1
|
Li K, Zhou Z, Liu F, Huang Z, Chen X, Zhou F. Unphosphorylated STAT1 binds to the BST2 transcription promoter, promoting increased AKBA anchoring on HPMECs to alleviate ARDS. Sci Rep 2025; 15:15207. [PMID: 40307322 DOI: 10.1038/s41598-025-00028-z] [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: 01/03/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025] Open
Abstract
Although the drug therapeutic targets of acute respiratory distress syndrome (ARDS) are still unclear and no specific drugs for ARDS treatment have been found, some breakthroughs have been gradually made in the biological target pathways such as endothelial injury. The Traditional Chinese Medicine Systems Pharmacology (TCMSP) database suggests that Acetyl-11-keto-β-boswellic acid (AKBA), a processed product of boswellic acid, may be an effective intervention for ARDS. After preliminary in vitro and in vivo verification of the protective role of AKBA on ARDS, in order to explore the mechanism of AKBA in ARDS, we used transcriptomic and proteomic methods to explore its main targets, and used molecular docking and cell thermal shift assay (CETSA) to further reveal the potential value of bone marrow stromal cell antigen 2 (BST2) as a target. We subsequently examined the effect of AKBA targeting BST2 on tubule formation, cell proliferation (Colony formation and EdU assay), migration (transwell and scratch assays), apoptosis and autophagy levels both in vitro and in vivo, and protein changes (analyzed by Western blotting analysis). Our results show that the unphosphorylated signal transducers and transcription activation factors (U-STAT1) bins to the BST2 transcription promoter to encourage more AKBA anchoring the human pulmonary microvascular endothelial cells (HPMECs), thus inhibiting apoptosis and autophagy, promoting migration and tube formation, and restraining the cecal ligation and puncture (CLP) induced lung tissue damage in mice. In conclusion, AKBA treatment may be a potential strategy in the intervention of ARDS. Alternatively, BST2 may contribute to the anchoring of AKBA to HPMECs, and STAT1 as a transcription factor promoting BST2 expression may bind to its promoter.
Collapse
Affiliation(s)
- Kaili Li
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Yuzhong District, 3rd Floor, Building 7, 400016, Chongqing, China
| | - Zixiang Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Feng Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Zuotian Huang
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, 400030, Chongqing Municipality, China.
| | - Xiaoying Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China.
| | - Fachun Zhou
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Yuzhong District, 3rd Floor, Building 7, 400016, Chongqing, China.
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
2
|
Li L, Zhu X, Xu W, Dai M, Liu Z, Li Y, Fang Y, Li J, Chen W. A prospective self-controlled study on the alterations of the ocular surface and conjunctival transcriptomic profile associated with prolonged exposure to video display terminals. Ocul Surf 2025; 36:94-105. [PMID: 39828134 DOI: 10.1016/j.jtos.2025.01.007] [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: 07/25/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE To assess the impact of prolonged and intense exposure to video display terminals (VDTs) on ocular surface homeostasis. METHODS 30 subjects limited daily VDT usage to less than 3 h for one week, then extended usage to more than 8 h/day for the next three weeks. Ocular symptoms and signs were evaluated weekly using the Ocular Surface Disease Index (OSDI) questionnaire and clinical examinations. Eyelid margins and meibomian glands were examined, and ocular surface samples were collected for transcriptomic analysis. RESULTS Average daily VDT time increased from 2.55 ± 0.46 h initially to 11.17 ± 2.45, 11.75 ± 2.63, and 10.89 ± 2.41 h over three weeks. The dry eye diagnosis rate rose from 6.67 % to 51.67 %. Total OSDI score (P = 0.008), symptoms score (P = 0.014), and visual function score (P = 0.002) significantly increased. Mean fluorescein break-up time (FBUT) decreased from 6.46s to 3.08s. Corneal fluorescein staining (CFS) score (P < 0.001) and lissamine green conjunctival staining (LCjs) score (P = 0.036) worsened. Ocular redness index (RI) increased at 1 week and 3 weeks (P = 0.007, P = 0.001). Telangiectasia scores of both upper and lower eyelid margins increased at 3 weeks (P = 0.002, P < 0.001). Meibomian gland orifice blockage worsened (P = 0.014, P = 0.002). Transcriptomic analysis revealed dynamic alterations in ocular surface gene expression, including inflammatory and hormonal responses. MUC5AC and TFF1 genes showed negative correlations with OSDI and conjunctival staining score, respectively. CONCLUSION Prolonged VDT exposure deteriorates ocular surface symptoms and signs, with significant inflammatory responses and hormonal activity indicating an imbalance in ocular surface homeostasis.
Collapse
Affiliation(s)
- Ling Li
- Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, 315042, China; National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xinhao Zhu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Weihao Xu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Mali Dai
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zihao Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yanxiao Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yiting Fang
- Hangzhou Lin'an Traditional Chinese Medicine Hospital, Hangzhou, 311300, China
| | - Jinyang Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; Department of Ophthalmology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 32400, Zhejiang Province, China.
| | - Wei Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China; Ningbo Eye Institute, Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, 315040, China.
| |
Collapse
|
3
|
Omara D, Natwijuka F, Kapaata A, Kato F, Kato L, Ndekezi C, Nakyanzi A, Ayebale ML, Yue L, Hunter E, Sande OJ, Ochsenbauer C, Kaleebu P, Balinda SN. Subtype AD Recombinant HIV-1 Transmitted/Founder Viruses Are Less Sensitive to Type I Interferons than Subtype D. Viruses 2025; 17:486. [PMID: 40284929 PMCID: PMC12031311 DOI: 10.3390/v17040486] [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: 02/14/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/29/2025] Open
Abstract
Initial interactions between HIV-1 and the immune system at mucosal exposure sites play a critical role in determining whether the virus is eliminated or progresses to establish systemic infection. The virus that successfully crosses the mucosal barrier to establish infection in the new host is referred to as the transmitted/founder (TF) virus. Following mucosal HIV-1 transmission, type 1 interferons (IFN-I) are rapidly induced at sites of initial virus replication. The resistance of TF variants to these antiviral effects of the IFN-I has been studied among HIV-1 subtypes B and C. However, their role in restricting HIV-1 replication among subtypes D and AD recombinant remains unexplored. This study assessed the sensitivity of HIV-1 subtype D and AD recombinant TF viruses to IFN-I by infecting peripheral blood mononuclear cells in vitro with infectious molecular clones of these viruses. Cells were exposed to varying concentrations of interferon-α and interferon-β, and viral replicative capacity was measured using HIV-1 p24 antigen ELISA from culture supernatants. Sensitivity to IFN-I was quantified based on viral replication levels. The results showed that interferon-α was more effective in inhibiting viral replication than interferon-β, regardless of the varying amounts of IFN-I used. However, recombinant AD viruses were found to be more resistant to the antiviral effects of IFN-I compared to subtype D viruses. These findings highlight the differential sensitivity of HIV-1 subtypes AD recombinant and D TF viruses to IFN-I and underscore the potential of IFN-I as a therapeutic strategy to target TF viruses and reduce HIV-1 transmission, particularly in populations where subtype D is prevalent.
Collapse
Affiliation(s)
- Denis Omara
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala P.O. Box 7062, Uganda; (D.O.); (F.N.); (F.K.); (C.N.); (O.J.S.)
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
| | - Fortunate Natwijuka
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala P.O. Box 7062, Uganda; (D.O.); (F.N.); (F.K.); (C.N.); (O.J.S.)
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
| | - Anne Kapaata
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
| | - Frank Kato
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala P.O. Box 7062, Uganda; (D.O.); (F.N.); (F.K.); (C.N.); (O.J.S.)
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
| | - Laban Kato
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
| | - Christian Ndekezi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala P.O. Box 7062, Uganda; (D.O.); (F.N.); (F.K.); (C.N.); (O.J.S.)
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
| | - Angella Nakyanzi
- Uganda Virus Research Institute (UVRI), Entebbe P.O. Box 49, Uganda
| | - Mercy L. Ayebale
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
| | - Ling Yue
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA 30329, USA; (L.Y.); (E.H.)
| | - Eric Hunter
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA 30329, USA; (L.Y.); (E.H.)
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Obondo J. Sande
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala P.O. Box 7062, Uganda; (D.O.); (F.N.); (F.K.); (C.N.); (O.J.S.)
| | - Christina Ochsenbauer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Pontiano Kaleebu
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
- Uganda Virus Research Institute (UVRI), Entebbe P.O. Box 49, Uganda
| | - Sheila N. Balinda
- Medical Research Council, Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine (MRC/UVRI & LSHTM), Uganda Research Unit, Entebbe P.O. Box 49, Uganda; (A.K.); (L.K.); (M.L.A.); (P.K.)
- Uganda Virus Research Institute (UVRI), Entebbe P.O. Box 49, Uganda
| |
Collapse
|
4
|
Lin Y, Zhu Y, Jing L, Lei X, Xie Z. Regulation of viral replication by host restriction factors. Front Immunol 2025; 16:1484119. [PMID: 39917304 PMCID: PMC11798991 DOI: 10.3389/fimmu.2025.1484119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Viral infectious diseases, caused by numerous viruses including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza A virus (IAV), enterovirus (EV), human immunodeficiency virus (HIV), hepatitis B virus (HBV), and human papillomavirus (HPV), pose a continuous threat to global health. As obligate parasites, viruses rely on host cells to replicate, and host cells have developed numerous defense mechanisms to counteract viral infection. Host restriction factors (HRFs) are critical components of the early antiviral response. These cellular proteins inhibit viral replication and spread by impeding essential steps in the viral life cycle, such as viral entry, genome transcription and replication, protein translation, viral particle assembly, and release. This review summarizes the current understanding of how host restriction factors inhibit viral replication, with a primary focus on their diverse antiviral mechanisms against a range of viruses, including SARS-CoV-2, influenza A virus, enteroviruses, human immunodeficiency virus, hepatitis B virus, and human papillomavirus. In addition, we highlight the crucial role of these factors in shaping the host-virus interactions and discuss their potential as targets for antiviral drug development.
Collapse
Affiliation(s)
- Ying Lin
- National Health Commission (NHC) Key Laboratory of System Biology of Pathogens and Christophe Merieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Zhu
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| | - Ling Jing
- National Health Commission (NHC) Key Laboratory of System Biology of Pathogens and Christophe Merieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Lei
- National Health Commission (NHC) Key Laboratory of System Biology of Pathogens and Christophe Merieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China
| | - Zhengde Xie
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Nguyen NM, Conrady CD. A Better Understanding of the Clinical and Pathological Changes in Viral Retinitis: Steps to Improve Visual Outcomes. Microorganisms 2024; 12:2513. [PMID: 39770716 PMCID: PMC11678148 DOI: 10.3390/microorganisms12122513] [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: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Infectious retinitis, though rare, poses a significant threat to vision, often leading to severe and irreversible damage. Various pathogens, including viruses, bacteria, tick-borne agents, parasites, and fungi, can cause this condition. Among these, necrotizing herpetic retinitis represents a critical spectrum of retinal infections primarily caused by herpes viruses such as varicella-zoster virus (VZV), herpes simplex virus (HSV), and cytomegalovirus (CMV). This review underscores the retina's susceptibility to viral infections, focusing on the molecular mechanisms through which herpetic viruses invade and damage retinal tissue, supported by clinical and preclinical evidence. We also identify existing knowledge gaps and propose future research directions to deepen our understanding and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Nghi M. Nguyen
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christopher D. Conrady
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
6
|
Allweier J, Bartels M, Torabi H, Tauler MDPM, Metwally NG, Roeder T, Gutsmann T, Bruchhaus I. Cytoadhesion of Plasmodium falciparum-Infected Red Blood Cells Changes the Expression of Cytokine-, Histone- and Antiviral Protein-Encoding Genes in Brain Endothelial Cells. Mol Microbiol 2024; 122:948-967. [PMID: 39630601 DOI: 10.1111/mmi.15331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Malaria remains a significant global health problem, mainly due to Plasmodium falciparum, which is responsible for most fatal infections. Infected red blood cells (iRBCs) evade spleen clearance by adhering to endothelial cells (ECs), triggering capillary blockage, inflammation, endothelial dysfunction and altered vascular permeability, prompting an endothelial transcriptional response. The iRBCIT4var04/HBEC-5i model, where iRBCs present IT4var04 (VAR2CSA) on their surface, was used to analyze the effects of iRBC binding on ECs at different temperature (37°C vs. 40°C). Binding of non-infected RBCs (niRBCs) and fever alone altered the expression of hundreds of genes in ECs. Comparing the expression profile of HBEC-5i cells cultured either in the presence of iRBCs or in the presence of niRBCs revealed significant upregulation of genes linked to immune response, nucleosome assembly, NF-kappa B signaling, angiogenesis, and antiviral immune response/interferon-alpha/beta signaling. Raising the temperature to 40°C, simulating fever, led to further upregulation of many genes, particularly those involved in cytokine production and angiogenesis. In summary, the presence of iRBCs stimulates ECs, activating several immunological pathways and affecting antiviral (-parasitic) mechanisms and angiogenesis. Our data uncovered the induction of the interferon-alpha/beta signaling pathway in ECs in response to iRBCs.
Collapse
Affiliation(s)
- Johannes Allweier
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Division of Biophysics, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Michael Bartels
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Hanifeh Torabi
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Nahla Galal Metwally
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Thomas Roeder
- Department Molecular Physiology, Zoology, Kiel University, Kiel, Germany
- DZL, German Center for Lung Research, ARCN, Airway Research Center North, Kiel, Germany
| | - Thomas Gutsmann
- Division of Biophysics, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Iris Bruchhaus
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Biology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
7
|
Zhou F, Sun Y, Chen X, Hou W, Shen J, Lai W, Han K, Zheng Y. Differences in cell subsets between sun-exposed and unexposed skin: preliminary single-cell sequencing and biological analysis from a single case. Front Med (Lausanne) 2024; 11:1453940. [PMID: 39540047 PMCID: PMC11558528 DOI: 10.3389/fmed.2024.1453940] [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: 06/24/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction The composition and subsets of skin cells continuously change in a dynamic manner. However, the specific microcosmic alterations of human photoaged skin, independent of chronologic aging, remain unclear and have been infrequently analyzed. This study aimed to evaluate the biological processes and mechanisms underlying cell-subgroup alterations in skin photoaging. Methods We utilized single-cell sequencing and biological analysis from a single case to investigate the effects of photoaging. Skin punch biopsies were taken from sun-exposed forearm skin and unexposed buttock skin from the same individual for comparative analysis. Results Our analysis identified 25 cell clusters and 12 skin cell types, revealing significant changes in unique gene expressions between the sun-exposed and unexposed skin samples. A comparison of cell numbers within each cluster revealed 9 dominant cell clusters in sun-exposed skin and 16 dominant cell clusters in unexposed skin. Enrichment analysis indicated that PD-L1 expression and the PD-1 checkpoint pathway were more prominent in sun-exposed skin, while MAPK, TNF-alpha, TGF-beta, and apoptosis pathways were more enriched in hair follicle cells of sun-exposed skin. Discussion This study reveals changes in cell components in photoaged skin from a single case and provides novel insights into cellular subpopulations and pathology during repeated UVA-induced skin damage. These findings enhance our understanding of the complex interplay between different cells in photoaged skin and offer potential targets for preventing human skin photoaging and UV-induced skin cancers.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Sun
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinling Chen
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenyi Hou
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Shen
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Lai
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kai Han
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Zheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Serrero MC, Paludan SR. Restriction factors regulating human herpesvirus infections. Trends Immunol 2024; 45:662-677. [PMID: 39198098 DOI: 10.1016/j.it.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Herpesviruses are DNA viruses and the cause of diseases ranging from mild skin conditions to severe brain diseases. Mammalian antiviral host defense comprises an array of mechanisms, including restriction factors (RFs), which block specific steps in viral replication cycles. In recent years, knowledge of RFs that contribute to controlling herpesvirus infections has expanded significantly, along with a new understanding of viral evasion mechanisms and disease pathogenesis. By integrating findings from human genetics, murine models, and cellular studies, this review provides a current view of RF control of herpesvirus infections. We also explore the regulation of RF expression, discuss the roles of RFs in diseases, and point towards their growing potential as candidate therapeutic targets.
Collapse
Affiliation(s)
- Manutea C Serrero
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus, Denmark
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus, Denmark.
| |
Collapse
|
9
|
Herreros-Cabello A, Del Moral-Salmoral J, Morato E, Marina A, Barrocal B, Fresno M, Gironès N. Quantitative Proteomic Analysis of Macrophages Infected with Trypanosoma cruzi Reveals Different Responses Dependent on the SLAMF1 Receptor and the Parasite Strain. Int J Mol Sci 2024; 25:7493. [PMID: 39000601 PMCID: PMC11242706 DOI: 10.3390/ijms25137493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Chagas disease is caused by the intracellular protozoan parasite Trypanosoma cruzi. This disease affects mainly rural areas in Central and South America, where the insect vector is endemic. However, this disease has become a world health problem since migration has spread it to other continents. It is a complex disease with many reservoirs and vectors and high genetic variability. One of the host proteins involved in the pathogenesis is SLAMF1. This immune receptor acts during the infection of macrophages controlling parasite replication and thus affecting survival in mice but in a parasite strain-dependent manner. Therefore, we studied the role of SLAMF1 by quantitative proteomics in a macrophage in vitro infection and the different responses between Y and VFRA strains of Trypanosoma cruzi. We detected different significant up- or downregulated proteins involved in immune regulation processes, which are SLAMF1 and/or strain-dependent. Furthermore, independently of SLAMF1, this parasite induces different responses in macrophages to counteract the infection and kill the parasite, such as type I and II IFN responses, NLRP3 inflammasome activation, IL-18 production, TLR7 and TLR9 activation specifically with the Y strain, and IL-11 signaling specifically with the VFRA strain. These results have opened new research fields to elucidate the concrete role of SLAMF1 and discover new potential therapeutic approaches for Chagas disease.
Collapse
Affiliation(s)
- Alfonso Herreros-Cabello
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Javier Del Moral-Salmoral
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Esperanza Morato
- Unidad de Proteómica, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
| | - Anabel Marina
- Unidad de Proteómica, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain
- Unidad de Técnicas Bioanalíticas (BAT), Instituto de Investigación de Ciencias de la Alimentación (CIAL), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Beatriz Barrocal
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
| | - Manuel Fresno
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid (IUBM-UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Núria Gironès
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049 Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid (IUBM-UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| |
Collapse
|
10
|
Chen S, Liang B, Xu J. Unveiling heterogeneity in MSCs: exploring marker-based strategies for defining MSC subpopulations. J Transl Med 2024; 22:459. [PMID: 38750573 PMCID: PMC11094970 DOI: 10.1186/s12967-024-05294-5] [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: 11/28/2023] [Accepted: 05/11/2024] [Indexed: 05/19/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a heterogeneous cell population distributed throughout various tissues, demonstrating remarkable adaptability to microenvironmental cues and holding immense promise for disease treatment. However, the inherent diversity within MSCs often leads to variability in therapeutic outcomes, posing challenges for clinical applications. To address this heterogeneity, purification of MSC subpopulations through marker-based isolation has emerged as a promising approach to ensure consistent therapeutic efficacy. In this review, we discussed the reported markers of MSCs, encompassing those developed through candidate marker strategies and high-throughput approaches, with the aim of explore viable strategies for addressing the heterogeneity of MSCs and illuminate prospective research directions in this field.
Collapse
Affiliation(s)
- Si Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Bowei Liang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, People's Republic of China
| | - Jianyong Xu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Fuqiang Avenue 1001, Shenzhen, 518060, Guangdong, People's Republic of China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China.
| |
Collapse
|
11
|
Li Q, Wen W, Wang Y, Gong T, Wang X, Tan Q, Fan B, Xie H, Li Y, Li S, Yang C, Zhou Z, Duan X, Lin W, Chen L. Autophagy-related protein 5 (ATG5) interacts with bone marrow stromal cell antigen 2 (BST2) to stimulate HBV replication through antagonizing the antiviral activity of BST2. J Med Virol 2024; 96:e29659. [PMID: 38747016 DOI: 10.1002/jmv.29659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Hepatitis B virus (HBV) infection is a major global health burden with 820 000 deaths per year. In our previous study, we found that the knockdown of autophagy-related protein 5 (ATG5) significantly upregulated the interferon-stimulated genes (ISGs) expression to exert the anti-HCV effect. However, the regulation of ATG5 on HBV replication and its underlying mechanism remains unclear. In this study, we screened the altered expression of type I interferon (IFN-I) pathway genes using RT² Profiler™ PCR array following ATG5 knock-down and we found the bone marrow stromal cell antigen 2 (BST2) expression was significantly increased. We then verified the upregulation of BST2 by ATG5 knockdown using RT-qPCR and found that the knockdown of ATG5 activated the Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling pathway. ATG5 knockdown or BST2 overexpression decreased Hepatitis B core Antigen (HBcAg) protein, HBV DNA levels in cells and supernatants of HepAD38 and HBV-infected NTCP-HepG2. Knockdown of BST2 abrogated the anti-HBV effect of ATG5 knockdown. Furthermore, we found that ATG5 interacted with BST2, and further formed a ternary complex together with HBV-X (HBx). In conclusion, our finding indicates that ATG5 promotes HBV replication through decreasing BST2 expression and interacting with it directly to antagonize its antiviral function.
Collapse
Affiliation(s)
- Qingyuan Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Wenxian Wen
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Yijin Wang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tao Gong
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Xinwei Wang
- Joint Laboratory on Transfusion-transmitted Infectious Diseases between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Nanning City, Nanning, Guangxi, China
| | - Qi Tan
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Bin Fan
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - He Xie
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xian, Shaanxi, China
| | - Yujia Li
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Shilin Li
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Chunhui Yang
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Zhonghui Zhou
- Department of Infectious Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaoqiong Duan
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Wenyu Lin
- Department of Medicine, Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Limin Chen
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
- Joint Laboratory on Transfusion-transmitted Infectious Diseases between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Nanning City, Nanning, Guangxi, China
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xian, Shaanxi, China
| |
Collapse
|
12
|
Cheng H, Zhang H, Cai H, Liu M, Wen S, Ren J. Molecular biology of canine parainfluenza virus V protein and its potential applications in tumor immunotherapy. Front Microbiol 2023; 14:1282112. [PMID: 38173672 PMCID: PMC10761501 DOI: 10.3389/fmicb.2023.1282112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
Canine parainfluenza virus (CPIV) is a zoonotic virus that is widely distributed and is the main pathogen causing canine infectious respiratory disease (CIRD), also known as "kennel cough," in dogs. The CPIV-V protein is the only nonstructural protein of the virus and plays an important role in multiple stages of the virus life cycle by inhibiting apoptosis, altering the host cell cycle and interfering with the interferon response. In addition, studies have shown that the V protein has potential applications in the field of immunotherapy in oncolytic virus therapy or self-amplifying RNA vaccines. In this review, the biosynthesis, structural characteristics and functions of the CPIV-V protein are reviewed with an emphasis on how it facilitates viral immune escape and its potential applications in the field of immunotherapy. Therefore, this review provides a scientific basis for research into the CPIV-V protein and its potential applications.
Collapse
Affiliation(s)
- Huai Cheng
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Hewei Zhang
- College of Food and Drugs, Luoyang Polytechnic, Luoyang, China
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang, China
| | - Huanchang Cai
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Min Liu
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
| | - Shubo Wen
- Preventive Veterinary Laboratory, College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China
| | - Jingqiang Ren
- Wenzhou Key Laboratory for Virology and Immunology, Institute of Virology, Wenzhou University, Wenzhou, China
- Animal Diseases and Public Health Engineering Research Center of Henan Province, Luoyang, China
| |
Collapse
|
13
|
Tanwattana N, Wanasen N, Jantraphakorn Y, Srisutthisamphan K, Chailungkarn T, Boonrungsiman S, Lumlertdacha B, Lekchareonsuk P, Kaewborisuth C. Human BST2 inhibits rabies virus release independently of cysteine-linked dimerization and asparagine-linked glycosylation. PLoS One 2023; 18:e0292833. [PMID: 37922253 PMCID: PMC10624315 DOI: 10.1371/journal.pone.0292833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 09/29/2023] [Indexed: 11/05/2023] Open
Abstract
The innate immune response is a first-line defense mechanism triggered by rabies virus (RABV). Interferon (IFN) signaling and ISG products have been shown to confer resistance to RABV at various stages of the virus's life cycle. Human tetherin, also known as bone marrow stromal cell antigen 2 (hBST2), is a multifunctional transmembrane glycoprotein induced by IFN that has been shown to effectively counteract many viruses through diverse mechanisms. Here, we demonstrate that hBST2 inhibits RABV budding by tethering new virions to the cell surface. It was observed that release of virus-like particles (VLPs) formed by RABV G (RABV-G VLPs), but not RABV M (RABV-G VLPs), were suppressed by hBST2, indicating that RABV-G has a specific effect on the hBST2-mediated restriction of RABV. The ability of hBST2 to prevent the release of RABV-G VLPs and impede RABV growth kinetics is retained even when hBST2 has mutations at dimerization and/or glycosylation sites, making hBST2 an antagonist to RABV, with multiple mechanisms possibly contributing to the hBST2-mediated suppression of RABV. Our findings expand the knowledge of host antiviral mechanisms that control RABV infection.
Collapse
Affiliation(s)
- Nathiphat Tanwattana
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Nanchaya Wanasen
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Yuparat Jantraphakorn
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Thanathom Chailungkarn
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Suwimon Boonrungsiman
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), KlongLuang, Pathum Thani, Thailand
| | - Boonlert Lumlertdacha
- Queen Saovabha Memorial Institute, Thai Red Cross Society, WHO Collaborating Center for Research and Training Prophylaxis on Rabies, Pathumwan, Bangkok, Thailand
| | - Porntippa Lekchareonsuk
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
- Center for Advance Studies in Agriculture and Food, KU Institute Studies, Kasetsart University, Bangkok, Thailand
| | - Challika Kaewborisuth
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| |
Collapse
|
14
|
Zhang Y, Kong N, Ti J, Cao D, Sui Z, Ge A, Pan L, Zhao K, Zhou Y, Tong G, Li L, Gao F. BST2 negatively regulates porcine reproductive and respiratory syndrome virus replication by restricting the expression of viral proteins. Virus Res 2023; 334:199181. [PMID: 37495116 PMCID: PMC10405318 DOI: 10.1016/j.virusres.2023.199181] [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: 05/27/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 07/28/2023]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has seriously affected the viability of swine industries worldwide, and effective measures to control PRRSV are urgently required. Understanding the mechanisms of action of antiviral proteins is crucial for developing antiviral strategies. Interferon-induced bone marrow stromal cell antigen 2 (BST2) can inhibit the replication of various viruses via different pathways. However, little is known about the effects of BST2 on PRRSV. Therefore, this study aimed to evaluate whether the interferon-induced BST2 can inhibit PRRSV replication. We used western blotting and RT-qPCR techniques to analyze the effect of BST2 overexpression and knockdown on PRRSV replication. Overexpression of BST2 inhibited the replication of PRRSV, whereas knockdown of BST2 by small interfering RNA promoted PRRSV replication. Additionally, the expression of BST2 was upregulated during the early phase of PRRSV infection in porcine alveolar macrophages. Analysis of PRRSV proteins showed that BST2 restricted the expression of several non-structural viral proteins. BST2 downregulated the expression of Nsp12 through a proteasome-dependent pathway and downregulated the expression and transcription of E protein. These findings demonstrate the potential of BST2 as a critical regulator of PRRSV replication.
Collapse
Affiliation(s)
- Yujiao Zhang
- Shandong Vocational Animal Science and Veterinary College, Weifang 261061, PR China; Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Ning Kong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Jinfeng Ti
- Shandong Vocational Animal Science and Veterinary College, Weifang 261061, PR China
| | - Dongshen Cao
- Shandong Vocational Animal Science and Veterinary College, Weifang 261061, PR China
| | - Zhaofeng Sui
- Shandong Vocational Animal Science and Veterinary College, Weifang 261061, PR China
| | - Aimin Ge
- Shandong Vocational Animal Science and Veterinary College, Weifang 261061, PR China
| | - Liuting Pan
- Shandong Vocational Animal Science and Veterinary College, Weifang 261061, PR China
| | - Kuan Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, PR China
| | - Yanjun Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, PR China
| | - Guangzhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, PR China
| | - Liwei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, PR China.
| | - Fei Gao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
15
|
Cruz Amaya J, Walcheck B, Smith-Gagen J, Lombardi VC, Hudig D. Detection of Antibody-Dependent Cell-Mediated Cytotoxicity-Supporting Antibodies by NK-92-CD16A Cell Externalization of CD107a: Recognition of Antibody Afucosylation and Assay Optimization. Antibodies (Basel) 2023; 12:44. [PMID: 37489366 PMCID: PMC10366760 DOI: 10.3390/antib12030044] [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: 05/24/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) by natural killer (NK) lymphocytes eliminates cells infected with viruses. Anti-viral ADCC requires three components: (1) antibody; (2) effector lymphocytes with the Fc-IgG receptor CD16A; and (3) viral proteins in infected cell membranes. Fc-afucosylated antibodies bind with greater affinity to CD16A than fucosylated antibodies; individuals' variation in afucosylation contributes to differences in ADCC. Current assays for afucosylated antibodies involve expensive methods. We report an improved bioassay for antibodies that supports ADCC, which encompasses afucosylation. This assay utilizes the externalization of CD107a by NK-92-CD16A cells after antibody recognition. We used anti-CD20 monoclonal antibodies, GA101 WT or glycoengineered (GE), 10% or ~50% afucosylated, and CD20-positive Raji target cells. CD107a increased detection 7-fold compared to flow cytometry to detect Raji-bound antibodies. WT and GE antibody effective concentrations (EC50s) for CD107a externalization differed by 20-fold, with afucosylated GA101-GE more detectable. The EC50s for CD107a externalization vs. 51Cr cell death were similar for NK-92-CD16A and blood NK cells. Notably, the % CD107a-positive cells were negatively correlated with dead Raji cells and were nearly undetectable at high NK:Raji ratios required for cytotoxicity. This bioassay is very sensitive and adaptable to assess anti-viral antibodies but unsuitable as a surrogate assay to monitor cell death after ADCC.
Collapse
Affiliation(s)
- Judith Cruz Amaya
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Bruce Walcheck
- Department of Veterinary and Biological Sciences, Center for Immunology and Masonic Cancer Center, University of Minnesota, 295J AS/VM Building, 1988 Fitch Avenue, Saint Paul, MN 55108, USA
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Vincent C. Lombardi
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Dorothy Hudig
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| |
Collapse
|
16
|
Suzuki Y. Interferon-induced restriction of Chikungunya virus infection. Antiviral Res 2023; 210:105487. [PMID: 36657882 DOI: 10.1016/j.antiviral.2022.105487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Chikungunya virus (CHIKV) is an enveloped RNA virus that causes Chikungunya fever (CHIKF), which is transmitted to humans through the bite of infected Aedes mosquitos. Although CHIKVF had been regarded as an endemic disease in limited regions of Africa and Asia, the recent global reemergence of CHIKV heightened awareness of this infectious disease, and CHIKV infection is currently considered an increasing threat to public health. However, no specific drug or licensed vaccine is available for CHIKV infection. As seen in other RNA virus infections, CHIKV triggers the interferon (IFN) response that plays a central role in host defense against pathogens. Experimental evidence has demonstrated that control of CHIVK replication by the IFN response is achieved by antiviral effector molecules called interferon-stimulated genes (ISGs), whose expressions are upregulated by IFN stimulation. This review details the molecular basis of the IFN-mediated suppression of CHIKV, particularly the ISGs restricting CHIKV replication.
Collapse
Affiliation(s)
- Youichi Suzuki
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan.
| |
Collapse
|