1
|
Slowikowski E, Willems C, Lemes RMR, Schuermans S, Berghmans N, Rocha RPF, Martens E, Proost P, Delang L, Marques RE, Filho JCA, Marques PE. A central role for CCR2 in monocyte recruitment and blood-brain barrier disruption during Usutu virus encephalitis. J Neuroinflammation 2025; 22:107. [PMID: 40241134 PMCID: PMC12004732 DOI: 10.1186/s12974-025-03435-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Usutu virus (USUV) is an emerging neurotropic flavivirus capable of causing encephalitis in humans. Here, our main goal was to characterize the innate immune response in the brain during USUV encephalitis and to identify strategies to control disease severity. Using an immunocompetent mouse model of USUV encephalitis, we showed that microglia activation, blood-brain barrier (BBB) disruption and inflammatory monocyte recruitment are hallmarks of disease 6 days post infection. Activated microglia were in close association to USUV-infected cells, concomitantly with elevated levels of IL-6, IFN-γ, CCL2, CCL5, CXCL10 and CXCL1 in the brain. Monocyte recruitment was CCR2-dependent and driven by IFN-γ and CCL2 production beneath the brain vasculature. Moreover, CCR2 deficiency inhibited microglia activation and BBB disruption, showing the central role of CCR2 in USUV encephalitis. Accordingly, treatment with dexamethasone prevented pro-inflammatory mediator production and reduced leukocyte recruitment significantly, restraining encephalitis severity. Concluding, USUV encephalitis is driven by CCR2-mediated monocyte recruitment and BBB disruption, and blocked therapeutically by glucocorticoids.
Collapse
Affiliation(s)
- Emily Slowikowski
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Céleste Willems
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Robertha Mariana Rodrigues Lemes
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sara Schuermans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Nele Berghmans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Rebeca Paiva Fróes Rocha
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Erik Martens
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Leen Delang
- Virus-Host Interactions and Therapeutic Approaches (VITA) Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - José Carlos Alves Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Spiteri AG, van Vreden C, Ashhurst TM, Niewold P, King NJC. Clodronate is not protective in lethal viral encephalitis despite substantially reducing inflammatory monocyte infiltration in the CNS. Front Immunol 2023; 14:1203561. [PMID: 37545511 PMCID: PMC10403146 DOI: 10.3389/fimmu.2023.1203561] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
Bone marrow (BM)-derived monocytes induce inflammation and tissue damage in a range of pathologies. In particular, in a mouse model of West Nile virus (WNV) encephalitis (WNE), nitric oxide-producing, Ly6Chi inflammatory monocytes from the BM are recruited to the central nervous system (CNS) and contribute to lethal immune pathology. Reducing the migration of these cells into the CNS using monoclonal antibody blockade, immune-modifying particles or CSF-1R inhibitors reduces neuroinflammation, improving survival and/or clinical outcomes. Macrophages can also be targeted more broadly by administration of clodronate-encapsulated liposomes, which induce apoptosis in phagocytes. In this study, clodronate reduced the inflammatory infiltrate by 70% in WNE, however, surprisingly, this had no effect on disease outcome. More detailed analysis demonstrated a compensatory increase in neutrophils and enhanced activation status of microglia in the brain. In addition, we observed increased numbers of Ly6Chi BM monocytes with an increased proliferative capacity and expression of SCA-1 and CD16/32, potentially indicating output of immature cells from the BM. Once in the brain, these cells were more phagocytic and had a reduced expression of antigen-presenting molecules. Lastly, we show that clodronate also reduces non-myeloid cells in the spleen and BM, as well as ablating red blood cells and their proliferation. These factors likely impeded the therapeutic potential of clodronate in WNE. Thus, while clodronate provides an excellent system to deplete macrophages in the body, it has larger and broader effects on the phagocytic and non-phagocytic system, which must be considered in the interpretation of data.
Collapse
Affiliation(s)
- Alanna G. Spiteri
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Caryn van Vreden
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Thomas M. Ashhurst
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
| | - Paula Niewold
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, Netherlands
| | - Nicholas J. C. King
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
3
|
Benzarti E, Murray KO, Ronca SE. Interleukins, Chemokines, and Tumor Necrosis Factor Superfamily Ligands in the Pathogenesis of West Nile Virus Infection. Viruses 2023; 15:v15030806. [PMID: 36992514 PMCID: PMC10053297 DOI: 10.3390/v15030806] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
West Nile virus (WNV) is a mosquito-borne pathogen that can lead to encephalitis and death in susceptible hosts. Cytokines play a critical role in inflammation and immunity in response to WNV infection. Murine models provide evidence that some cytokines offer protection against acute WNV infection and assist with viral clearance, while others play a multifaceted role WNV neuropathogenesis and immune-mediated tissue damage. This article aims to provide an up-to-date review of cytokine expression patterns in human and experimental animal models of WNV infections. Here, we outline the interleukins, chemokines, and tumor necrosis factor superfamily ligands associated with WNV infection and pathogenesis and describe the complex roles they play in mediating both protection and pathology of the central nervous system during or after virus clearance. By understanding of the role of these cytokines during WNV neuroinvasive infection, we can develop treatment options aimed at modulating these immune molecules in order to reduce neuroinflammation and improve patient outcomes.
Collapse
Affiliation(s)
- Emna Benzarti
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA
- William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Kristy O Murray
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA
- William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Immunology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shannon E Ronca
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX 77030, USA
- William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Immunology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
4
|
Nobs SP, Kopf M. Tissue-resident macrophages: guardians of organ homeostasis. Trends Immunol 2021; 42:495-507. [PMID: 33972166 DOI: 10.1016/j.it.2021.04.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Tissue-resident macrophages (MTR) have recently emerged as a key rheostat capable of regulating the balance between organ health and disease. In most organs, ontogenetically and functionally distinct macrophage subsets fulfill a plethora of functions specific to their tissue environment. In this review, we summarize recent findings regarding the ontogeny and functions of macrophage populations in different mammalian tissues, describing how these cells regulate tissue homeostasis and how they can contribute to inflammation. Furthermore, we highlight new developments concerning certain general principles of tissue macrophage biology, including the importance of metabolism for understanding macrophage activation states and the influence of intrinsic and extrinsic factors on macrophage metabolic control. We also shed light on certain open questions in the field and how answering these might pave the way for tissue-specific therapeutic approaches.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
5
|
Saiz JC, Martín-Acebes MA, Blázquez AB, Escribano-Romero E, Poderoso T, Jiménez de Oya N. Pathogenicity and virulence of West Nile virus revisited eight decades after its first isolation. Virulence 2021; 12:1145-1173. [PMID: 33843445 PMCID: PMC8043182 DOI: 10.1080/21505594.2021.1908740] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
West Nile virus (WNV) is a flavivirus which transmission cycle is maintained between mosquitoes and birds, although it occasionally causes sporadic outbreaks in horses and humans that can result in serious diseases and even death. Since its first isolation in Africa in 1937, WNV had been considered a neglected pathogen until its recent spread throughout Europe and the colonization of America, regions where it continues to cause outbreaks with severe neurological consequences in humans and horses. Although our knowledge about the characteristics and consequences of the virus has increased enormously lately, many questions remain to be resolved. Here, we thoroughly update our knowledge of different aspects of the WNV life cycle: virology and molecular classification, host cell interactions, transmission dynamics, host range, epidemiology and surveillance, immune response, clinical presentations, pathogenesis, diagnosis, prophylaxis (antivirals and vaccines), and prevention, and we highlight those aspects that are still unknown and that undoubtedly require further investigation.
Collapse
Affiliation(s)
- Juan-Carlos Saiz
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Miguel A Martín-Acebes
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Ana B Blázquez
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Estela Escribano-Romero
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| | - Teresa Poderoso
- Molecular Virology Group, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nereida Jiménez de Oya
- Department of Biotechnology, National Institute for Agricultural and Food Research and Technology (INIA), Madrid, Spain
| |
Collapse
|
6
|
Immune outcomes of Zika virus infection in nonhuman primates. Sci Rep 2020; 10:13069. [PMID: 32747639 PMCID: PMC7400481 DOI: 10.1038/s41598-020-69978-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/21/2020] [Indexed: 11/23/2022] Open
Abstract
Although the Zika virus (ZIKV) epidemic is subsiding, immune responses that are important for controlling acute infection have not been definitively characterized. Nonhuman primate (NHP) models were rapidly developed to understand the disease and to test vaccines, and these models have since provided an understanding of the immune responses that correlate with protection during natural infection and vaccination. Here, we infected a small group of male rhesus (Macaca mulatta) and cynomolgus (Macaca fascicularis) macaques with a minimally passaged Brazilian ZIKV isolate and used multicolor flow cytometry and transcriptional profiling to describe early immune patterns following infection. We found evidence of strong innate antiviral responses together with induction of neutralizing antibodies and T cell responses. We also assessed the relative importance of CD8 T cells in controlling infection by carrying out CD8 T cell depletion in an additional two animals of each species. CD8 depletion appeared to dysregulate early antiviral responses and possibly increase viral persistence, but the absence of CD8 T cells ultimately did not impair control of the virus. Together, these data describe immunological trends in two NHP species during acute ZIKV infection, providing an account of early responses that may be important in controlling infection.
Collapse
|
7
|
Tsetsarkin KA, Acklin JA, Liu G, Kenney H, Teterina NL, Pletnev AG, Lim JK. Zika virus tropism during early infection of the testicular interstitium and its role in viral pathogenesis in the testes. PLoS Pathog 2020; 16:e1008601. [PMID: 32614902 PMCID: PMC7331987 DOI: 10.1371/journal.ppat.1008601] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/05/2020] [Indexed: 12/18/2022] Open
Abstract
Sexual transmission and persistence of Zika virus (ZIKV) in the testes pose new challenges for controlling virus outbreaks and developing live-attenuated vaccines. It has been shown that testicular infection of ZIKV is initiated in the testicular interstitium, followed by spread of the virus in the seminiferous tubules. This leads to testicular damage and/or viral dissemination into the epididymis and eventually into semen. However, it remains unknown which cell types are targeted by ZIKV in the testicular interstitium, and what is the specific order of infectious events leading to ZIKV invasion of the seminiferous tubules. Here, we demonstrate that interstitial leukocytes expressing mir-511-3p microRNA are the initial targets of ZIKV in the testes, and infection of mir-511-3p-expressing cells in the testicular interstitium is necessary for downstream infection of the seminiferous tubules. Mir-511-3p is expressed concurrently with CD206, a marker of lineage 2 (M2) macrophages and monocyte derived dendritic cells (moDCs). Selective restriction of ZIKV infection of CD206-expressing M2 macrophages/moDCs results in the attenuation of macrophage-associated inflammatory responses in vivo and prevents the disruption of the Sertoli cell barrier in vitro. Finally, we show that targeting of viral genome for mir-511-3p significantly attenuates early ZIKV replication not only in the testes, but also in many peripheral organs, including spleen, epididymis, and pancreas. This incriminates M2 macrophages/moDCs as important targets for visceral ZIKV replication following hematogenous dissemination of the virus from the site of infection.
Collapse
Affiliation(s)
- Konstantin A. Tsetsarkin
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Joshua A. Acklin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Guangping Liu
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Heather Kenney
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Natalia L. Teterina
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Alexander G. Pletnev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Jean K. Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
8
|
Carpentier KS, Davenport BJ, Haist KC, McCarthy MK, May NA, Robison A, Ruckert C, Ebel GD, Morrison TE. Discrete viral E2 lysine residues and scavenger receptor MARCO are required for clearance of circulating alphaviruses. eLife 2019; 8:e49163. [PMID: 31596239 PMCID: PMC6839921 DOI: 10.7554/elife.49163] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
The magnitude and duration of vertebrate viremia is a critical determinant of arbovirus transmission, geographic spread, and disease severity. We find that multiple alphaviruses, including chikungunya (CHIKV), Ross River (RRV), and o'nyong 'nyong (ONNV) viruses, are cleared from the circulation of mice by liver Kupffer cells, impeding viral dissemination. Clearance from the circulation was independent of natural antibodies or complement factor C3, and instead relied on scavenger receptor SR-A6 (MARCO). Remarkably, lysine to arginine substitutions at distinct residues within the E2 glycoproteins of CHIKV and ONNV (E2 K200R) as well as RRV (E2 K251R) allowed for escape from clearance and enhanced viremia and dissemination. Mutational analysis revealed that viral clearance from the circulation is strictly dependent on the presence of lysine at these positions. These findings reveal a previously unrecognized innate immune pathway that controls alphavirus viremia and dissemination in vertebrate hosts, ultimately influencing disease severity and likely transmission efficiency.
Collapse
Affiliation(s)
- Kathryn S Carpentier
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Bennett J Davenport
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Kelsey C Haist
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Mary K McCarthy
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Nicholas A May
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Alexis Robison
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Claudia Ruckert
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Gregory D Ebel
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Thomas E Morrison
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| |
Collapse
|
9
|
RIG-I-like receptors direct inflammatory macrophage polarization against West Nile virus infection. Nat Commun 2019; 10:3649. [PMID: 31409781 PMCID: PMC6692387 DOI: 10.1038/s41467-019-11250-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 06/17/2019] [Indexed: 01/20/2023] Open
Abstract
RIG-I-Like Receptors (RLRs) RIG-I, MDA5, and LGP2, are vital pathogen recognition receptors in the defense against RNA viruses. West Nile Virus (WNV) infections continue to grow in the US. Here, we use a systems biology approach to define the contributions of each RLR in the innate immune response to WNV. Genome-wide RNAseq and bioinformatics analyses of macrophages from mice lacking either RLR reveal that the RLRs drive distinct immune gene activation and response polarization to mediate an M1/inflammatory signature while suppressing the M2/wound healing phenotype. While LGP2 functions to modulate inflammatory signaling, RIG-I and MDA5 together are essential for M1 macrophage polarization in vivo and the control of WNV infection through potential downstream control of ATF4 and SMAD4 to regulate target gene expression for cell polarization. These analyses reveal the RLR-driven signature of macrophage polarization, innate immune protection, and immune programming against WNV infection.
Collapse
|
10
|
Zhu G, Gui Z. Effect of silkworm peptide on inducting M1 type polarization and Th1 activation via TLR2-induced MyD88-dependent pathway. Food Sci Nutr 2019; 7:1251-1260. [PMID: 31024698 PMCID: PMC6475741 DOI: 10.1002/fsn3.954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/04/2018] [Accepted: 12/16/2018] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to explore immune activity and molecular mechanism of silkworm peptide. The cell subsets induced by silkworm peptides were detected by flow cytometry. The IFN-γ and IL-4 levels in CD4+ cells were measured by ELISA. TLR2 mRNA expression in mouse CD4+ T cells was detected by qRT-PCR. Western blot was used to detect the protein expression levels of MyD88 and p-IκB. The growth rate of Lewis lung cancer xenografts in mice of the medium-dose group was significantly reduced, and the tumor volume was significantly smaller than that of the control group on the 14th day. The relative vitality values of spleen lymphocytes in the medium-dose and high-dose groups were higher than the control group. The IFN-γ levels in the medium-dose and high-dose groups were significantly higher than the control group. The levels of IL-4 were no significant change among different groups. Compared with the control group, different doses of silkworm peptide groups could increase the levels of NO, IL-6, IL-12, and IL-1β. Compared with the control group, the protein expression levels of MyD88 and p-IκB in 10 μg/ml group and 20 μg/ml groups were significantly increased compared with the control group. Silkworm peptide could induce Th1 activation and M1 type polarization, which was dose-dependent and was relative to the effect of silkworm peptide on inhibiting tumor growth. Silkworm peptide could directly induce M1 type polarization and Th1 activation via TLR2-induced MyD88-dependent pathway in vitro.
Collapse
Affiliation(s)
- Guanglai Zhu
- School of BiotechnologyJiangsu University of Science and TechnologyZhenjiangChina
- Department of Aquatic Science and TechnologyJiangsu Animal Husbandry and Veterinary CollegeTaizhouChina
| | - Zhongzheng Gui
- School of BiotechnologyJiangsu University of Science and TechnologyZhenjiangChina
- Sericultural Research InstituteChinese Academy of Agricultural SciencesZhenjiangChina
| |
Collapse
|
11
|
A case of West Nile virus encephalitis accompanied by diabetic ketoacidosis and rhabdomyolysis. IDCases 2019; 15:e00505. [PMID: 30815360 PMCID: PMC6378900 DOI: 10.1016/j.idcr.2019.e00505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 11/22/2022] Open
Abstract
Introduction We present here a case of West Nile Virus (WNV) encephalitis that initially presented with diabetic ketoacidosis and rhabdomyolysis. Case presentation A 35-year-old male with no past medical history presented to the emergency department complaining of polydipsia, generalized weakness, lightheadedness, and visual disturbances of one week duration. He was found to be in diabetic ketoacidosis. His hemoglobin A1c was 11%. The patient was appropriately treated for diabetic ketoacidosis and it resolved on hospital day 1. On hospital day 2, the patient developed a fever of 101.6 °F and his mental status became severely altered. He developed auditory and visual hallucinations. IgM and IgG antibodies to West Nile Virus were positive in the cerebral spinal fluid (CSF). The patient's creatine kinase level rose to 118,400 U/L during his hospitalization and eventually returned to baseline. The patient made a full recovery with no residual neurologic deficits after an 11 day hospital course. Discussion In this patient, neuroinvasive WNV was confirmed with positive CSF IgM. The patient's newly diagnosed diabetes likely contributed to his susceptibility to neuroinvasive disease. Furthermore, WNV encephalitis in a background of DKA has not been previously described in the literature and this case demonstrates WNV neuroinvasive disease should be in the differential diagnosis for patients presenting with unexplained neurological symptoms. Conclusion Diagnosis of neuroinvasive WNV is imperative to stop unnecessary therapies, limit further diagnostic evaluation, help predict patient outcomes, direct public health prevention measures, and further provide investigations into the clinical conditions that define the spectrum of WNV disease.
Collapse
|
12
|
Garcia M, Alout H, Diop F, Damour A, Bengue M, Weill M, Missé D, Lévêque N, Bodet C. Innate Immune Response of Primary Human Keratinocytes to West Nile Virus Infection and Its Modulation by Mosquito Saliva. Front Cell Infect Microbiol 2018; 8:387. [PMID: 30450338 PMCID: PMC6224356 DOI: 10.3389/fcimb.2018.00387] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/12/2018] [Indexed: 12/13/2022] Open
Abstract
West Nile Virus (WNV) is a flavivirus involved in many human infections worldwide. This arthropod-borne virus is directly co-inoculated with mosquito saliva through the epidermis and the dermis during blood meal. WNV starts replicating in the skin before migrating to the draining lymph node, leading to widespread viremia and in some cases to neurological symptoms. Skin is a complex organ composed of different cell types that together perform essential functions such as pathogen sensing, barrier maintenance and immunity. Keratinocytes, which represent 90% of the cells of the epidermis, are the organism's first line of defense, initiating innate immune response by recognizing pathogens through their pattern recognition receptors. Although WNV was previously known to replicate in human primary keratinocytes, the induced inflammatory response remains unknown. The aim of this study was first to characterize the inflammatory response of human primary keratinocytes to WNV infection and then, to assess the potential role of co-inoculated mosquito saliva on the keratinocyte immune response and viral replication. A type I and III interferon inflammatory response associated with an increase of IRF7 but not IRF3 mRNA expression, and dependent on infectious dose, was observed during keratinocyte infection with WNV. Expression of several interferon-stimulated gene mRNA was also increased at 24 h post-infection (p.i.); they included CXCL10 and interferon-induced proteins with tetratricopeptide repeats (IFIT)-2 sustained up until 48 h p.i. Moreover, WNV infection of keratinocyte resulted in a significant increase of pro-inflammatory cytokines (TNFα, IL-6) and various chemokines (CXCL1, CXCL2, CXCL8 and CCL20) expression. The addition of Aedes aegypti or Culex quinquefasciatus mosquito saliva, two vectors of WNV infection, to infected keratinocytes led to a decrease of inflammatory response at 24 h p.i. However, only Ae. Aegypti saliva adjunction induced modulation of viral replication. In conclusion, this work describes for the first time the inflammatory response of human primary keratinocytes to WNV infection and its modulation in presence of vector mosquito saliva. The effects of mosquito saliva assessed in this work could be involved in the early steps of WNV replication in skin promoting viral spread through the body.
Collapse
Affiliation(s)
- Magali Garcia
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France.,Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, EA 4331, Université de Poitiers, Poitiers, France
| | - Haoues Alout
- Institut des Sciences de l'Evolution, Université de Montpellier, Montpellier, France
| | - Fodé Diop
- MIVEGEC UMR 224, Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Alexia Damour
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, EA 4331, Université de Poitiers, Poitiers, France
| | - Michèle Bengue
- MIVEGEC UMR 224, Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Mylène Weill
- Institut des Sciences de l'Evolution, Université de Montpellier, Montpellier, France
| | - Dorothée Missé
- MIVEGEC UMR 224, Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Nicolas Lévêque
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France.,Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, EA 4331, Université de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, EA 4331, Université de Poitiers, Poitiers, France
| |
Collapse
|