1
|
Tomatis C, Charo N, Ferrer MF, Aquila S, Fuentes F, Scalise ML, Aguiar MCAM, Valdivieso G, Carrera Silva EA, Gómez RM. Yellow fever virus infection alters mitochondrial network dynamics and trigger IFN-I response via TLR2 pathway. Int Immunopharmacol 2025; 157:114699. [PMID: 40300356 DOI: 10.1016/j.intimp.2025.114699] [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: 10/09/2024] [Revised: 03/24/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025]
Abstract
It has been emphasized that mitochondria play a fundamental role not only in cellular bioenergetics but also in the defense against infections. Here, we investigated mitochondrial network dynamics (MND) and IFN-Iβ signaling response in epithelial A549 cells after yellow fever virus (YFV) infection. We analyze the MND when only some cells are infected at 1 day post-infection (dpi) and after the spread of viral infection, at 3 dpi. Confocal microscopy and MiNA analysis showed that YFV infection leads to a decrease in the number of branches at 3 dpi and an increase in the length of branches at 1 and 3 dpi, suggesting that mitochondrial fission and fusion occur. Consistent with both processes, we found increased transcription of mitofusin 1 and Drp1 and increased colocalization of mitochondria with Drp1 at 3 dpi. In addition, mitochondrial membrane polarization decreased, mtROS production increased, p62 expression decreased, and LC3 expression increased, suggesting an increase in mitophagy flux. We found decreased expression of the IFN inducers RIG-I and MAVS sensors in YFV-infected A549 cells a t 3 dpi. Surprisingly, increased IFN-Iβ levels were observed at transcriptional and protein levels along with IRF7 induction at 1 and 3 dpi. Using the blocking antibody against TLR2, we showed that IFN-Iβ and IL-6 synthesis is maintained by TLR2 signaling. Mechanistically, infection led to activation of the NFκB pathway by degradation of IkBα, and increased phosphorylation of P65 and ERK MAPK signaling. Our results show that YFV infection induces altered MND in epithelial cells and triggers TLR2 signaling.
Collapse
Affiliation(s)
- Carla Tomatis
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular, CONICET-UNLP, La Plata, Argentina; Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| | - Nancy Charo
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| | - María F Ferrer
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular, CONICET-UNLP, La Plata, Argentina
| | - Silvia Aquila
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular, CONICET-UNLP, La Plata, Argentina
| | - Federico Fuentes
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| | - María L Scalise
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular, CONICET-UNLP, La Plata, Argentina
| | - Mara C A M Aguiar
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular, CONICET-UNLP, La Plata, Argentina; Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| | - Gabriel Valdivieso
- Laboratorio de Biología Celular y Molecular, Instituto de Investigación Médica CONICET-UCA, Buenos Aires, Argentina
| | - Eugenio A Carrera Silva
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina.
| | - Ricardo M Gómez
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular, CONICET-UNLP, La Plata, Argentina.
| |
Collapse
|
2
|
Monyama MC, Molefe LR, Meddows-Taylor S. A review of the mosquito-borne flaviviruses: Dengue virus and West Nile virus in Southern Africa. Virusdisease 2025; 36:1-11. [PMID: 40290767 PMCID: PMC12022202 DOI: 10.1007/s13337-025-00917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/02/2025] [Indexed: 04/30/2025] Open
Abstract
Dengue virus (DENV) and West Nile (WNV) viruses are important re-emerging mosquito-borne members of the genus Flavivirus that are under-recognized in many parts of Africa. This review aims to evaluate the existing literature on the transmission, epidemiology, diagnostic techniques, clinical presentation and prevention of infection with DENV and WNV in Southern Africa. Literature shows that both DENV and WNV are transmitted by mosquitoes of Aedes spp. and Culex species., respectively, and both viruses are widespread in the Southern African region. Epidemiologically, sporadic outbreaks have been reported of both DENV and WNV in various Southern African countries, indicating the ongoing threat of these viruses. However, the lack of comprehensive surveillance and diagnostic capacity challenges accurate estimation of their true prevalence. Diagnostic techniques for DENV and WNV involve serological tests, molecular tests and viral isolation, enabling prompt diagnosis and differentiation from other febrile illnesses. In Southern Africa, infection with DENV and WNV presents significant public health concerns, with the clinical presentation of both infections ranging from asymptomatic cases to severe manifestations. Symptoms of infection include high fever, myalgia, rash, and, in severe cases, haemorrhagic fever for DENV and neurological complications for WNV. No specific antiviral treatment exists for either virus, underscoring the importance of supportive care and symptom management. To prevent the spread of DENV and WNV in Southern African countries, a combination of prevention and treatment strategies should be employed, including effective mosquito control, continuous monitoring of vector population dynamics, public health education, and surveillance and reporting systems for averting future outbreaks.
Collapse
Affiliation(s)
- Maropeng C. Monyama
- Department of Life and Consumer Sciences, University of South Africa, Private Bag X6, Florida, Johannesburg 1710 South Africa
| | - Letlhogonolo R. Molefe
- Department of Life and Consumer Sciences, University of South Africa, Private Bag X6, Florida, Johannesburg 1710 South Africa
| | - Stephen Meddows-Taylor
- Department of Life and Consumer Sciences, University of South Africa, Private Bag X6, Florida, Johannesburg 1710 South Africa
| |
Collapse
|
3
|
Stegeman SK, Kourko O, Amsden H, Pellizzari Delano IE, Mamatis JE, Roth M, Colpitts CC, Gee K. RNA Viruses, Toll-Like Receptors, and Cytokines: The Perfect Storm? J Innate Immun 2025; 17:126-153. [PMID: 39820070 PMCID: PMC11845175 DOI: 10.1159/000543608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs. BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs.
Collapse
Affiliation(s)
- Sophia K Stegeman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Olena Kourko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Heather Amsden
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - John E Mamatis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Madison Roth
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
4
|
Guo T, Xiong L, Xie J, Zeng J, Huang Z, Yao M, Zhang X, Mo J. TLR2 promotes traumatic deep venous thrombosis of the lower extremity following femoral fracture by activating the NF‑κB/COX‑2 signaling pathway in rats. Exp Ther Med 2024; 28:436. [PMID: 39355523 PMCID: PMC11443593 DOI: 10.3892/etm.2024.12725] [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: 11/24/2023] [Accepted: 06/28/2024] [Indexed: 10/03/2024] Open
Abstract
Endothelial cells (ECs) are crucial for maintaining the integrity of blood vessel walls and reducing thrombosis. Deep venous thrombosis (DVT) is a common thrombotic disease and its diagnosis and treatment remain at the stage of coagulation function examination and post-onset treatment. Thus, identifying the pathogenesis of DVT is important. The present study investigated the significance of the Toll-like receptor 2 (TLR2)/nuclear factor kappa B (NF-κB)/cyclooxygenase-2 (COX-2) signaling pathway in a human umbilical vein EC (HUVECs) oxygen glucose deprivation (OGD) model and femoral fractures were induced in anesthetized rats using a quantifiable impact device delivering 5 J of energy to each side of the proximal outer thigh, followed by external fixation with a hip spica cast to create a traumatic deep venous thrombosis (TDVT) animal model. Rats were subjected to quantitative impact fixation to establish a TDVT model. The rats were treated with a TLR2 agonist (Pam3CSK4) and a TLR2 inhibitor (C29) via intraperitoneal injection and thrombus formation was examined. HUVECs were subjected to OGD and treated with Pam3CSK4 or C29 and cell viability and apoptosis were assessed. Western blotting, immunofluorescence and reverse transcription-quantitative PCR were used to examine the inflammatory responses and signaling pathways. In vivo experiments showed that Pam3CSK4 promoted thrombus formation and increased the mRNA and protein expression of NF-κB, COX-2, Tissue factor (TF), IL-6 and P-selectin compared with the model and C29 groups. In vitro experiments showed that Pam3CSK4 treatment resulted in a higher number of apoptotic cells than C29 treatment and that it increased the levels of NF-κB, COX-2, IL-6 and P-selectin, whereas C29 decreased them. Thus, TLR2 promotes the inflammatory response in EC through the NF-κB/COX-2 signaling pathway, which may lead to EC apoptosis and the occurrence of TDVT.
Collapse
Affiliation(s)
- Tianting Guo
- Department of Orthopedics, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi 341000, P.R. China
| | - Lijiao Xiong
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, Guangdong 518055, P.R. China
- Department of Geriatrics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Junbin Xie
- Department of Orthopedics, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi 341000, P.R. China
| | - Jiwei Zeng
- Department of Orthopedics, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi 341000, P.R. China
| | - Zhihua Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Mengting Yao
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Xiaoan Zhang
- Department of Orthopedics, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi 341000, P.R. China
| | - Jianwen Mo
- Department of Geriatrics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
5
|
Dash MK, Samal S, Rout S, Behera CK, Sahu MC, Das B. Immunomodulation in dengue: towards deciphering dengue severity markers. Cell Commun Signal 2024; 22:451. [PMID: 39327552 PMCID: PMC11425918 DOI: 10.1186/s12964-024-01779-4] [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: 05/06/2024] [Accepted: 08/06/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Dengue is a vector-borne debilitating disease that is manifested as mild dengue fever, dengue with warning signs, and severe dengue. Dengue infection provokes a collective immune response; in particular, the innate immune response plays a key role in primary infection and adaptive immunity during secondary infection. In this review, we comprehensively walk through the various markers of immune response against dengue pathogenesis and outcome. MAIN BODY Innate immune response against dengue involves a collective response through the expression of proinflammatory cytokines, such as tumor necrosis factors (TNFs), interferons (IFNs), and interleukins (ILs), in addition to anti-inflammatory cytokines and toll-like receptors (TLRs) in modulating viral pathogenesis. Monocytes, dendritic cells (DCs), and mast cells are the primary innate immune cells initially infected by DENV. Such immune cells modulate the expression of various markers, which can influence disease severity by aiding virus entry and proinflammatory responses. Adaptive immune response is mainly aided by B and T lymphocytes, which stimulate the formation of germinal centers for plasmablast development and antibody production. Such antibodies are serotype-dependent and can aid in virus entry during secondary infection, mediated through a different serotype, such as in antibody-dependent enhancement (ADE), leading to DENV severity. The entire immunological repertoire is exhibited differently depending on the immune status of the individual. SHORT CONCLUSION Dengue fever through severe dengue proceeds along with the modulated expression of several immune markers. In particular, TLR2, TNF-α, IFN-I, IL-6, IL-8, IL-17 and IL-10, in addition to intermediate monocytes (CD14+CD16+) and Th17 (CD4+IL-17+) cells are highly expressed during severe dengue. Such markers could assist greatly in severity assessment, prompt diagnosis, and treatment.
Collapse
Affiliation(s)
- Manoj Kumar Dash
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | - Sagnika Samal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | - Shailesh Rout
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | - Chinmay Kumar Behera
- Department of Pediatrics, Kalinga Institute of Medical Sciences, Deemed to Be University, Bhubaneswar, Odisha, 751024, India
| | | | - Biswadeep Das
- School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to Be University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
6
|
Kang N, Chawla A, Hillman H, Tippalagama R, Kim C, Mikulski Z, Seumois G, Vijayanand P, Scriba TJ, De Silva AD, Balmaseda A, Harris E, Weiskopf D, Sette A, Arlehamn CL, Peters B, Burel JG. A novel method for characterizing cell-cell interactions at single-cell resolution reveals unique signatures in blood T cell-monocyte complexes during infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.20.612103. [PMID: 39386643 PMCID: PMC11463634 DOI: 10.1101/2024.09.20.612103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Communication between immune cells through direct contact is a critical feature of immune responses. Here, we developed a novel high-throughput method to study the transcriptome and adaptive immune receptor repertoire of single cells forming complexes without needing bioinformatic deconvolution. We found that T cells and monocytes forming complexes in blood during active tuberculosis (TB) and dengue hold unique transcriptomic signatures indicative of TCR/MCH-II immune synapses. Additionally, T cells in complexes showed enrichment for effector phenotypes, imaging and transcriptomic features of active TCR signaling, and increased immune activity at diagnosis compared to after anti-TB therapy. We also found evidence for bidirectional RNA exchange between T cells and monocytes, since complexes were markedly enriched for "dual-expressing" cells (i.e., co-expressing T cell and monocyte genes). Thus, studying immune cell complexes at a single-cell resolution offers novel perspectives on immune synaptic interactions occurring in blood during infection.
Collapse
Affiliation(s)
- Ningxin Kang
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| | - Ashu Chawla
- Bioinformatics Core, La Jolla Institute for Immunology, CA 92037, United States
| | - Hannah Hillman
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| | - Rashmi Tippalagama
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| | - Cheryl Kim
- Flow Cytometry Core, La Jolla Institute for Immunology, CA 92037, United States
| | - Zbigniew Mikulski
- Microscopy Core, La Jolla Institute for Immunology, CA 92037, United States
| | - Grégory Seumois
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, CA, United States
| | - Pandurangan Vijayanand
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Aruna D De Silva
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Faculty of Medicine, General Sir John Kotelawala Defence University, Sri Lanka
| | | | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA 94720-3370, USA
| | - Daniela Weiskopf
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | | | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Julie G Burel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| |
Collapse
|
7
|
Lim SJ, Gan SC, Ong HT, Ngeow YF. In vitro analysis of VEGF-mediated endothelial permeability and the potential therapeutic role of Anti-VEGF in severe dengue. Biochem Biophys Rep 2024; 39:101814. [PMID: 39263317 PMCID: PMC11387214 DOI: 10.1016/j.bbrep.2024.101814] [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: 06/12/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Background Vascular endothelial growth factor (VEGF) is one of the proteins involved in dengue immunopathogenesis. It is overexpressed in severe dengue and contributes to vascular permeability and plasma leakage. In this study, we investigated the effects of VEGF and anti-VEGF treatments on endothelial cells in vitro, to assess the potential use of anti-VEGF antibodies in managing severe dengue. Methods Human pulmonary microvascular endothelial cells were treated with VEGF and a VEGF/anti-VEGF combination. The effects of the treatments were studied using an endothelial permeability assay and microarray gene expression profiling. In the permeability assay, the fluorescein isothiocyanate (FITC)-dextran fluorescence signal across the endothelial monolayer was recorded, and the cells were stained with PECAM-1 to detect gap formation. RNA was extracted from treated cells for microarray gene profiling and analysis. The results were analyzed for differentially expressed genes (DEGs) and gene enrichment analysis. The DEGs were subjected to STRING to construct the protein-protein interaction network and then Cytoscape to identify the hub genes. Results VEGF-treated endothelial cells showed greater movement of FITC-dextran across the monolayer than VEGF/anti-VEGF-treated cells. There were 111 DEGs for VEGF-treated cells and 118 DEGs for VEGF/anti-VEGF-treated cells. The genes upregulated in VEGF-treated cells were enriched in inflammatory responses and regulation of the endothelial barrier, nitric oxide synthesis, angiogenesis, and the nucleotide-binding oligomerization domain-like receptor signaling pathway. Top 10 hub genes were identified from the DEGs. Conclusions VEGF treatment increased permeability across endothelial cells, while anti-VEGF reduced this leakage. Analysis of VEGF-treated endothelial cells identified hub genes implicated in severe dengue. The top 10 hub genes were TNF, IL1B, IL6, CCL2, PTGS2, ICAM1, CXCL2, CXCL1, CSF2, and TLR2. The results of this study show that using anti-VEGF antibodies to neutralize VEGF may be a promising therapy to prevent the progression of dengue to severe dengue.
Collapse
Affiliation(s)
- Sheng Jye Lim
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| | - Seng Chiew Gan
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| | - Hooi Tin Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
- Center for Cancer Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| | - Yun Fong Ngeow
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
- Centre for Research on Communicable Diseases, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras 43000, Kajang, Selangor, Malaysia
| |
Collapse
|
8
|
Malavige GN, Ogg GS. Molecular mechanisms in the pathogenesis of dengue infections. Trends Mol Med 2024; 30:484-498. [PMID: 38582622 DOI: 10.1016/j.molmed.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024]
Abstract
Dengue is the most rapidly emerging climate-sensitive infection, and morbidity/mortality and disease incidence are rising markedly, leading to healthcare systems being overwhelmed. There are currently no specific treatments for dengue or prognostic markers to identify those who will progress to severe disease. Owing to an increase in the burden of illness and a change in epidemiology, many patients experience severe disease. Our limited understanding of the complex mechanisms of disease pathogenesis has significantly hampered the development of safe and effective treatments, vaccines, and biomarkers. We discuss the molecular mechanisms of dengue pathogenesis, the gaps in our knowledge, and recent advances, as well as the most crucial questions to be answered to enable the development of therapeutics, biomarkers, and vaccines.
Collapse
Affiliation(s)
- Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka; Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| | - Graham S Ogg
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka; Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Hernández-Sarmiento LJ, Tamayo-Molina YS, Valdés-López JF, Urcuqui-Inchima S. Mayaro virus infection elicits a robust pro-inflammatory and antiviral response in human macrophages. Acta Trop 2024; 252:107146. [PMID: 38342287 DOI: 10.1016/j.actatropica.2024.107146] [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: 09/04/2023] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Mayaro virus (MAYV), the etiological agent of Mayaro fever (MAYF), is an emergent arbovirus pathogen belonging to Togaviridae family. MAYF is characterized by high inflammatory component that can cause long-lasting arthralgia that persists for months. Macrophages are viral targets and reservoirs, key components of innate immunity and host response. Given the importance of this pathogen, our aim was to determine the inflammatory and antiviral response of human monocyte-derived macrophages (MDMs) infected with MAYV. First, we established the replication kinetics of the virus. Thereafter, we determined the expression of pattern recognition receptors, NF-ĸB complex, interferons (IFNs), two interleukin 27 (IL27) subunits, IFN-stimulated genes (ISGs), and the production of cytokines/chemokines. We found that human MDMs are susceptible to MAYV infection in vitro, with a peak of viral particles released between 24- and 48-hours post-infection (h.p.i) at MOI 0.5, and between 12 and 24 h.p.i at MOI 1. Interestingly, we observed a significant decline in the production of infectious viral particles at 72 h.p.i that was associated with the induction of antiviral response and high cytotoxic effect of MAYV infection in MDMs. We observed modulation of several genes after MAYV infection, as well, we noted the activation of antiviral detection and response pathways (Toll-like receptors, RIG-I/MDA5, and PKR) at 48 h.p.i but not at 6 h.p.i. Furthermore, MAYV-infected macrophages express high levels of the three types of IFNs and the two IL27 subunits at 48 h.p.i. Moreover, we found higher production of IL6, IL1β, CXCL8/IL8, CCL2, and CCL5 at 48 h.p.i as compared to 6 h.p.i. A robust antiviral response (ISG15, APOBEC3A, IFITM1, and MX2) was observed at 48 but not at 6 h.p.i. The innate and antiviral responses of MAYV-infected MDMs differ at 6 and 48 h.p.i. We conclude that MAYV infection induces robust pro-inflammatory and antiviral responses in human primary macrophages.
Collapse
Affiliation(s)
| | - Y S Tamayo-Molina
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
10
|
Huang H, He X, Shi L, Yu J, Lu Z, Cao H, Ou J, Chen X, Yan L, Yang J, Zhao W, Liu J, Yu L. Tanreqing injection inhibits dengue virus encephalitis by suppressing the activation of NLRP3 inflammasome. Chin Med 2024; 19:24. [PMID: 38355571 PMCID: PMC10868054 DOI: 10.1186/s13020-024-00893-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Encephalitis caused by dengue virus (DENV) is considered a manifestation of severe dengue. Tanreqing injection (TRQ) is a well-known Chinese patented medicine, which has been used to treat brain-related disorders by inhibiting inflammation. Nevertheless, the effects of TRQ on DENV encephalitis have not been studied. The aim of this study was to evaluate the effects of TRQ on DENV encephalitis and to explore its potential mechanisms. METHODS The cytotoxicity of TRQ was examined by MTT assay, and the anti-DENV activities of TRQ in BHK-21 baby hamster kidney fibroblast were evaluated through CCK-8 and plaque assays. The expression levels of NO, IL1B/IL-1β, TNFα and IL6 were measured by qRT‒PCR and ELISA in the BV2 murine microglial cell line. The inhibitory effects of TRQ on NLRP3 inflammasome activation in BV2 cells were examined by Western blotting, qRT‒PCR and ELISA. The effects of TRQ on HT22 mouse hippocampal neuronal cells were examined by CCK-8 assay, morphology observation and flow cytometry. Moreover, a DENV-infected ICR suckling mouse model was developed to investigate the protective role of TRQ in vivo. RESULTS TRQ decreased the release of NO, IL6, TNFα and IL1B from BV2 cells and inhibited the activation of NLRP3. The presence of the NLRP3 agonist nigericin reversed the anti-inflammatory activities of TRQ. Furthermore, TRQ inhibited the death of HT22 cells by decreasing IL1B in DENV-infected BV2 cells. In addition, TRQ significantly attenuated weight loss, reduced clinical scores and extended the survival in DENV-infected ICR suckling mice. Critically, TRQ ameliorated pathological changes in ICR suckling mice brain by inhibiting microglia and NLRP3 activation and decreasing the production of inflammatory factors and the number of dead neurons. CONCLUSION TRQ exerts potent inhibitory effects on dengue encephalitis in vitro and in vivo by reducing DENV-2-induced microglial activation and subsequently decreasing the inflammatory response, thereby protecting neurons. These findings demonstrate the potential of TRQ in the treatment of dengue encephalitis.
Collapse
Affiliation(s)
- Hefei Huang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xuemei He
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Lingzhu Shi
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Huihui Cao
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jinying Ou
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xi Chen
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Lijun Yan
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jiabin Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
11
|
Kurosu T, Okuzaki D, Sakai Y, Kadi MA, Phanthanawiboon S, Ami Y, Shimojima M, Yoshikawa T, Fukushi S, Nagata N, Suzuki T, Kamimura D, Murakami M, Ebihara H, Saijo M. Dengue virus infection induces selective expansion of Vγ4 and Vγ6TCR γδ T cells in the small intestine and a cytokine storm driving vascular leakage in mice. PLoS Negl Trop Dis 2023; 17:e0011743. [PMID: 37939119 PMCID: PMC10659169 DOI: 10.1371/journal.pntd.0011743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/20/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
Dengue is a major health problem in tropical and subtropical regions. Some patients develop a severe form of dengue, called dengue hemorrhagic fever, which can be fatal. Severe dengue is associated with a transient increase in vascular permeability. A cytokine storm is thought to be the cause of the vascular leakage. Although there are various research reports on the pathogenic mechanism, the complete pathological process remains poorly understood. We previously reported that dengue virus (DENV) type 3 P12/08 strain caused a lethal systemic infection and severe vascular leakage in interferon (IFN)-α/β and γ receptor knockout mice (IFN-α/β/γRKO mice), and that blockade of TNF-α signaling protected mice. Here, we performed transcriptome analysis of liver and small intestine samples collected chronologically from P12/08-infected IFN-α/β/γRKO mice in the presence/absence of blockade of TNF-α signaling and evaluated the cytokine and effector-level events. Blockade of TNF-α signaling mainly protected the small intestine but not the liver. Infection induced the selective expansion of IL-17A-producing Vγ4 and Vγ6 T cell receptor (TCR) γδ T cells in the small intestine, and IL-17A, together with TNF-α, played a critical role in the transition to severe disease via the induction of inflammatory cytokines such as TNF-α, IL-1β, and particularly the excess production of IL-6. Infection also induced the infiltration of neutrophils, as well as neutrophil collagenase/matrix metalloprotease 8 production. Blockade of IL-17A signaling reduced mortality and suppressed the expression of most of these cytokines, including TNF-α, indicating that IL-17A and TNF-α synergistically enhance cytokine expression. Blockade of IL-17A prevented nuclear translocation of NF-κB p65 in stroma-like cells and epithelial cells in the small intestine but only partially prevented recruitment of immune cells to the small intestine. This study provides an overall picture of the pathogenesis of infection in individual mice at the cytokine and effector levels.
Collapse
Affiliation(s)
- Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mohamad Al Kadi
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, Japan
| | | | - Yasusi Ami
- Management Department of Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Shimojima
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomoki Yoshikawa
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shuetsu Fukushi
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Daisuke Kamimura
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Team of Quantumimmunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology (QST), Chiba, Japan
- Division of Molecular Neuroimmunology, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
| | - Hideki Ebihara
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
12
|
Xie L, Fang Y, Chen J, Meng W, Guan Y, Gong W. TTC13 expression and STAT3 activation may form a positive feedback loop to promote ccRCC progression. PeerJ 2023; 11:e16316. [PMID: 37927783 PMCID: PMC10621595 DOI: 10.7717/peerj.16316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023] Open
Abstract
Background Renal cell carcinoma (RCC) originates from renal tubular epithelial cells and is mainly classified into three histological types, including clear cell renal cell carcinoma (ccRCC) which accounts for about 75% of all kidney cancers and is characterized by its strong invasiveness and poor prognosis. Hence, it is imperative to understand the mechanisms underlying the occurrence and progression of ccRCC to identify effective biomarkers for the early diagnosis and the prognosis prediction. Methods The mRNA level of TTC13 was quantified by RT-PCR, while the protein level was determined by western blot and immunohistochemistry (IHC) staining. Cell proliferation was measured by cck-8, and cell apoptosis was detected by flow cytometry. The binding of STAT3 to the promoter region of TTC13 was determined by the luciferase reporter assay and chip experiments. STAT3 nuclear translocation was assessed by immunofluorescence staining. Results We found that TTC13 was up-regulated in ccRCC, and TTC13 promoted cell proliferation as well as inhibited cell apoptosis and autophagy of ccRCC through wnt/β-catenin and IL6-JAK-STAT3 signaling pathways. Furthermore, TTC13 might play a role in the immune infiltration and immunotherapy of ccRCC. Mechanistically, STAT3 activated the transcription of TTC13 gene. Conclusions STAT3 directly regulated TTC13 expression through a positive feedback loop mechanism to promote ccRCC cell proliferation as well as reduce cell apoptosis and autophagy. These findings suggested new and effective therapeutic targets for more accurate and personalized treatment strategies.
Collapse
Affiliation(s)
- Lingling Xie
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu Fang
- Department of Urology, The First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, China
| | - Jianping Chen
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Wei Meng
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yangbo Guan
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wenliang Gong
- Department of Urology, The First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, China
- Department of Urology, The Third Affiliated Hospital of Naval Medical University (Shanghai Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| |
Collapse
|
13
|
Valdés-López JF, Urcuqui-Inchima S. Antiviral response and immunopathogenesis of interleukin 27 in COVID-19. Arch Virol 2023; 168:178. [PMID: 37310504 DOI: 10.1007/s00705-023-05792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/11/2023] [Indexed: 06/14/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with a high mortality rate. The clinical course is attributed to the severity of pneumonia and systemic complications. In COVID-19 patients and murine models of SARS-CoV-2 infection, the disease may be accompanied by excessive production of cytokines, leading to an accumulation of immune cells in affected organs such as lungs. Previous reports have shown that SARS-CoV-2 infection antagonizes interferon (IFN)-dependent antiviral response, thereby preventing the expression of IFN-stimulated genes (ISGs). Lower IFN levels have been linked to more-severe COVID-19. Interleukin 27 (IL27) is a heterodimeric cytokine composed of IL27p28 and EBI3 subunits, which induce both pro- and anti-inflammatory responses. Recently, we and others have reported that IL27 also induces a strong antiviral response in an IFN-independent manner. Here, we investigated transcription levels of both IL27 subunits in COVID-19 patients. The results show that SARS-CoV-2 infection modulates TLR1/2-MyD88 signaling in PBMCs and monocytes and induces NF-κB activation and expression of NF-κB-target genes that are dependent on a robust pro-inflammatory response, including EBI3; and activates IRF1 signaling which induces IL27p28 mRNA expression. The results suggest that IL27 induces a robust STAT1-dependent pro-inflammatory and antiviral response in an IFN-independent manner in COVID-derived PBMCs and monocytes as a function of a severe clinical course of COVID-19. Similar results were observed in macrophages stimulated with the SARS-CoV-2 spike protein. Thus, IL27 can trigger an antiviral response in the host, suggesting the possibility of novel therapeutics against SARS-CoV-2 infection in humans.
Collapse
Affiliation(s)
- Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
14
|
Upasani V, ter Ellen BM, Sann S, Lay S, Heng S, Laurent D, Ly S, Duong V, Dussart P, Smit JM, Cantaert T, Rodenhuis-Zybert IA. Characterization of soluble TLR2 and CD14 levels during acute dengue virus infection. Heliyon 2023; 9:e17265. [PMID: 37416678 PMCID: PMC10320027 DOI: 10.1016/j.heliyon.2023.e17265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 06/02/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Dengue virus infection results in a broad spectrum of diseases ranging from mild dengue fever (DF) to severe dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Hitherto, there is no consensus biomarker for the prediction of severe dengue disease in patients. Yet, early identification of patients who progress to severe dengue is pivotal for better clinical management. We have recently reported that an increased frequency of classical (CD14 ++CD16-) monocytes with sustained high TLR2 expression in acutely infected dengue patients correlates with severe dengue development. Here, we hypothesized that the relatively lower TLR2 and CD14 expression in mild dengue patients is due to the shedding of their soluble forms (sTLR2 and sCD14) and that these could be used as indicators of disease progression. Therefore, using commercial sandwich ELISAs, we evaluated the release of sTLR2 and sCD14 by peripheral blood mononuclear cells (PBMCs) in response to in vitro dengue virus (DENV) infection and assessed their levels in acute-phase plasma of 109 dengue patients. We show that while both sTLR2 and sCD14 are released by PBMCs in response to DENV infection in vitro, their co-circulation in an acute phase of the disease is not always apparent. In fact, sTLR2 was found only in 20% of patients irrespective of disease status. In contrast, sCD14 levels were detected in all patients and were significantly elevated in DF patients when compared to DHF patients and age-matched healthy donors. Altogether, our results suggest that sCD14 may help in identifying patients at risk of severe dengue at hospital admittance.
Collapse
Affiliation(s)
- Vinit Upasani
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Bram M. ter Ellen
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sothy Heng
- Kantha Bopha Children Hospital, Phnom Penh, Cambodia
| | - Denis Laurent
- Kantha Bopha Children Hospital, Phnom Penh, Cambodia
| | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Jolanda M. Smit
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur 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
| |
Collapse
|
15
|
Li W, Cai Z, Schindler F, Bahiraii S, Brenner M, Heiss EH, Weckwerth W. Norbergenin prevents LPS-induced inflammatory responses in macrophages through inhibiting NFκB, MAPK and STAT3 activation and blocking metabolic reprogramming. Front Immunol 2023; 14:1117638. [PMID: 37251401 PMCID: PMC10213229 DOI: 10.3389/fimmu.2023.1117638] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/28/2023] [Indexed: 05/31/2023] Open
Abstract
Inflammation is thought to be a key cause of many chronic diseases and cancer. However, current therapeutic agents to control inflammation have limited long-term use potential due to various side-effects. This study aimed to examine the preventive effects of norbergenin, a constituent of traditional anti-inflammatory recipes, on LPS-induced proinflammatory signaling in macrophages and elucidate the underlying mechanisms by integrative metabolomics and shotgun label-free quantitative proteomics platforms. Using high-resolution mass spectrometry, we identified and quantified nearly 3000 proteins across all samples in each dataset. To interpret these datasets, we exploited the differentially expressed proteins and conducted statistical analyses. Accordingly, we found that LPS-induced production of NO, IL1β, TNFα, IL6 and iNOS in macrophages was alleviated by norbergenin via suppressed activation of TLR2 mediated NFκB, MAPKs and STAT3 signaling pathways. In addition, norbergenin was capable of overcoming LPS-triggered metabolic reprogramming in macrophages and restrained the facilitated glycolysis, promoted OXPHOS, and restored the aberrant metabolites within the TCA cycle. This is linked to its modulation of metabolic enzymes to support its anti-inflammatory activity. Thus, our results uncover that norbergenin regulates inflammatory signaling cascades and metabolic reprogramming in LPS stimulated macrophages to exert its anti-inflammatory potential.
Collapse
Affiliation(s)
- Wan Li
- Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Ecology and Evolution, University of Vienna, Vienna, Austria
| | - Zhengnan Cai
- Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Ecology and Evolution, University of Vienna, Vienna, Austria
| | - Florian Schindler
- Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sports Sciences, University of Vienna, Vienna, Austria
| | - Sheyda Bahiraii
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sports Sciences, University of Vienna, Vienna, Austria
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Martin Brenner
- Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sports Sciences, University of Vienna, Vienna, Austria
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Elke H Heiss
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Wolfram Weckwerth
- Molecular Systems Biology (MOSYS), Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
- Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Teo A, Tan HD, Loy T, Chia PY, Chua CLL. Understanding antibody-dependent enhancement in dengue: Are afucosylated IgG1s a concern? PLoS Pathog 2023; 19:e1011223. [PMID: 36996026 PMCID: PMC10062565 DOI: 10.1371/journal.ppat.1011223] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Affiliation(s)
- Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Hao Dong Tan
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Thomas Loy
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Po Ying Chia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Caroline Lin Lin Chua
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor’s University, Subang Jaya, Malaysia
| |
Collapse
|
17
|
Hernández-Sarmiento LJ, Valdés-López JF, Urcuqui-Inchima S. American-Asian- and African lineages of Zika virus induce differential pro-inflammatory and Interleukin 27-dependent antiviral responses in human monocytes. Virus Res 2023; 325:199040. [PMID: 36610657 PMCID: PMC10194209 DOI: 10.1016/j.virusres.2023.199040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/06/2023]
Abstract
Zika virus (ZIKV) is an arbovirus that belongs to the Flaviviridae family and inflammatory responses play a critical role in ZIKV pathogenesis. As a first-line defense, monocytes are key components of innate immunity and host response to viruses. Monocytes are considered the earliest blood cell type to be infected by ZIKV and have been shown to be associated with ZIKV pathogenesis. The first ZIKV epidemic was reported in Africa and Asia although, it is less well known whether African- and Asian- lineages of ZIKV have different impacts on host immune response. We studied the pro-inflammatory and antiviral response of ZIKV-infected monocytes using publicly available RNA-seq analysis (GSE103114). We compared the transcriptomic profiles of human monocytes infected with ZIKV Puerto Rico strain (PRVABC59), American-Asian lineage, and ZIKV Nigeria strain (IBH30656), African lineage. We validated RNA-seq results by ELISA or RT-qPCR, in human monocytes infected with a clinical isolate of ZIKV from Colombia (American-Asian lineage), or with ZIKV from Dakar (African lineage). The transcriptomic analysis showed that ZIKV Puerto Rico strain promotes a higher pro-inflammatory response through TLR2 signaling and NF-kB activation and induces a strong IL27-dependent antiviral activity than ZIKV Nigeria strain. Furthermore, human monocytes are more susceptible to infection with ZIKV from Colombia than ZIKV from Dakar. Likewise, Colombian ZIKV isolate activated IL27 signaling and induced a robust antiviral response in an IFN-independent manner. Moreover, we show that treatment of monocytes with IL27 results in decreased release of ZIKV particles in a dose-dependent manner with an EC50 =2.870 ng/mL for ZIKV from Colombia and EC50 =10.23 ng/mL to ZIKV from Dakar. These findings highlight the differential inflammatory response and antiviral activity of monocytes infected with different lineages of ZIKV and may help better management of ZIKV-infected patients.
Collapse
Affiliation(s)
| | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
18
|
Hatton AA, Guerra FE. Scratching the Surface Takes a Toll: Immune Recognition of Viral Proteins by Surface Toll-like Receptors. Viruses 2022; 15:52. [PMID: 36680092 PMCID: PMC9863796 DOI: 10.3390/v15010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Early innate viral recognition by the host is critical for the rapid response and subsequent clearance of an infection. Innate immune cells patrol sites of infection to detect and respond to invading microorganisms including viruses. Surface Toll-like receptors (TLRs) are a group of pattern recognition receptors (PRRs) that can be activated by viruses even before the host cell becomes infected. However, the early activation of surface TLRs by viruses can lead to viral clearance by the host or promote pathogenesis. Thus, a plethora of research has attempted to identify specific viral ligands that bind to surface TLRs and mediate progression of viral infection. Herein, we will discuss the past two decades of research that have identified specific viral proteins recognized by cell surface-associated TLRs, how these viral proteins and host surface TLR interactions affect the host inflammatory response and outcome of infection, and address why controversy remains regarding host surface TLR recognition of viral proteins.
Collapse
Affiliation(s)
- Alexis A. Hatton
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT 59718, USA
| | - Fermin E. Guerra
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
19
|
Aguilar Briseño JA, Ramos Pereira L, van der Laan M, Pauzuolis M, ter Ellen BM, Upasani V, Moser J, de Souza Ferreira LC, Smit JM, Rodenhuis-Zybert IA. TLR2 axis on peripheral blood mononuclear cells regulates inflammatory responses to non-infectious immature dengue virus particles. PLoS Pathog 2022; 18:e1010499. [PMID: 36240261 PMCID: PMC9605289 DOI: 10.1371/journal.ppat.1010499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/26/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Severe dengue virus (DENV) infection is characterized by exacerbated inflammatory responses that lead to endothelial dysfunction and plasma leakage. We have recently demonstrated that Toll-like receptor 2 (TLR2) on blood monocytes senses DENV infection leading to endothelial activation. Here, we report that non-infectious immature DENV particles, which are released in large numbers by DENV-infected cells, drive endothelial activation via the TLR2 axis. We show that fully immature DENV particles induce a rapid, within 6 hours post-infection, inflammatory response in PBMCs. Furthermore, pharmacological blocking of TLR2/TLR6/CD14 and/or NF-kB prior to exposure of PBMCs to immature DENV reduces the initial production of inter alia TNF-α and IL-1β by monocytes and prevents endothelial activation. However, prolonged TLR2 block induces TNF-α production and leads to exacerbated endothelial activation, indicating that TLR2-mediated responses play an important role not only in the initiation but also the resolution of inflammation. Altogether, these data indicate that the maturation status of the virus has the potential to influence the kinetics and extent of inflammatory responses during DENV infection.
Collapse
Affiliation(s)
- José Alberto Aguilar Briseño
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Lennon Ramos Pereira
- Vaccine Development Laboratory, Microbiology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marleen van der Laan
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Mindaugas Pauzuolis
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Bram M. ter Ellen
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Vinit Upasani
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Jill Moser
- Departments of Critical Care, Pathology & Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Luís Carlos de Souza Ferreira
- Vaccine Development Laboratory, Microbiology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jolanda M. Smit
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Izabela A. Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
20
|
Wang T, Jiang F, Su J, Chu X, Cao Y, Lv H, Deng X, Wang J. A Dual-Action Molecule Suppresses S. aureus Infection as an Inhibitor Targeting Hla Pore Formation and TLR2 Signaling. Adv Biol (Weinh) 2022; 6:e2200109. [PMID: 35754300 DOI: 10.1002/adbi.202200109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/06/2022] [Indexed: 01/28/2023]
Abstract
Antibiotic resistance is the greatest challenge for the treatment of Staphylococcus aureus (S. aureus) infection under the global antibiotic resistance crisis. With the bottleneck period of the development of new antibiotics, novel alternative agents are urgently in need. In this study, the small molecule amentoflavone is identified as a dual-action inhibitor of Hla, a pore-forming virulence determinant particularly important for S. aureus pathogenicity and Toll-like receptor 2 (TLR2) signaling, which triggers inflammation response upon recognizing pathogen-associated molecular patterns. Amentoflavone treatment conferred effective protection against S. aureus pneumonia through this dual-action mechanism. Mechanically, amentoflavone effectively inhibited Hla pore formation, thereby reducing Hla-mediated cytotoxicity and tissue damage; at the same time, amentoflavone suppressed TLR2-mediated inflammatory response by blocking the interaction between TLR2 and its adapter myeloid differentiation primary response gene 88 (MyD88). Surprisingly, TLR2 signaling induced by synthetic bacterial TLR2 agonists and other heat-killed gram-positive bacteria is also blocked by amentoflavone. In summary, these results presented amentoflavone as a potential antibiotic alternative that curbed S. aureus infection by simultaneously suppressing host-damaging virulence determinants derived from bacteria and the detrimental effect of excessive inflammation derived from the host rather than bacteria viability.
Collapse
Affiliation(s)
- Tingting Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Fan Jiang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jianqing Su
- College of Agronomy, Liaocheng University, Liaocheng, 252000, China
| | - Xiuling Chu
- College of Agronomy, Liaocheng University, Liaocheng, 252000, China
| | - Yongguo Cao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Hongfa Lv
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jianfeng Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| |
Collapse
|
21
|
Zhao G, Gao Y, Zhang J, Zhang H, Xie C, Nan F, Feng S, Ha Z, Li C, Zhu X, Li Z, Zhang P, Zhang Y, Lu H, Jin N. Toll-like receptor 2 signaling pathway activation contributes to a highly efficient inflammatory response in Japanese encephalitis virus-infected mouse microglial cells by proteomics. Front Microbiol 2022; 13:989183. [PMID: 36171749 PMCID: PMC9511957 DOI: 10.3389/fmicb.2022.989183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Thousands of people die each year from Japanese encephalitis (JE) caused by the Japanese encephalitis virus (JEV), probably due to exacerbation of the inflammatory response that impairs the course of the disease. Microglia are mononuclear phagocytic cells located within the parenchyma of the central nervous system; these play a key role in the innate immune response against JEV infections. However, the involvement of toll-like receptor 2 (TLR2) in the inflammatory response during the early stages of JEV infection in BV2 cells remains. Here, we evaluated protein profiles and determined the role of TLR2 in the inflammatory response of JEV-infected BV2 cells. High-depth tandem mass tags labeling for quantitative proteomics was used to assess JEV infected-BV2 cells and compare immune response profiles at 6, 12, and 24 h post-infection (hpi). In total, 212 upregulated proteins were detected at 6 hpi, 754 at 12 h, and 191 at 24 h. According to GO and KEGG enrichment analysis, the upregulated proteins showed enrichment for proteins related to the immune response. Parallel reaction monitoring tests, western blotting, and qPCR results showed that the adaptor protein MyD88 was not activated. The expression levels of key proteins downstream of MyD88, such as IRAK1, IRAK4, and TRAF6 did not increase; however, the expression levels of PI3K-AKT did increase. By inhibiting key proteins (TLR2, PI3K, and AKT) we confirmed that JEV activated TLR2, thus resulting in a robust inflammatory response. Consequently, the TLR2-PI3K-AKT signaling axis was proven to play a critical in the early stages of the JEV infection-induced inflammatory response in microglia.
Collapse
Affiliation(s)
- Guanyu Zhao
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China
| | - Yan Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jiaqi Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - He Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun, China
| | - Changzhan Xie
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Fulong Nan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Sheng Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhuo Ha
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun, China
| | - Chenghui Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xiangyu Zhu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhuoxin Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ping Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ying Zhang
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China
- *Correspondence: Ying Zhang,
| | - Huijun Lu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun, China
- Huijun Lu,
| | - Ningyi Jin
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun, China
- Ningyi Jin,
| |
Collapse
|
22
|
Córdova-Dávalos LE, Hernández-Mercado A, Barrón-García CB, Rojas-Martínez A, Jiménez M, Salinas E, Cervantes-García D. Impact of genetic polymorphisms related to innate immune response on respiratory syncytial virus infection in children. Virus Genes 2022; 58:501-514. [PMID: 36085536 PMCID: PMC9462631 DOI: 10.1007/s11262-022-01932-6] [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: 03/25/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022]
Abstract
Respiratory syncytial virus (RSV) causes lower respiratory tract infections and bronchiolitis, mainly affecting children under 2 years of age and immunocompromised patients. Currently, there are no available vaccines or efficient pharmacological treatments against RSV. In recent years, tremendous efforts have been directed to understand the pathological mechanisms of the disease and generate a vaccine against RSV. Although RSV is highly infectious, not all the patients who get infected develop bronchiolitis and severe disease. Through various sequencing studies, single nucleotide polymorphisms (SNPs) have been discovered in diverse receptors, cytokines, and transcriptional regulators with crucial role in the activation of the innate immune response, which is implicated in the susceptibility to develop or protect from severe forms of the infection. In this review, we highlighted how variations in the key genes affect the development of innate immune response against RSV. This data would provide crucial information about the mechanisms of viral infection, and in the future, could help in generation of new strategies for vaccine development or generation of the pharmacological treatments.
Collapse
Affiliation(s)
- Laura Elena Córdova-Dávalos
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Alicia Hernández-Mercado
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Claudia Berenice Barrón-García
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Augusto Rojas-Martínez
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Av. Morones Prieto 3000 Pte, Los Doctores, 64710, Monterrey, Nuevo León, México
| | - Mariela Jiménez
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México
| | - Eva Salinas
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México.
| | - Daniel Cervantes-García
- Laboratorio de Inmunología, Departamento de Microbiología, Universidad Autónoma de Aguascalientes, 20100, Aguascalientes, México. .,Consejo Nacional de Ciencia y Tecnología, 03940, Ciudad de México, México.
| |
Collapse
|
23
|
Wang Y, Lv S, Zhou X, Niu X, Chen L, Yang Z, Peng D. Identification of TLR2 as a Key Target in Neuroinflammation in Vascular Dementia. Front Genet 2022; 13:860122. [PMID: 35873459 PMCID: PMC9296774 DOI: 10.3389/fgene.2022.860122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular dementia (VaD) is the second most common cause of dementia. At present, precise molecular processes of VaD are unclear. We attempted to discover the VaD relevant candidate genes, enrichment biological processes and pathways, key targets, and the underlying mechanism by microarray bioinformatic analysis. We selected GSE122063 related to the autopsy samples of VaD for analysis. We first took use of Weighted Gene Co-expression Network Analysis (WGCNA) to achieve modules related to VaD and hub genes. Second, we filtered out significant differentially expressed genes (DEGs). Third, significant DEGs then went through Geno Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Fourth, Gene Set Enrichment Analysis (GSEA) was performed. At last, we constructed the protein–protein interaction (PPI) network. The results showed that the yellow module had the strongest correlation with VaD, and we finally identified 21 hub genes. Toll-like receptor 2 (TLR2) was the top hub gene and was strongly correlated with other possible candidate genes. In total, 456 significant DEGs were filtered out and these genes were found to be enriched in the Toll receptor signaling pathway and several other immune-related pathways. In addition, Gene Set Enrichment Analysis results showed that similar pathways were significantly over-represented in TLR2-high samples. In the PPI network, TLR2 was still an important node with high weight and combined scores. We concluded that the TLR2 acts as a key target in neuroinflammation which may participate in the pathophysiological process of VaD.
Collapse
Affiliation(s)
- Yuye Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shuang Lv
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiao Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoqian Niu
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Leian Chen
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ziyuan Yang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- *Correspondence: Dantao Peng,
| |
Collapse
|
24
|
Yong YK, Wong WF, Vignesh R, Chattopadhyay I, Velu V, Tan HY, Zhang Y, Larsson M, Shankar EM. Dengue Infection - Recent Advances in Disease Pathogenesis in the Era of COVID-19. Front Immunol 2022; 13:889196. [PMID: 35874775 PMCID: PMC9299105 DOI: 10.3389/fimmu.2022.889196] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
The dynamics of host-virus interactions, and impairment of the host’s immune surveillance by dengue virus (DENV) serotypes largely remain ambiguous. Several experimental and preclinical studies have demonstrated how the virus brings about severe disease by activating immune cells and other key elements of the inflammatory cascade. Plasmablasts are activated during primary and secondary infections, and play a determinative role in severe dengue. The cross-reactivity of DENV immune responses with other flaviviruses can have implications both for cross-protection and severity of disease. The consequences of a cross-reactivity between DENV and anti-SARS-CoV-2 responses are highly relevant in endemic areas. Here, we review the latest progress in the understanding of dengue immunopathogenesis and provide suggestions to the development of target strategies against dengue.
Collapse
Affiliation(s)
- Yean Kong Yong
- Laboratory Centre, Xiamen University Malaysia, Sepang, Malaysia
- *Correspondence: Esaki M. Shankar, ; Yean Kong Yong,
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ramachandran Vignesh
- Preclinical Department, Royal College of Medicine Perak (UniKL RCMP), Universiti Kuala Lumpur, Ipoh, Malaysia
| | - Indranil Chattopadhyay
- Cancer and Microbiome Biology, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Vijayakumar Velu
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Pathology and Laboratory Medicine, Emory National Primate Research Center, Emory University, Atlanta GA, United States
| | - Hong Yien Tan
- School of Traditional Chinese Medicine, Xiamen University Malaysia, Sepang, Malaysia
| | - Ying Zhang
- Chemical Engineering, Xiamen University Malaysia, Sepang, Malaysia
| | - Marie Larsson
- Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Esaki M. Shankar
- Infection Biology, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
- *Correspondence: Esaki M. Shankar, ; Yean Kong Yong,
| |
Collapse
|
25
|
Innate Immune Response to Dengue Virus: Toll-like Receptors and Antiviral Response. Viruses 2022; 14:v14050992. [PMID: 35632732 PMCID: PMC9147118 DOI: 10.3390/v14050992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
Dengue is a mosquito-borne viral disease caused by the dengue virus (DENV1-4). The clinical manifestations range from asymptomatic to life-threatening dengue hemorrhagic fever (DHF) and/or Dengue Shock Syndrome (DSS). Viral and host factors are related to the clinical outcome of dengue, although the disease pathogenesis remains uncertain. The innate antiviral response to DENV is implemented by a variety of immune cells and inflammatory mediators. Blood monocytes, dendritic cells (DCs) and tissue macrophages are the main target cells of DENV infection. These cells recognize pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs). Pathogen recognition is a critical step in eliciting the innate immune response. Toll-like receptors (TLRs) are responsible for the innate recognition of pathogens and represent an essential component of the innate and adaptive immune response. Ten different TLRs are described in humans, which are expressed in many different immune cells. The engagement of TLRs with viral PAMPs triggers downstream signaling pathways leading to the production of inflammatory cytokines, interferons (IFNs) and other molecules essential for the prevention of viral replication. Here, we summarize the crucial TLRs’ roles in the antiviral innate immune response to DENV and their association with viral pathogenesis.
Collapse
|
26
|
Han F, Zhang Y, Xu A, Wang X, He Y, Song N, Gao T. Genome-wide identification and characterization of Toll-like receptor genes in black rockfish (Sebastes schlegelii) and their response mechanisms following poly (I:C) injection. Comp Biochem Physiol C Toxicol Pharmacol 2022; 254:109277. [PMID: 35085815 DOI: 10.1016/j.cbpc.2022.109277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 12/13/2022]
Abstract
Toll-like receptors (TLRs) are canonical transmembrane receptors that play an important role in defending against invading pathogens. In this study, we identified a total of 12 TLR genes in black rockfish (Sebastes schlegelii) with an analysis of their sequence characterizations. The phylogenetic analysis suggested that 12 distinct TLRs were grouped into five subfamilies (i.e., TLR1, TLR3, TLR5, TLR7, and TLR11 subfamilies), and each SsTLR gene respectively corresponded to the orthologs genes of other species. The protein domain analysis indicated that TLRs are type I transmembrane proteins, including an extracellular leucine-rich repeat (LRR), a transmembrane region (TM) domain and an intracellular Toll/IL-1 receptor (TIR) domain. The evolutionary ratios indicted that 12 SsTLRs were under purifying selection. qRT-PCR assays exhibited diverse TLRs molecular expression patterns in the heart, brain, head kidney, kidney, liver, intestine, and spleen of 3 black rockfish, and the expression levels were high in some immune tissues (e.g., head kidney, kidney, and spleen). Subsequently, 30 fish were equally divided into 2 groups i.e., poly (I:C)-treated and PBS-Control groups. After poly (I:C) injection, eight SsTLRs, i.e., SsTLR2, SsTLR2-1, SsTLR2-2, SsTLR3, SsTLR5S, SsTLR7, SsTLR8 and SsTLR22, were dramatically increased. Altogether these results contribute to understanding how SsTLRs respond to immune defense after poly (I:C) injection and provide researchers with comprehensive TLR gene family data of black rockfish.
Collapse
Affiliation(s)
- Fei Han
- Fisheries College, Ocean University of China, Qingdao, Shandong 266003, China
| | - Yuan Zhang
- Fisheries College, Ocean University of China, Qingdao, Shandong 266003, China
| | - Anle Xu
- Fishery College, Zhejiang Ocean University, Zhoushan, Zhejiang 316022, China
| | - Xiaoyan Wang
- Fishery College, Zhejiang Ocean University, Zhoushan, Zhejiang 316022, China
| | - Yan He
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong 266003, China
| | - Na Song
- Fisheries College, Ocean University of China, Qingdao, Shandong 266003, China
| | - Tianxiang Gao
- Fishery College, Zhejiang Ocean University, Zhoushan, Zhejiang 316022, China.
| |
Collapse
|
27
|
Valdés-López JF, Fernandez GJ, Urcuqui-Inchima S. Synergistic Effects of Toll-Like Receptor 1/2 and Toll-Like Receptor 3 Signaling Triggering Interleukin 27 Gene Expression in Chikungunya Virus-Infected Macrophages. Front Cell Dev Biol 2022; 10:812110. [PMID: 35223841 PMCID: PMC8863767 DOI: 10.3389/fcell.2022.812110] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) is the etiological agent of chikungunya fever (CHIKF), a self-limiting disease characterized by myalgia and severe acute or chronic arthralgia. CHIKF is associated with immunopathology and high levels of pro-inflammatory factors. CHIKV is known to have a wide range of tropism in human cell types, including keratinocytes, fibroblasts, endothelial cells, monocytes, and macrophages. Previously, we reported that CHIKV-infected monocytes-derived macrophages (MDMs) express high levels of interleukin 27 (IL27), a heterodimeric cytokine consisting of IL27p28 and EBI3 subunits, that triggers JAK-STAT signaling and promotes pro-inflammatory and antiviral response, in interferon (IFN)-independent manner. Based on the transcriptomic analysis, we now report that induction of IL27-dependent pro-inflammatory and antiviral response in CHIKV-infected MDMs relies on two signaling pathways: an early signal dependent on recognition of CHIKV-PAMPs by TLR1/2-MyD88 to activate NF-κB-complex that induces the expression of EBI3 mRNA; and second signaling dependent on the recognition of intermediates of CHIKV replication (such as dsRNA) by TLR3-TRIF, to activate IRF1 and the induction of IL27p28 mRNA expression. Both signaling pathways were required to produce a functional IL27 protein involved in the induction of ISGs, including antiviral proteins, cytokines, CC- and CXC- chemokines in an IFN-independent manner in MDMs. Furthermore, we reported that activation of TLR4 by LPS, both in human MDMs and murine BMDM, results in the induction of both subunits of IL27 that trigger strong IL27-dependent pro-inflammatory and antiviral response independent of IFNs signaling. Our findings are a significant contribution to the understanding of molecular and cellular mechanisms of CHIKV infection.
Collapse
|
28
|
In silico Design and Characterization of Multi-epitopes Vaccine for SARS-CoV2 from Its Spike Protein. Int J Pept Res Ther 2022; 28:37. [PMID: 35002585 PMCID: PMC8722413 DOI: 10.1007/s10989-021-10348-z] [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] [Accepted: 12/12/2021] [Indexed: 11/04/2022]
Abstract
COVID 19 is a disease caused by a novel coronavirus, SARS-CoV2 originated in China most probably of Bat origin. Multiepitopes vaccine would be useful in eliminating SARS-CoV2 infections as asymptomatic patients are in large numbers. In response to this, we utilized bioinformatic tools to develop an efficient vaccine candidate against SARS-CoV2. The designed vaccine has effective BCR and TCR epitopes screened from the sequence of S-protein of SARS-CoV2. Predicted BCR and TCR epitopes found antigenic, non-toxic and probably non-allergen. Modeled and the refined tertiary structure predicted as valid for further use. Protein–Protein interaction prediction of TLR2/4 and designed vaccine indicates promising binding. The designed multiepitope vaccine has induced cell-mediated and humoral immunity along with increased interferon-gamma response. Macrophages and dendritic cells were also found to increase upon the vaccine exposure. In silico codon optimization and cloning in expression vector indicates that the vaccine can be efficiently expressed in E. coli. In conclusion, the predicted vaccine is a good antigen, probable no allergen, and has the potential to induce cellular and humoral immunity.
Collapse
|
29
|
Li X, Dong Z, Liu Y, Song W, Pu J, Jiang G, Wu Y, Liu L, Huang X. A Novel Role for the Regulatory Nod-Like Receptor NLRP12 in Anti-Dengue Virus Response. Front Immunol 2021; 12:744880. [PMID: 34956178 PMCID: PMC8695442 DOI: 10.3389/fimmu.2021.744880] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/22/2021] [Indexed: 11/14/2022] Open
Abstract
Dengue Virus (DENV) infection can cause severe illness such as highly fatality dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Innate immune activation by Nod-like receptors (NLRs) is a critical part of host defense against viral infection. Here, we revealed a key mechanism of NLRP12-mediated regulation in DENV infection. Firstly, NLRP12 expression was inhibited in human macrophage following DENV or other flaviviruses (JEV, YFV, ZIKV) infection. Positive regulatory domain 1 (PRDM1) was induced by DENV or poly(I:C) and suppressed NLRP12 expression, which was dependent on TBK-1/IRF3 and NF-κB signaling pathways. Moreover, NLRP12 inhibited DENV and other flaviviruses (JEV, YFV, ZIKV) replication, which relied on the well-conserved nucleotide binding structures of its NACHT domain. Furthermore, NLRP12 could interact with heat shock protein 90 (HSP90) dependent on its Walker A and Walker B sites. In addition, NLRP12 enhanced the production of type I IFNs (IFN-α/β) and interferon-stimulated genes (ISGs), including IFITM3, TRAIL and Viperin. Inhibition of HSP90 with 17-DMAG impaired the upregulation of type I IFNs and ISGs induced by NLRP12. Taken together, we demonstrated a novel mechanism that NLRP12 exerted anti-viral properties in DENV and other flaviviruses (JEV, YFV, ZIKV) infection, which brings up a potential target for the treatment of DENV infection.
Collapse
Affiliation(s)
- Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhuo Dong
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yan Liu
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Weifeng Song
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jieying Pu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Lei Liu
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
30
|
SARS-CoV-2 Exposed Mesenchymal Stromal Cell from Congenital Pulmonary Airway Malformations: Transcriptomic Analysis and the Expression of Immunomodulatory Genes. Int J Mol Sci 2021; 22:ijms222111814. [PMID: 34769246 PMCID: PMC8584055 DOI: 10.3390/ijms222111814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
The inflammatory response plays a central role in the complications of congenital pulmonary airway malformations (CPAM) and severe coronavirus disease 2019 (COVID-19). The aim of this study was to evaluate the transcriptional changes induced by SARS-CoV-2 exposure in pediatric MSCs derived from pediatric lung (MSCs-lung) and CPAM tissues (MSCs-CPAM) in order to elucidate potential pathways involved in SARS-CoV-2 infection in a condition of exacerbated inflammatory response. MSCs-lung and MSCs-CPAM do not express angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TRMPSS2). SARS-CoV-2 appears to be unable to replicate in MSCs-CPAM and MSCs-lung. MSCs-lung and MSCs-CPAM maintained the expression of stemness markers MSCs-lung show an inflammatory response (IL6, IL1B, CXCL8, and CXCL10), and the activation of Notch3 non-canonical pathway; this route appears silent in MSCs-CPAM, and cytokine genes expression is reduced. Decreased value of p21 in MSCs-lung suggested no cell cycle block, and cells did not undergo apoptosis. MSCs-lung appears to increase genes associated with immunomodulatory function but could contribute to inflammation, while MSCs-CPAM keeps stable or reduce the immunomodulatory receptors expression, but they also reduce their cytokines expression. These data indicated that, independently from their perilesional or cystic origin, the MSCs populations already present in a patient affected with CPAM are not permissive for SARS-CoV-2 entry, and they will not spread the disease in case of infection. Moreover, these MSCs will not undergo apoptosis when they come in contact with SARS-CoV-2; on the contrary, they maintain their staminality profile.
Collapse
|
31
|
Castillo JA, Giraldo DM, Hernandez JC, Smit JM, Rodenhuis-Zybert IA, Urcuqui-Inchima S. Regulation of innate immune responses in macrophages differentiated in the presence of vitamin D and infected with dengue virus 2. PLoS Negl Trop Dis 2021; 15:e0009873. [PMID: 34634046 PMCID: PMC8530315 DOI: 10.1371/journal.pntd.0009873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/21/2021] [Accepted: 10/05/2021] [Indexed: 12/26/2022] Open
Abstract
A dysregulated or exacerbated inflammatory response is thought to be the key driver of the pathogenesis of severe disease caused by the mosquito-borne dengue virus (DENV). Compounds that restrict virus replication and modulate the inflammatory response could thus serve as promising therapeutics mitigating the disease pathogenesis. We and others have previously shown that macrophages, which are important cellular targets for DENV replication, differentiated in the presence of bioactive vitamin D (VitD3) are less permissive to viral replication, and produce lower levels of pro-inflammatory cytokines. Therefore, we here evaluated the extent and kinetics of innate immune responses of DENV-2 infected monocytes differentiated into macrophages in the presence (D3-MDMs) or absence of VitD3 (MDMs). We found that D3-MDMs expressed lower levels of RIG I, Toll-like receptor (TLR)3, and TLR7, as well as higher levels of SOCS-1 in response to DENV-2 infection. D3-MDMs produced lower levels of reactive oxygen species, related to a lower expression of TLR9. Moreover, although VitD3 treatment did not modulate either the expression of IFN-α or IFN-β, higher expression of protein kinase R (PKR) and 2'-5'-oligoadenylate synthetase 1 (OAS1) mRNA were found in D3-MDMs. Importantly, the observed effects were independent of reduced infection, highlighting the intrinsic differences between D3-MDMs and MDMs. Taken together, our results suggest that differentiation of MDMs in the presence of VitD3 modulates innate immunity in responses to DENV-2 infection.
Collapse
Affiliation(s)
- Jorge Andrés Castillo
- Grupo de Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, (Antioquia), Colombia
- Department of Medical Microbiology and infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Diana M. Giraldo
- Grupo de Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, (Antioquia), Colombia
| | - Juan C. Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, (Antioquia), Colombia
| | - Jolanda M. Smit
- Department of Medical Microbiology and infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Izabela A. Rodenhuis-Zybert
- Department of Medical Microbiology and infection Prevention, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Silvio Urcuqui-Inchima
- Grupo de Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, (Antioquia), Colombia
| |
Collapse
|
32
|
Sharma KB, Chhabra S, Aggarwal S, Tripathi A, Banerjee A, Yadav AK, Vrati S, Kalia M. Proteomic landscape of Japanese encephalitis virus-infected fibroblasts. J Gen Virol 2021; 102. [PMID: 34546869 DOI: 10.1099/jgv.0.001657] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Advances in proteomics have enabled a comprehensive understanding of host-pathogen interactions. Here we have characterized Japanese encephalitis virus (JEV) infection-driven changes in the mouse embryonic fibroblast (MEF) proteome. Through tandem mass tagging (TMT)-based mass spectrometry, we describe changes in 7.85 % of the identified proteome due to JEV infection. Pathway enrichment analysis showed that proteins involved in innate immune sensing, interferon responses and inflammation were the major upregulated group, along with the immunoproteasome and poly ADP-ribosylation proteins. Functional validation of several upregulated anti-viral innate immune proteins, including an active cGAS-STING axis, was performed. Through siRNA depletion, we describe a crucial role of the DNA sensor cGAS in restricting JEV replication. Further, many interferon-stimulated genes (ISGs) were observed to be induced in infected cells. We also observed activation of TLR2 and inhibition of TLR2 signalling using TLR1/2 inhibitor CU-CPT22-blocked production of inflammatory cytokines IL6 and TNF-α from virus-infected N9 microglial cells. The major proteins that were downregulated by infection were involved in cell adhesion (collagens), transport (solute carrier and ATP-binding cassette transporters), sterol and lipid biosynthesis. Several collagens were found to be transcriptionally downregulated in infected MEFs and mouse brain. Collectively, our data provide a bird's-eye view into how fibroblast protein composition is rewired following JEV infection.
Collapse
Affiliation(s)
- Kiran Bala Sharma
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India.,Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Simran Chhabra
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Suruchi Aggarwal
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Aarti Tripathi
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Arup Banerjee
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India.,Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Amit Kumar Yadav
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Sudhanshu Vrati
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India.,Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Manjula Kalia
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, Haryana, India.,Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
33
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Recent Insights Into the Molecular Mechanism of Toll-Like Receptor Response to Dengue Virus Infection. Front Microbiol 2021; 12:744233. [PMID: 34603272 PMCID: PMC8483762 DOI: 10.3389/fmicb.2021.744233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Dengue is the most prevalent and rapidly spreading mosquito-borne viral disease caused by dengue virus (DENV). Recently, DENV has been affecting humans within an expanding geographic range due to the warming of the earth. Innate immune responses play a significant role in antiviral defense, and Toll-like receptors (TLRs) are key regulators of innate immunity. Therefore, a detailed understanding of TLR and DENV interactions is important for devising therapeutic and preventive strategies. Several studies have indicated the ability of DENV to modulate the TLR signaling pathway and host immune response. Vaccination is considered one of the most successful medical interventions for preventing viral infections. However, only a partially protective dengue vaccine, the first licensed dengue vaccine CYD-TDV, is available in some dengue-endemic countries to protect against DENV infection. Therefore, the development of a fully protective, durable, and safe DENV vaccine is a priority for global health. Here, we demonstrate the progress made in our understanding of the host response to DENV infection, with a particular focus on TLR response and how DENV avoids the response toward establishing infection. We also discuss dengue vaccine candidates in late-stage development and the issues that must be overcome to enable their success.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
34
|
Hussman JP. Severe Clinical Worsening in COVID-19 and Potential Mechanisms of Immune-Enhanced Disease. Front Med (Lausanne) 2021; 8:637642. [PMID: 34239884 PMCID: PMC8258105 DOI: 10.3389/fmed.2021.637642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Infection by the novel SARS-CoV-2 coronavirus produces a range of outcomes, with the majority of cases producing mild or asymptomatic effects, and a smaller subset progressing to critical or fatal COVID-19 disease featuring severe acute respiratory distress. Although the mechanisms driving severe disease progression remain unknown, it is possible that the abrupt clinical deterioration observed in patients with critical disease corresponds to a discrete underlying expansion of viral tropism, from infection of cells comprising respiratory linings and alveolar epithelia to direct infection and activation of inflammatory monocytes and macrophages. Dysregulated immune responses could then contribute to disease severity. This article discusses the potential role of monocyte/macrophage (Mo/Mϕ) infection by SARS-CoV-2 in mediating the immune response in severe COVID-19. Additional mechanisms of immune-enhanced disease, comprising maladaptive immune responses that may aggravate rather than alleviate severity, are also discussed. Severe acute clinical worsening in COVID-19 patients may be influenced by the emergence of antibodies that participate in hyperinflammatory monocyte response, release of neutrophil extracellular traps (NETs), thrombosis, platelet apoptosis, viral entry into Fc gamma receptor (FcγR)-expressing immune cells, and induction of autoantibodies with cross-reactivity against host proteins. While the potential roles of Mo/Mϕ infection and immune-enhanced pathology in COVID-19 are consistent with a broad range of clinical and laboratory findings, their prominence remains tentative pending further validation. In the interim, these proposed mechanisms present immediate avenues of inquiry that may help to evaluate the safety of candidate vaccines and antibody-based therapeutics, and to support consideration of pathway-informed, well-tolerated therapeutic candidates targeting the dysregulated immune response.
Collapse
|
35
|
Bournazos S, Vo HTM, Duong V, Auerswald H, Ly S, Sakuntabhai A, Dussart P, Cantaert T, Ravetch JV. Antibody fucosylation predicts disease severity in secondary dengue infection. Science 2021; 372:1102-1105. [PMID: 34083490 DOI: 10.1126/science.abc7303] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 01/19/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022]
Abstract
Although antiviral antibodies generally confer protective functions, antibodies against dengue virus (DENV) are associated with enhanced disease susceptibility. Antibodies can mediate DENV infection of leukocytes via Fcγ receptors, likely contributing to dengue disease pathogenesis. To determine if this mechanism accounts for variable disease severity, we examined Fab and Fc structures of anti-DENV antibodies from patients before and after infection and with variable disease outcomes. Neither antibody titers nor neutralizing activity correlated with disease severity in DENV-infected populations. Rather, DENV infection induced a specific increase in immunoglobulin G1 (IgG1) afucosylation, and the levels of afucosylated IgG1 were predictive of dengue disease severity. Thus, the IgG1 fucosylation status represents a robust prognostic tool for dengue disease, highlighting the key role of the Fc glycan structure in dengue pathogenesis.
Collapse
Affiliation(s)
- Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology 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
| | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Anavaj Sakuntabhai
- Functional Genetics of Infectious Diseases Unit, Department of Global Health, Institut Pasteur, Paris Cedex 15, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - 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.
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
36
|
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: 1.0] [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
|
37
|
Sellau J, Puengel T, Hoenow S, Groneberg M, Tacke F, Lotter H. Monocyte dysregulation: consequences for hepatic infections. Semin Immunopathol 2021; 43:493-506. [PMID: 33829283 PMCID: PMC8025899 DOI: 10.1007/s00281-021-00852-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
Liver disorders due to infections are a substantial health concern in underdeveloped and industrialized countries. This includes not only hepatotropic viruses (e.g., hepatitis B, hepatitis C) but also bacterial and parasitic infections such as amebiasis, leishmaniasis, schistosomiasis, or echinococcosis. Recent studies of the immune mechanisms underlying liver disease show that monocytes play an essential role in determining patient outcomes. Monocytes are derived from the mononuclear phagocyte lineage in the bone marrow and are present in nearly all tissues of the body; these cells function as part of the early innate immune response that reacts to challenge by external pathogens. Due to their special ability to develop into tissue macrophages and dendritic cells and to change from an inflammatory to an anti-inflammatory phenotype, monocytes play a pivotal role in infectious and non-infectious liver diseases: they can maintain inflammation and support resolution of inflammation. Therefore, tight regulation of monocyte recruitment and termination of monocyte-driven immune responses in the liver is prerequisite to appropriate healing of organ damage. In this review, we discuss monocyte-dependent immune mechanisms underlying hepatic infectious disorders. Better understanding of these immune mechanisms may lead to development of new interventions to treat acute liver disease and prevent progression to organ failure.
Collapse
Affiliation(s)
- Julie Sellau
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Tobias Puengel
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Stefan Hoenow
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marie Groneberg
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Hannelore Lotter
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
38
|
Chung NPY, Khan KMF, Kaner RJ, O'Beirne SL, Crystal RG. HIV induces airway basal progenitor cells to adopt an inflammatory phenotype. Sci Rep 2021; 11:3988. [PMID: 33597552 PMCID: PMC7889866 DOI: 10.1038/s41598-021-82143-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the introduction of anti-retroviral therapy, chronic HIV infection is associated with an increased incidence of other comorbidities such as COPD. Based on the knowledge that binding of HIV to human airway basal stem/progenitor cells (BC) induces a destructive phenotype by increased MMP-9 expression through MAPK signaling pathways, we hypothesized that HIV induces the BC to express inflammatory mediators that contribute to the pathogenesis of emphysema. Our data demonstrate that airway BC isolated from HAART-treated HIV+ nonsmokers spontaneously release inflammatory mediators IL-8, IL-1β, ICAM-1 and GM-CSF. Similarly, exposure of normal BC to HIV in vitro up-regulates expression of the same inflammatory mediators. These HIV-BC derived mediators induce migration of alveolar macrophages (AM) and neutrophils and stimulate AM proliferation. This HIV-induced inflammatory phenotype likely contributes to lung inflammation in HIV+ individuals and provides explanation for the increased incidence of COPD in HIV+ individuals.
Collapse
Affiliation(s)
- Nancy P Y Chung
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - K M Faisal Khan
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Robert J Kaner
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Sarah L O'Beirne
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
39
|
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: 20] [Impact Index Per Article: 5.0] [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
|
40
|
Gadanec LK, McSweeney KR, Qaradakhi T, Ali B, Zulli A, Apostolopoulos V. Can SARS-CoV-2 Virus Use Multiple Receptors to Enter Host Cells? Int J Mol Sci 2021; 22:992. [PMID: 33498183 PMCID: PMC7863934 DOI: 10.3390/ijms22030992] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
The occurrence of the novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), responsible for coronavirus disease 2019 (COVD-19), represents a catastrophic threat to global health. Protruding from the viral surface is a densely glycosylated spike (S) protein, which engages angiotensin-converting enzyme 2 (ACE2) to mediate host cell entry. However, studies have reported viral susceptibility in intra- and extrapulmonary immune and non-immune cells lacking ACE2, suggesting that the S protein may exploit additional receptors for infection. Studies have demonstrated interactions between S protein and innate immune system, including C-lectin type receptors (CLR), toll-like receptors (TLR) and neuropilin-1 (NRP1), and the non-immune receptor glucose regulated protein 78 (GRP78). Recognition of carbohydrate moieties clustered on the surface of the S protein may drive receptor-dependent internalization, accentuate severe immunopathological inflammation, and allow for systemic spread of infection, independent of ACE2. Furthermore, targeting TLRs, CLRs, and other receptors (Ezrin and dipeptidyl peptidase-4) that do not directly engage SARS-CoV-2 S protein, but may contribute to augmented anti-viral immunity and viral clearance, may represent therapeutic targets against COVID-19.
Collapse
|
41
|
Aguilar-Briseño JA, Moser J, Rodenhuis-Zybert IA. Understanding immunopathology of severe dengue: lessons learnt from sepsis. Curr Opin Virol 2020; 43:41-49. [PMID: 32896675 DOI: 10.1016/j.coviro.2020.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022]
Abstract
Endothelial dysfunction leading to vascular permeability and plasma leakage are characteristic features of severe dengue and sepsis. However, the mechanisms underlying these immune-pathologies remain unclear. The risk of severe dengue and sepsis development depend on patient-related and pathogen-related factors. Additionally, comorbidities increase the risk of severe disease and their incidence hampers correct diagnosis and treatments. To date, there is no efficient therapy to combat severe dengue and sepsis. Here, we discuss the differences and similarities between the pathogenesis of severe dengue and that of bacterial sepsis. We identify gaps in knowledge that need to be better understood in order to move towards the rational development and/or usage of therapeutic strategies to ameliorate severe dengue disease.
Collapse
Affiliation(s)
- José A Aguilar-Briseño
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Jill Moser
- Departments of Critical Care, Pathology & Medical Biology, Medical Biology Section, University of Groningen and University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Izabela A Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University of Groningen and University Medical Center Groningen, 9700 RB Groningen, The Netherlands.
| |
Collapse
|