 7:e2409. [PMID: 24040431 PMCID: PMC3764236 DOI: 10.1371/journal.pntd.0002409] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 07/24/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Elevated IL-10 has been shown to be associated with severe dengue infection (DI). We proceeded to investigate the role of IL-10 in the pathogenesis of acute DI. MATERIALS AND METHODS Ex vivo and cultured IFNγ ELISpot assays for dengue virus (DENV) NS3 protein and non dengue viral proteins were carried out in 26 patients with acute DI (16 with dengue haemorrhagic fever) and 12 healthy dengue seropositive individuals from Sri Lanka. DENV serotype specific (SS) responses were determined by using a panel of SS peptides. RESULTS Serum IL-10 level were significantly higher (p = 0.02) in those who did not have in vitro responses to DENV-SS peptides (mean 144.2 pg/ml) when compared to those who responded (mean 75.7 pg/ml). DENV-NS3 specific ex vivo IFNγ ELISpot responses were also significantly lower (p = 0.0001) in those who did not respond to DENV-SS peptides (mean 42 SFU/million PBMCs) when compared to those who responded to DENV-SS peptides (mean 1024 SFU/million PBMCs). Serum IL-10 levels correlated significantly (p = 0.03) and inversely (Spearmans R = -0.45) with ex vivo DENV-NS3 specific responses but not with ex vivo non DENV specific responses (Spearmans R = -014, p = 0.52). Blockage of IL-10 in vitro significantly increased (p = 0.04) the ex vivo IFNγ ELISpot DENV-NS3 specific responses but had no effect on responses to non DENV proteins. CONCLUSION IL-10 appears to contribute to the pathogenesis of acute dengue infections by inhibiting DENV-specific T cell responses, which can be restored by blocking IL-10.
Collapse
Affiliation(s)
- Gathsaurie Neelika Malavige
- Centre for Dengue Research, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Nugegoda, Sri Lanka
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | - Chandima Jeewandara
- Centre for Dengue Research, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Nugegoda, Sri Lanka
| | - K. M. Luckmaal Alles
- Centre for Dengue Research, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Nugegoda, Sri Lanka
| | - Maryam Salimi
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Laksiri Gomes
- Centre for Dengue Research, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Nugegoda, Sri Lanka
| | - Achala Kamaladasa
- Centre for Dengue Research, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Nugegoda, Sri Lanka
| | - S. D. Jayaratne
- Department of Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Gangodawila, Nugegoda, Sri Lanka
| | - Graham Stuart Ogg
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Dermatology, Churchill Hospital, Oxford, United Kingdom
| |
Collapse
|
34
|
Moi ML, Takasaki T, Saijo M, Kurane I. Determination of antibody concentration as the main parameter in a dengue virus antibody-dependent enhancement assay using FcγR-expressing BHK cells. Arch Virol 2013; 159:103-16. [DOI: 10.1007/s00705-013-1787-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 04/26/2013] [Indexed: 11/29/2022]
|
35
|
Richter K, Oxenius A. Non-neutralizing antibodies protect from chronic LCMV infection independently of activating FcγR or complement. Eur J Immunol 2013; 43:2349-60. [PMID: 23749374 DOI: 10.1002/eji.201343566] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/22/2013] [Accepted: 06/04/2013] [Indexed: 01/10/2023]
Abstract
Chronic viral infections lead to CD8(+) T cell exhaustion, characterized by impaired cytokine secretion. The presence of the immune-regulatory cytokine IL-10 promotes chronic lymphocytic choriomeningitis virus (LCMV) Clone 13 infection in mice, whereas the absence of IL-10/IL-10R signaling early during infection results in viral clearance and higher percentages and numbers of antiviral, cytokine-producing T cells. However, it is currently unclear which cell populations and effector molecules are crucial to protect against chronic infection. In this study, we demonstrate that antiviral, LCMV-binding, non-neutralizing antibodies are needed, in addition to CD4(+) and CD8(+) T cells, to clear a high-dose LCMV infection in mice, in the absence of IL-10. The interaction between CD4(+) T cells and B cells in B-cell follicles via CD40/CD40L, in addition to class switch and/or somatic hypermutation, is crucial for viral control in the absence of IL-10. Interestingly, transfer of LCMV-binding non-neutralizing antibodies protected recipients from chronic infection. In addition, viral clearance in the absence of IL-10R signaling was independent of activating Fcγ receptors and complement. These data highlight that non-neutralizing antibodies effectively contribute to the control of LCMV infection when present prior to infection, suggesting that the induction of neutralizing antibodies is not implicitly necessary for the generation of successful vaccines.
Collapse
|
36
|
Torres S, Hernández JC, Giraldo D, Arboleda M, Rojas M, Smit JM, Urcuqui-Inchima S. Differential expression of Toll-like receptors in dendritic cells of patients with dengue during early and late acute phases of the disease. PLoS Negl Trop Dis 2013; 7:e2060. [PMID: 23469297 PMCID: PMC3585035 DOI: 10.1371/journal.pntd.0002060] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 01/01/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Dengue hemorrhagic fever (DHF) is observed in individuals that have pre-existing heterotypic dengue antibodies and is associated with increased viral load and high levels of pro-inflammatory cytokines early in infection. Interestingly, a recent study showed that dengue virus infection in the presence of antibodies resulted in poor stimulation of Toll-like receptors (TLRs), thereby facilitating virus particle production, and also suggesting that TLRs may contribute to disease pathogenesis. METHODOLOGY/PRINCIPAL FINDINGS We evaluated the expression levels of TLR2, 3, 4 and 9 and the co-stimulatory molecules CD80 and CD86 by flow cytometry. This was evaluated in monocytes, in myeloid and plasmacytoid dendritic cells (mDCs and pDCs) from 30 dengue patients with different clinical outcomes and in 20 healthy controls. Increased expression of TLR3 and TLR9 in DCs of patients with dengue fever (DF) early in infection was detected. In DCs from patients with severe manifestations, poor stimulation of TLR3 and TLR9 was observed. In addition, we found a lower expression of TLR2 in patients with DF compared to DHF. Expression levels of TLR4 were not affected. Furthermore, the expression of CD80 and CD86 was altered in mDCs and CD86 in pDCs of severe dengue cases. We show that interferon alpha production decreased in the presence of dengue virus after stimulation of PBMCs with the TLR9 agonist (CpG A). This suggests that the virus can affect the interferon response through this signaling pathway. CONCLUSIONS/SIGNIFICANCE These results show that during dengue disease progression, the expression profile of TLRs changes depending on the severity of the disease. Changes in TLRs expression could play a central role in DC activation, thereby influencing the innate immune response.
Collapse
Affiliation(s)
- Silvia Torres
- Grupo Inmunovirología, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Juan Carlos Hernández
- Grupo Inmunovirología, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
- Grupo Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Diana Giraldo
- Grupo Inmunovirología, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| | - Margarita Arboleda
- Instituto Colombiano de Medicina Tropical, Universidad CES, Sabaneta, Antioquia, Colombia
| | - Mauricio Rojas
- Grupo de Inmunología Celular e Inmunogenética, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Jolanda M. Smit
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
- * E-mail:
| |
Collapse
|
37
|
The battle between infection and host immune responses of dengue virus and its implication in dengue disease pathogenesis. ScientificWorldJournal 2013; 2013:843469. [PMID: 23476150 PMCID: PMC3582169 DOI: 10.1155/2013/843469] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 01/03/2013] [Indexed: 01/01/2023] Open
Abstract
Dengue virus (DENV) is a mosquito-transmitted single stranded RNA virus belonging to genus Flavivirus. The virus is endemic in the tropical and subtropical countries of the world, causing diseases classified according to symptoms and severity (from mild to severe) as dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. Among a variety of human cell types targeted by DENV, monocytes, macrophages, and dendritic cells are members of innate immunity, capable of mounting rapid inflammatory responses. These cells are also major antigen presenting cells, responsible for activating the adaptive immunity for long-term memory. This paper is an overview of the current understanding of the following mutually affected aspects: DENV structure, viral infectivity, cellular receptors, innate immune response, and adaptive immunity.
Collapse
|
38
|
Flipse J, Wilschut J, Smit JM. Molecular mechanisms involved in antibody-dependent enhancement of dengue virus infection in humans. Traffic 2012; 14:25-35. [PMID: 22998156 DOI: 10.1111/tra.12012] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 12/19/2022]
Abstract
Dengue is the most common arthropod-borne viral infection in humans with ∼50 million cases annually worldwide. In recent decades, a steady increase in the number of severe dengue cases has been seen. Severe dengue disease is most often observed in individuals that have pre-existing immunity against heterotypic dengue subtypes and in infants with low levels of maternal dengue antibodies. The generally accepted hypothesis explaining the immunopathogenesis of severe dengue is called antibody-dependent enhancement of dengue infection. Here, circulating antibodies bind to the newly infecting virus but do not neutralize infection. Rather, these antibodies increase the infected cell mass and virus production. Additionally, antiviral responses are diminished allowing massive virus particle production early in infection. The large infected cell mass and the high viral load are prelude for severe disease development. In this review, we discuss what is known about the trafficking of dengue virus in its human host cells, and the signalling pathways activated after virus detection, both in the absence and presence of antibodies against the virus. This review summarizes work that aims to better understand the complex immunopathogenesis of severe dengue disease.
Collapse
Affiliation(s)
- Jacky Flipse
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|