1
|
Bajinka O, Ouedraogo SY, Li N, Zhan X. Multiomics as instrument to promote 3P medical approaches for the overall management of respiratory syncytial viral infections. EPMA J 2025; 16:217-238. [PMID: 39991100 PMCID: PMC11842696 DOI: 10.1007/s13167-024-00395-z] [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: 10/26/2024] [Accepted: 12/29/2024] [Indexed: 02/25/2025]
Abstract
Respiratory syncytial viral (RSV) infection is a leading persisting pulmonary disease-causing agent. It causes loss of lives especially among infants, old ages, and adults immunocompromised individuals. This viral pathogen infects children more especially those under the age of 2 and may lead to death. It causes 3 million hospitalizations and up to 60,000 deaths annually for under the age of 5. The most vulnerable are immunocompromised individuals and asthmatic children with suboptimal antiviral defenses. It is associated with bronchiolitis, pneumonia, and bronchopneumonia. Despite all the current interventions and clinical trials, the only available therapeutic strategies for this viral infection are palliative care. Therefore, it is imperative to understand the pathogenicity of RSV and the corresponding host immune response to depict a sort of a targeted intervention. With the increasingly cutting-edge methods in harnessing the pathogenicity of this viral infection, high throughput systems including omics technological advances are at the spotlight. For instance, the associated genes with RSV complications for the host, the set of microbiome identified as operational taxonomic unit, the upregulated or downregulated metabolites, the protein subtypes, and the small molecules can help explain the viral microenvironment. Moreover, these big data will lead to RSV patients' stratification through individualized patient profiles that will bring in targeted prevention and treatment algorithms tailored to individualized patients' profiles. Through this, the virus and host interactions based on the pathogenicity of infection will provide a strong ground for depicting the prevention, prediction, and personalized medicine (3PM) for RSV. The 3PM approach brought cutting edge functional medicine to the healthcare givers, thus conferring targeted prevention and precision medicine while observing personalized treatment as well as preventive regularities. The viral replication mechanisms against the host defense mechanisms are crucial for the development of safe and effective therapy. Integrative personal omics profiles, whose analysis is based on the combined proteomics, transcriptomics, genomics, proteoformics, metabolomics, and autoantibody profiles, are very robust for predicting the risk of RSV infection. The targeted prevention will emerge from the patient stratification when the diagnosis is accurately predicted. In addition, the personalized medical services will give an effective prognostic assessment for RSV complications.
Collapse
Affiliation(s)
- Ousman Bajinka
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Serge Yannick Ouedraogo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
2
|
Kierbiedź-Guzik N, Sozańska B. The Molecular Basis of Asthma Exacerbations Triggered by Viral Infections: The Role of Specific miRNAs. Int J Mol Sci 2024; 26:120. [PMID: 39795977 PMCID: PMC11720134 DOI: 10.3390/ijms26010120] [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: 11/30/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Viral respiratory infections are a significant clinical problem among the pediatric population and are one of the leading causes of hospitalization. Most often, upper respiratory tract infections are self-limiting. Still, those that involve the lower respiratory tract are usually associated with asthma exacerbations, leading to worsening or even the initiation of the disease. A key role in regulating the immune response and inflammation during viral infections and their impact on the progression of asthma has been demonstrated for miRNA molecules (microRNA). Their interaction with mRNA (messenger RNA) regulates gene expression in innate and acquired immune responses, making them valuable biomarkers for diagnostics, monitoring, and predicting asthma exacerbations. The following paper presents changes in the expression of miRNAs during the five most common viral infections causing asthma worsening, with particular emphasis on the pediatric population. In addition, we describe the molecular mechanisms by which miRNAs influence the pathogenesis of viral infection, immune responses, and asthma exacerbations. These molecules represent promising targets for future innovative therapeutic strategies, paving the way for developing personalized medicine for patients with viral-induced asthma exacerbations.
Collapse
Affiliation(s)
- Natalia Kierbiedź-Guzik
- Department and Clinic of Paediatrics, Allergology and Cardiology, Wroclaw Medical University, ul. Chałubińskiego 2a, 50-368 Wrocław, Poland
| | | |
Collapse
|
3
|
Yang TN, Wang YX, Jian PA, Ma XY, Ren YF, Huang NN, Li XN, Li JL. Rab8a Is a Key Target That Melatonin Prevents Lipid Disorder from Atrazine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23511-23519. [PMID: 39382334 DOI: 10.1021/acs.jafc.4c07006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Atrazine (ATZ), a widely used herbicide, disrupts mitochondrial function and lipid metabolism in the liver. Melatonin (MLT), a naturally synthesized hormone, combats mitochondrial dysfunction and alleviates lipid toxicity. However, the mechanisms behind ATZ-induced lipid metabolism toxicity and the protective effects of MLT remain unexplored. Mice were randomly assigned to four groups: control (Con), 5 mg/kg MLT, 170 mg/kg ATZ, and a cotreatment group receiving 170 mg/kg ATZ with 5 mg/kg MLT (ATZ+MLT). Additionally, we analyzed the effects of MLT and Rab8a on mRNA and proteins related to mitochondrial function and lipid metabolism disrupted by ATZ in AML12 cells. In conclusion, ATZ induced mitochondrial stress and disrupted fatty acid metabolism in mouse hepatocytes and AML12 cells. Exogenous MLT restores Rab8a levels, regulating fatty acid utilization in mitochondria and mitochondrial function. Notably, targeting Rab8a does not significantly affect mitochondrial function but prevents ATZ-induced lipid metabolism disorders in hepatocytes.
Collapse
Affiliation(s)
- Tian-Ning Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Yu-Xiang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Ping-An Jian
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Xiang-Yu Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Yi-Fei Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Ning-Ning Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P.R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P.R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| |
Collapse
|
4
|
Yang M, Zhang X, Liu Q, Wang Y. Network pharmacology, molecular docking, and untargeted metabolomics reveal molecular mechanisms of multi-targets effects of Qingfei Tongluo Plaster improving respiratory syncytial virus pneumonia. CHINESE HERBAL MEDICINES 2024; 16:638-655. [PMID: 39606255 PMCID: PMC11589485 DOI: 10.1016/j.chmed.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 02/20/2024] [Accepted: 07/26/2024] [Indexed: 11/29/2024] Open
Abstract
Objective Qingfei Tongluo Plaster (QFP), an improved Chinese medicine hospital preparation, is an attractive treatment option due to its well clinical efficacy, convenience, economy, and patient compliance in the treatment of respiratory syncytial virus (RSV) pneumonia. The aim of this study was to investigate the efficacy mechanism of QFP on RSV rats from the perspective of alleviating lung inflammation and further explore the changes of serum metabolites and metabolic pathways in RSV rats under the influence of QFP. Methods This study used network pharmacological methods and molecular docking combined with molecular biology and metabolomics from multi-dimensional perspectives to screen and verify the therapeutic targets. Open online databases were used to speculate the gene targets of efficient ingredients and diseases. Then, we used the String database to examine the fundamental interaction of common targets of drugs and diseases. An online enrichment analysis was performed to predict the functional pathways. Molecular docking was applied to discover the binding modes between essential ingredients and crucial gene targets. Finally, we demonstrated the anti-inflammatory ability of QFP in the RSV-evoked pneumonia rat model and explained the mechanism in combination with the metabolomics results. Results There were 19 critical targets defined as the core targets: tumor necrosis factor (TNF), inducible nitric oxide synthase 2 (NOS2), mitogen-activated protein kinase 14 (MAPK14), g1/S-specific cyclin-D1 (CCND1), signal transducer and activator of transcription 1-alpha/beta (STAT1), proto-oncogene tyrosine-protein kinase Src (SRC), cellular tumor antigen p53 (TP53), interleukin-6 (IL6), hypoxia-inducible factor 1-alpha (HIF1A), RAC-alpha serine/threonine-protein kinase (AKT1), signal transducer and activator of transcription 3 (STAT3), heat shock protein HSP 90-alpha (HSP90AA1), tyrosine-protein kinase JAK2 (JAK2), cyclin-dependent kinase inhibitor 1 (CDKN1A), mitogen-activated protein kinase 3 (MAPK3), epidermal growth factor receptor (EGFR), myc proto-oncogene protein (MYC), protein c-Fos (FOS) and transcription factor p65 (RELA). QFP treated RSV pneumonia mainly through the phosphatidylinositol 3-kinase (PI3K)/RAC AKT pathway, HIF-1 pathway, IL-17 pathway, TNF pathway, and MAPK pathway. Animal experiments proved that QFP could effectively ameliorate RSV-induced pulmonary inflammation. A total of 28 metabolites underwent significant changes in the QFP treatment, and there are four metabolic pathways consistent with the KEGG pathway analyzed by network pharmacology, suggesting that they may be critical processes related to treatment. Conclusion These results provide essential perspicacity into the mechanisms of action of QFP as a promising anti-RSV drug.
Collapse
Affiliation(s)
- Mengfei Yang
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
| | - Xiuying Zhang
- Department of Pediatrics, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Qing Liu
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
| | - Yongxue Wang
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
| |
Collapse
|
5
|
Farías MA, Diethelm-Varela B, Kalergis AM, González PA. Interplay between lipid metabolism, lipid droplets and RNA virus replication. Crit Rev Microbiol 2024; 50:515-539. [PMID: 37348003 DOI: 10.1080/1040841x.2023.2224424] [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: 02/23/2022] [Revised: 09/20/2022] [Accepted: 01/29/2023] [Indexed: 06/24/2023]
Abstract
Lipids play essential roles in the cell as components of cellular membranes, signaling molecules, and energy storage sources. Lipid droplets are cellular organelles composed of neutral lipids, such as triglycerides and cholesterol esters, and are also considered as cellular energy reserves, yet new functions have been recently associated with these structures, such as regulators of oxidative stress and cellular lipotoxicity, as well as modulators of pathogen infection through immune regulation. Lipid metabolism and lipid droplets participate in the infection process of many RNA viruses and control their replication and assembly, among others. Here, we review and discuss the contribution of lipid metabolism and lipid droplets over the replication cycle of RNA viruses, altogether pointing out potentially new pharmacological antiviral targets associated with lipid metabolism.
Collapse
Affiliation(s)
- Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Benjamín Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
6
|
Jia X, Crawford JC, Gebregzabher D, Monson EA, Mettelman RC, Wan Y, Ren Y, Chou J, Novak T, McQuilten HA, Clarke M, Bachem A, Foo IJ, Fritzlar S, Carrera Montoya J, Trenerry AM, Nie S, Leeming MG, Nguyen THO, Kedzierski L, Littler DR, Kueh A, Cardamone T, Wong CY, Hensen L, Cabug A, Laguna JG, Agrawal M, Flerlage T, Boyd DF, Van de Velde LA, Habel JR, Loh L, Koay HF, van de Sandt CE, Konstantinov IE, Berzins SP, Flanagan KL, Wakim LM, Herold MJ, Green AM, Smallwood HS, Rossjohn J, Thwaites RS, Chiu C, Scott NE, Mackenzie JM, Bedoui S, Reading PC, Londrigan SL, Helbig KJ, Randolph AG, Thomas PG, Xu J, Wang Z, Chua BY, Kedzierska K. High expression of oleoyl-ACP hydrolase underpins life-threatening respiratory viral diseases. Cell 2024; 187:4586-4604.e20. [PMID: 39137778 DOI: 10.1016/j.cell.2024.07.026] [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: 07/07/2023] [Revised: 03/07/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024]
Abstract
Respiratory infections cause significant morbidity and mortality, yet it is unclear why some individuals succumb to severe disease. In patients hospitalized with avian A(H7N9) influenza, we investigated early drivers underpinning fatal disease. Transcriptomics strongly linked oleoyl-acyl-carrier-protein (ACP) hydrolase (OLAH), an enzyme mediating fatty acid production, with fatal A(H7N9) early after hospital admission, persisting until death. Recovered patients had low OLAH expression throughout hospitalization. High OLAH levels were also detected in patients hospitalized with life-threatening seasonal influenza, COVID-19, respiratory syncytial virus (RSV), and multisystem inflammatory syndrome in children (MIS-C) but not during mild disease. In olah-/- mice, lethal influenza infection led to survival and mild disease as well as reduced lung viral loads, tissue damage, infection-driven pulmonary cell infiltration, and inflammation. This was underpinned by differential lipid droplet dynamics as well as reduced viral replication and virus-induced inflammation in macrophages. Supplementation of oleic acid, the main product of OLAH, increased influenza replication in macrophages and their inflammatory potential. Our findings define how the expression of OLAH drives life-threatening viral disease.
Collapse
Affiliation(s)
- Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jeremy Chase Crawford
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Deborah Gebregzabher
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ebony A Monson
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robert C Mettelman
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yanmin Wan
- Shanghai Public Health Clinical Centre and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Yanqin Ren
- Shanghai Public Health Clinical Centre, Fudan University, Shanghai 201508, China
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tanya Novak
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital and Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Michele Clarke
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Isabelle J Foo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Julio Carrera Montoya
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Alice M Trenerry
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Dene R Littler
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Andrew Kueh
- Walter Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Tina Cardamone
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Aira Cabug
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jaime Gómez Laguna
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group, University of Córdoba, International Excellence Agrifood Campus "CeiA3", 14014 Córdoba, Spain
| | - Mona Agrawal
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David F Boyd
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Lee-Ann Van de Velde
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Carolien E van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Igor E Konstantinov
- Department of Cardiothoracic Surgery, Royal Children's Hospital, University of Melbourne, Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Parkville, VIC 3052, Australia
| | - Stuart P Berzins
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Katie L Flanagan
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS 7248, Australia; School of Health and Biomedical Science, RMIT University, Bundoora, VIC 3083, Australia; Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Marco J Herold
- Walter Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Amanda M Green
- Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Institute of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Karla J Helbig
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Adrienne G Randolph
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital and Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA
| | - Jianqing Xu
- Shanghai Public Health Clinical Centre and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Zhongfang Wang
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China.
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA.
| |
Collapse
|
7
|
Hsia JZ, Liu D, Haynes L, Cruz-Cosme R, Tang Q. Lipid Droplets: Formation, Degradation, and Their Role in Cellular Responses to Flavivirus Infections. Microorganisms 2024; 12:647. [PMID: 38674592 PMCID: PMC11051834 DOI: 10.3390/microorganisms12040647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Lipid droplets (LDs) are cellular organelles derived from the endoplasmic reticulum (ER), serving as lipid storage sites crucial for maintaining cellular lipid homeostasis. Recent attention has been drawn to their roles in viral replication and their interactions with viruses. However, the precise biological functions of LDs in viral replication and pathogenesis remain incompletely understood. To elucidate the interaction between LDs and viruses, it is imperative to comprehend the biogenesis of LDs and their dynamic interactions with other organelles. In this review, we explore the intricate pathways involved in LD biogenies within the cytoplasm, encompassing the uptake of fatty acid from nutrients facilitated by CD36-mediated membranous protein (FABP/FATP)-FA complexes, and FA synthesis via glycolysis in the cytoplasm and the TCL cycle in mitochondria. While LD biogenesis primarily occurs in the ER, matured LDs are intricately linked to multiple organelles. Viral infections can lead to diverse consequences in terms of LD status within cells post-infection, potentially involving the breakdown of LDs through the activation of lipophagy. However, the exact mechanisms underlying LD destruction or accumulation by viruses remain elusive. The significance of LDs in viral replication renders them effective targets for developing broad-spectrum antivirals. Moreover, considering that reducing neutral lipids in LDs is a strategy for anti-obesity treatment, LD depletion may not pose harm to cells. This presents LDs as promising antiviral targets for developing therapeutics that are minimally or non-toxic to the host.
Collapse
Affiliation(s)
| | | | | | | | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (J.Z.H.); (D.L.); (L.H.); (R.C.-C.)
| |
Collapse
|
8
|
Lu X, Li G, Liu Y, Luo G, Ding S, Zhang T, Li N, Geng Q. The role of fatty acid metabolism in acute lung injury: a special focus on immunometabolism. Cell Mol Life Sci 2024; 81:120. [PMID: 38456906 PMCID: PMC10923746 DOI: 10.1007/s00018-024-05131-4] [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: 11/09/2023] [Revised: 01/06/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Reputable evidence from multiple studies suggests that excessive and uncontrolled inflammation plays an indispensable role in mediating, amplifying, and protracting acute lung injury (ALI). Traditionally, immunity and energy metabolism are regarded as separate functions regulated by distinct mechanisms, but recently, more and more evidence show that immunity and energy metabolism exhibit a strong interaction which has given rise to an emerging field of immunometabolism. Mammalian lungs are organs with active fatty acid metabolism, however, during ALI, inflammation and oxidative stress lead to a series metabolic reprogramming such as impaired fatty acid oxidation, increased expression of proteins involved in fatty acid uptake and transport, enhanced synthesis of fatty acids, and accumulation of lipid droplets. In addition, obesity represents a significant risk factor for ALI/ARDS. Thus, we have further elucidated the mechanisms of obesity exacerbating ALI from the perspective of fatty acid metabolism. To sum up, this paper presents a systematical review of the relationship between extensive fatty acid metabolic pathways and acute lung injury and summarizes recent advances in understanding the involvement of fatty acid metabolism-related pathways in ALI. We hold an optimistic believe that targeting fatty acid metabolism pathway is a promising lung protection strategy, but the specific regulatory mechanisms are way too complex, necessitating further extensive and in-depth investigations in future studies.
Collapse
Affiliation(s)
- Xiao Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Tianyu Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
9
|
Tan YJ, Jin Y, Zhou J, Yang YF. Lipid droplets in pathogen infection and host immunity. Acta Pharmacol Sin 2024; 45:449-464. [PMID: 37993536 PMCID: PMC10834987 DOI: 10.1038/s41401-023-01189-1] [Citation(s) in RCA: 1] [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: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
As the hub of cellular lipid metabolism, lipid droplets (LDs) have been linked to a variety of biological processes. During pathogen infection, the biogenesis, composition, and functions of LDs are tightly regulated. The accumulation of LDs has been described as a hallmark of pathogen infection and is thought to be driven by pathogens for their own benefit. Recent studies have revealed that LDs and their subsequent lipid mediators contribute to effective immunological responses to pathogen infection by promoting host stress tolerance and reducing toxicity. In this comprehensive review, we delve into the intricate roles of LDs in governing the replication and assembly of a wide spectrum of pathogens within host cells. We also discuss the regulatory function of LDs in host immunity and highlight the potential for targeting LDs for the diagnosis and treatment of infectious diseases.
Collapse
Affiliation(s)
- Yan-Jie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Yi Jin
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yun-Fan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
10
|
Ruan P, Dai P, Mao Y, Tang Z, He H, Wu G, Qin L, Tan Y. The in vitro and in vivo antiviral effects of IGF1R inhibitors against respiratory syncytial virus infection. J Biomol Struct Dyn 2024:1-12. [PMID: 38299600 DOI: 10.1080/07391102.2024.2309643] [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: 08/11/2023] [Accepted: 01/18/2024] [Indexed: 02/02/2024]
Abstract
The insulin-like growth factor 1 receptor (IGF1R) was recognized as a pivotal receptor that facilitated the cellular entry of RSV. Small molecule inhibitors designed to target IGF1R exhibited potential as potent antiviral agents. Through virtual screening, we conducted a screening process involving small molecule compounds derived from natural products, aiming to target the IGF1R protein against respiratory syncytial virus infection. The molecular dynamics simulation analysis showed that tannic acid and daptomycin interacted with the IGF1R. The experimental results in vivo and in vitro showed that tannic acid and daptomycin had anti-RSV infection potential through reducing viral loads, inflammation, airway resistance and protecting alveolar integrity. The CC50 values of tannic acid and daptomycin were 6 nM and 0.45 μM, respectively. Novel small-molecule inhibitors targeting the IGF1R, tannic acid and daptomycin, may be effective anti-RSV therapy agents. This study may in future broaden the arsenal of therapeutics for use against RSV infection and lead to more effective care against the virus.
Collapse
Affiliation(s)
- Pinglang Ruan
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Pei Dai
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Second Department of Laboratory, Hunan Provincial People's Hospital (The First Affifiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Yu Mao
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhongxiang Tang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hanlin He
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guojun Wu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ling Qin
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yurong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Herrera-Moro Huitron L, De Jesús-González LA, Martínez-Castillo M, Ulloa-Aguilar JM, Cabello-Gutierrez C, Helguera-Repetto C, Garcia-Cordero J, León Juárez M. Multifaceted Nature of Lipid Droplets in Viral Interactions and Pathogenesis. Microorganisms 2023; 11:1851. [PMID: 37513023 PMCID: PMC10386712 DOI: 10.3390/microorganisms11071851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Once regarded as inert organelles with limited and ill-defined roles, lipid droplets (LDs) have emerged as dynamic entities with multifaceted functions within the cell. Recent research has illuminated their pivotal role as primary energy reservoirs in the form of lipids, capable of being metabolized to meet cellular energy demands. Their high dynamism is underscored by their ability to interact with numerous cellular organelles, notably the endoplasmic reticulum (the site of LD genesis) and mitochondria, which utilize small LDs for energy production. Beyond their contribution to cellular bioenergetics, LDs have been associated with viral infections. Evidence suggests that viruses can co-opt LDs to facilitate their infection cycle. Furthermore, recent discoveries highlight the role of LDs in modulating the host's immune response. Observations of altered LD levels during viral infections suggest their involvement in disease pathophysiology, potentially through production of proinflammatory mediators using LD lipids as precursors. This review explores these intriguing aspects of LDs, shedding light on their multifaceted nature and implications in viral interactions and disease development.
Collapse
Affiliation(s)
- Luis Herrera-Moro Huitron
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | | | - Macario Martínez-Castillo
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - José Manuel Ulloa-Aguilar
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Carlos Cabello-Gutierrez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Departamento de Investigación en Virología y Micología, Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico
| | - Cecilia Helguera-Repetto
- Laboratorio de Microbiología y Diagnóstico Molecular, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Julio Garcia-Cordero
- Departamento de Biomedicina Molecular, Cinvestav, Av. IPN# 2508, Mexico City 07360, Mexico
| | - Moisés León Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| |
Collapse
|
12
|
Qu Y, Wang W, Xiao MZX, Zheng Y, Liang Q. The interplay between lipid droplets and virus infection. J Med Virol 2023; 95:e28967. [PMID: 37496184 DOI: 10.1002/jmv.28967] [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: 05/31/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023]
Abstract
As an intracellular parasite, the virus usurps cellular machinery and modulates cellular metabolism pathways to replicate itself in cells. Lipid droplets (LDs) are universally conserved energy storage organelles that not only play vital roles in maintaining lipid homeostasis but are also involved in viral replication. Increasing evidence has demonstrated that viruses take advantage of cellular lipid metabolism by targeting the biogenesis, hydrolysis, and lipophagy of LD during viral infection. In this review, we summarize the current knowledge about the modulation of cellular LD by different viruses, with a special emphasis on the Hepatitis C virus, Dengue virus, and SARS-CoV-2.
Collapse
Affiliation(s)
- Yafei Qu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weili Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maggie Z X Xiao
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai University of Traditional Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Medicine, Shanghai, China
| | - Qiming Liang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Liu D, Tang Z, Bajinka O, Dai P, Wu G, Qin L, Tan Y. miR-34b/c-5p/CXCL10 Axis Induced by RSV Infection Mediates a Mechanism of Airway Hyperresponsive Diseases. BIOLOGY 2023; 12:biology12020317. [PMID: 36829591 PMCID: PMC9953223 DOI: 10.3390/biology12020317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Background: RSV is closely correlated with post-infection airway hyperresponsive diseases (AHD), but the mechanism remains unclear. Objective: Due to the pivotal role of miRNAs in AHD, we analyzed the differentially expressed miRNAs (DEmiRs) in RSV-infected patients, asthma patients, and COPD patients from public datasets and explored the mechanisms of association between RSV and AHD. Methods: We obtained miRNA and mRNA databases of patients with RSV infection, as well as miRNA databases of asthma and COPD patients from the GEO database. Through integrated analysis, we screened DEmiRs and DEGs. Further analysis was carried out to obtain the hub genes through the analysis of biological pathways and enrichment pathways of DEGs targeted by DEmiRs and the construction of a protein-protein interaction (PPI) network. Results: The five differential molecules (miR-34b/c-5p, Cd14, Cxcl10, and Rhoh) were verified through in vivo experiments that had the same expression trend in the acute and chronic phases of RSV infection. Following infection of BEAS-2B cells with RSV, we confirmed that RSV infection down-regulated miR-34b/c-5p, and up-regulated the expression levels of CXCL10 and CD14. Furthermore, the results of the dual-luciferase reporter assay showed that CXCL10 was the target of hsa-miR-34c-5p. Conclusions: miR-34b/c-5p/CXCL10 axis mediates a mechanism of AHD.
Collapse
Affiliation(s)
- Dan Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China
- Hunan Provincial Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Zhongxiang Tang
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha 410078, China
| | - Ousman Bajinka
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha 410078, China
| | - Pei Dai
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha 410078, China
| | - Guojun Wu
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
| | - Ling Qin
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Central South University, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China
- Hunan Provincial Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha 410078, China
- Correspondence: (L.Q.); (Y.T.)
| | - Yurong Tan
- Department of Medical Microbiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha 410078, China
- Correspondence: (L.Q.); (Y.T.)
| |
Collapse
|
14
|
Shi P, Nian D, Qu H, Ye M, Wang C, Li L, Li Q. B.infantis enhances immunotherapy for Guillain-Barre syndrome through regulating PD-1. BMC Neurol 2023; 23:48. [PMID: 36709251 PMCID: PMC9883859 DOI: 10.1186/s12883-022-03046-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 12/28/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Guillain-Barré syndrome (GBS) is a rare, autoimmune disease. B.infantis is reported to be effective in alleviating GBS by regulating abnormal function of T helper (Th) cells. OBJECTIVES In this study, T cells were isolated from healthy and GBS patients. The therapeutic effect of Bifidobacterium infantis (B.infantis) and whether it is achieved by PD-1 was examined at cellular and animal models. METHODS We used CCK-8, flow cytometry and real-time PCR to determine the differentiation of T cell subsets at cellular level. Then, an experimental autoimmune neuritis (EAN) animal model using six-week SD rats (n = 30, male) weighing 180-200 g was established to support the role of B. infantis in GBS through PD-1. RESULTS B. infantis inhibited the proliferation and promoted apoptosis of T cells from GBS. At the same time, the expression levels of PD-1 increased, which was correlated with decreased T-bet (Th1) and ROR-γt (Th17) and increased Foxp3 (Treg) expression. Moreover, B. infantis alleviated the symptoms of GBS. Th1 and Th17 cells decreased while Treg cells increased after B. infantis treatment, which could be partly abrogated by PD-1 inhibitor. CONCLUSIONS We concluded from this study that B.infantis alleviated GBS partly through PD-1.
Collapse
Affiliation(s)
- Peng Shi
- grid.414884.5Department of Neurology, first Affiliated Hospital of Bengbu Medical College, Bengbu, 233004 Anhui China
| | - Di Nian
- grid.252957.e0000 0001 1484 5512Department of Medical Examination, Bengbu Medical College, Bengbu, 233030 Anhui China
| | - Hongdang Qu
- grid.414884.5Department of Neurology, first Affiliated Hospital of Bengbu Medical College, Bengbu, 233004 Anhui China
| | - Ming Ye
- grid.414884.5Department of Neurology, first Affiliated Hospital of Bengbu Medical College, Bengbu, 233004 Anhui China
| | - Chun Wang
- grid.414884.5Department of Neurology, first Affiliated Hospital of Bengbu Medical College, Bengbu, 233004 Anhui China
| | - Li Li
- grid.414884.5Department of Neurology, first Affiliated Hospital of Bengbu Medical College, Bengbu, 233004 Anhui China
| | - Qian Li
- grid.414884.5Department of Neurology, first Affiliated Hospital of Bengbu Medical College, Bengbu, 233004 Anhui China
| |
Collapse
|
15
|
He H, Tan Y, Tang Z, Wang L, Liu S, Wu G. ADAM9: A regulator between HCMV infection and function of smooth muscle cells. J Med Virol 2023; 95:e28352. [PMID: 36437481 DOI: 10.1002/jmv.28352] [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: 07/22/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
Lots of epidemiological and clinical studies have shown that human cytomegalovirus (HCMV) is related to the pathogenesis of atherosclerosis. Released by inflammatory cells and vascular smooth muscle cell (VSMCs), metalloproteinases are observed in many pathological vessel conditions, including atherosclerosis and restenosis. This study was designed to investigate the effect of HCMV infection on the expression of metalloproteinases and their involvements in the HCMV-induced functional changes of VSMCs. Differential metalloproteinase after HCMV infection was assayed using reverse transcription-polymerase chain reaction (RT-PCR) microarray. The most significant increased a disintegrin and metalloprotease 9 (ADAM9) was chosen to investigate the mechanism of its specific increase after infection using the treatment of UV-irradiated replication-deficient HCMV, HCMV-infected cell lysate filters or Foscarnet. The function of proliferation, migration, production of inflammatoty factors and phenotypic transformation were determined by using cell counting kit-8, transwell, Enzyme-linked immunosorbent assay, RT-quantitative PCR (qPCR) and Western blot, respectively. Moreover, the effect of ADAM9 deficiency on HCMV replication was also determined using RT-qPCR and immunofluorescence. The expression levels of 6 genes were upregulated and 14 genes were downregulated at different time points after HCMV infection. Among these, the expression level of ADAM9 increased most significantly at each time point and the abnormal expression of ADAM9 might be induced by the early gene products of HCMV. Further studies found that ADAM9 promoted the proliferation, the migration, the production of inflammatory factors and the transit from the contractile phenotype (decreased ACTA2 expression) to the synthetic phenotype (increased osteopontin [OPN] expression). Knockdown theADAM9 expression could rescue the decreased ACTA2 expression, but has no effect on OPN expression. ADAM-9 deficiency didn't affect the replication of HCMV. The findings of our study suggest that HCMV infection changed VSMC function through ADAM9 expression, which may contribute to the understanding of the underlying pathological mechanisms of HCMV-induced atherosclerosis.
Collapse
Affiliation(s)
- Hanlin He
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yurong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhongxiang Tang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lili Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shuiping Liu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guojun Wu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,School of Basic Medical Sciences, China-Africa Research Centre of Infectious Diseases, Central South University, Changsha, Hunan, China
| |
Collapse
|
16
|
Mao Y, Bajinka O, Tang Z, Qiu X, Tan Y. Lung-brain axis: Metabolomics and pathological changes in lungs and brain of respiratory syncytial virus-infected mice. J Med Virol 2022; 94:5885-5893. [PMID: 35945613 DOI: 10.1002/jmv.28061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/04/2022] [Accepted: 08/06/2022] [Indexed: 01/06/2023]
Abstract
The lung-brain axis is an emerging area of study that got its basis from the gut-brain axis biological pathway. Using Respiratory Synctial Virus (RSV) as the model of respiratory viral pathogen, this study aims to establish some biological pathways. After establishing the mice model, the inflammation in lung and brain were assayed using Hematoxylin-eosin staining, indirect immunofluorescence (IFA), and quantitative reverse-transcription polymerase chain reaction. The biological pathways between lung and brain were detected through metabolomics analysis. In lung, RSV infection promoted epithelial shedding and infiltration of inflammatory cells. Also, RSV immunofluorescence and titerss were significantly increased. Moreover, interleukin (IL)-1, IL-6 and tumor necrosis factor-α (TNF-α) were also significantly increased after RSV infection. In brain, the cell structure of hippocampal CA1 area was loose and disordered. Inflammatory cytokines IL-6 and IL-1β expression in the brain also increased, however, TNF-α expression showed no differences among the control and RSV group. We observed an increased expression of microglia biomarker IBA-1 and decreased neuronal biomarker NeuN. In addition, RSV mRNA expression levels were also increased in the brains. 15 metabolites were found upregulated in the RSV group including nerve-injuring metabolite glutaric acid, hydroxyglutaric acid and Spermine. ɑ-Estradiol increased significantly while normorphine decreased significantly at Day 7 of infection among the RSV group. This study established a mouse model for exploring the pathological changes in lungs and brains. There are many biological pathways between lung and brain, including direct translocation of RSV and metabolite pathway.
Collapse
Affiliation(s)
- Yu Mao
- Department of Medical Microbiology, School of Basic and Medical Sciences, Central South University, Changsha, Hunan Provinces, China.,China-Africa Research Center of Infectious Diseases, School of Basic and Medical Sciences, Central South University, Changsha, Hunan, China
| | - Ousman Bajinka
- Department of Medical Microbiology, School of Basic and Medical Sciences, Central South University, Changsha, Hunan Provinces, China.,China-Africa Research Center of Infectious Diseases, School of Basic and Medical Sciences, Central South University, Changsha, Hunan, China.,Department of Medicine, School of Medicine and Allied Health Sciences, University of The Gambia, Serekunda, Gambia
| | - Zhongxiang Tang
- Department of Medical Microbiology, School of Basic and Medical Sciences, Central South University, Changsha, Hunan Provinces, China.,China-Africa Research Center of Infectious Diseases, School of Basic and Medical Sciences, Central South University, Changsha, Hunan, China
| | - Xiangjie Qiu
- Department of Medical Microbiology, School of Basic and Medical Sciences, Central South University, Changsha, Hunan Provinces, China.,China-Africa Research Center of Infectious Diseases, School of Basic and Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yurong Tan
- Department of Medical Microbiology, School of Basic and Medical Sciences, Central South University, Changsha, Hunan Provinces, China.,China-Africa Research Center of Infectious Diseases, School of Basic and Medical Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
17
|
lncRNA STAT4-AS1 Inhibited TH17 Cell Differentiation by Targeting ROR γt Protein. J Immunol Res 2022; 2022:8307280. [PMID: 35528611 PMCID: PMC9071868 DOI: 10.1155/2022/8307280] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/05/2022] [Indexed: 11/18/2022] Open
Abstract
Objective From our previous study, we obtained long noncoding RNA (lncRNA) STAT4-AS1, which is related to asthma through high-throughput screening. However, we could not determine the specific mechanism involved and in response to this. We further designed this study. Results First, we found that lncRNA STAT4-AS1 was downregulated in T cells from patients with asthma when compared to healthy controls. Next, we confirmed that lncRNA STAT4-AS1 was significantly negatively correlated with T helper 17 (TH17) differentiation in vitro experiments. The decreases of STAT4-AS1 promoted TH17 differentiation, while the increases of STAT4-AS1 inhibited TH17 differentiation. Subsequently, through RNA pull-down, RNA-binding protein immunoprecipitation (RIP), and dual luciferase reporter assay, we found that STAT4-AS1 could inhibit the binding of retinoid-related orphan receptor-γt (RORγt) protein with an IL-17A promoter after binding with RORγt protein. Fluorescence in situ hybridization (FISH) and nuclear-cytoplasmic separation assay showed that STAT4-AS1 is bonded to RORγt in the cytoplasm, preventing RORγt from entering the nucleus. Conclusion Overall, STAT4-AS1 directly targets RORγt protein, inhibits the mutual binding of RORγt and IL-17 gene promoter, and eventually inhibits TH17 differentiation. To this end, STAT4-AS1 as a potential target may confer applications in the clinical treatment and diagnosis of TH17-related diseases.
Collapse
|
18
|
Liu B, Yang Q, Zhao L, Shui H, Si X. Vitamin D receptor gene polymorphism predicts left ventricular hypertrophy in maintenance hemodialysis. BMC Nephrol 2022; 23:32. [PMID: 35033017 PMCID: PMC8761333 DOI: 10.1186/s12882-021-02640-3] [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: 08/06/2021] [Accepted: 12/13/2021] [Indexed: 01/04/2023] Open
Abstract
Background To verify that the single nucleotide polymorphisms (SNP) of vitamin D receptor (VDR) may lead to genetic susceptibility to left ventricular hypertrophy (LVH), the present study was designed to study four SNPs of VDR associated with LVH in maintenance hemodialysis (MHD) patients of Han nationality. Methods 120 MHD patients were recruited at Department of Nephrology, Zhongnan Hospital of Wuhan University to analyze the expression of genotype, allele and haplotype of Fok I, Bsm I, Apa I and Taq I in blood samples, and to explore their correlation with blood biochemical indexes and ventricular remodeling. Results The results showed that the risks of CVD included gender, dialysis time, heart rate, SBP, glycated hemoglobin, calcium, iPTH and CRP concentration. Moreover, LAD, LVDd, LVDs, IVST and LVMI in B allele of Bsm I increased significantly. Fok I, Apa I and Taq I polymorphisms have no significant difference between MHD with LVH and without LVH. Further study showed that VDR expression level decreased significantly in MHD patients with LVH, and the B allele was positively correlated with VDR Expression. Conclusion VDR Bsm I gene polymorphism may predict cardiovascular disease risk of MDH patients, and provided theoretical basis for early detection and prevention of cardiovascular complications.
Collapse
Affiliation(s)
- Bingman Liu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Qingqing Yang
- Department of Nephrology, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Liangyu Zhao
- Department of Nephrology, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Hua Shui
- Department of Nephrology, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Xiaoyun Si
- Department of Nephrology, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China.
| |
Collapse
|
19
|
Luo X, Gong Y, Cai L, Zhang L, Dong X. Chemerin regulates autophagy to participate in polycystic ovary syndrome. J Int Med Res 2021; 49:3000605211058376. [PMID: 34816741 PMCID: PMC8647268 DOI: 10.1177/03000605211058376] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder in women of reproductive age. Chemerin has recently been discovered as a novel adipokine associated with obesity and metabolic syndrome. Excessive autophagy activity and overexpression of autophagy-related genes in follicular granulosa cells are important mechanisms of PCOS. This study aimed to investigate the effect of chemerin on autophagy in PCOS. METHODS A rat model of PCOS was established by subcutaneous injection of testosterone propionate under a high-fat diet. Expression levels of chemerin and its receptor CMKLR1 were determined by real-time polymerase chain reaction and western blot. Proliferation and apoptosis of human granulosa cells in vitro and expression of autophagy-related genes were examined using bafilomycin A1 (autophagy inhibitor) and Torin1 (autophagy inducer). RESULTS Chemerin and CMKLR1 expression were significantly increased in the ovary in a rat model of PCOS. Ectopic expression of chemerin promoted the proliferation and inhibited the apoptosis of COV434 cells. Ectopic expression of chemerin also induced autophagy by inhibiting the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway. CONCLUSIONS Chemerin and CMKLR1 were overexpressed in PCOS rats. Chemerin promoted autophagy through inhibiting the PI3K/Akt/mTOR pathway, and may provide a potential target and biomarker of PCOS.
Collapse
Affiliation(s)
- Xiaodong Luo
- Department of Obstetrics and Gynecology, 585250The Second Affiliated Hospital of Chongqing Medical University, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, P. R. China
| | - Yangyang Gong
- Department of Obstetrics and Gynecology, 585250The Second Affiliated Hospital of Chongqing Medical University, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, P. R. China
| | - Liuyun Cai
- Department of Obstetrics and Gynecology, 585250The Second Affiliated Hospital of Chongqing Medical University, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, P. R. China
| | - Lei Zhang
- Department of Obstetrics and Gynecology, 585250The Second Affiliated Hospital of Chongqing Medical University, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, P. R. China
| | - Xiaojing Dong
- Department of Obstetrics and Gynecology, 585250The Second Affiliated Hospital of Chongqing Medical University, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, P. R. China
| |
Collapse
|
20
|
Li W, Du X, Yang Y, Yuan L, Yang M, Qin L, Wang L, Zhou K, Xiang Y, Qu X, Liu H, Qin X, Xiao G, Liu C. miRNA-34b/c regulates mucus secretion in RSV-infected airway epithelial cells by targeting FGFR1. J Cell Mol Med 2021; 25:10565-10574. [PMID: 34636482 PMCID: PMC8581336 DOI: 10.1111/jcmm.16988] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/28/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection in airway epithelial cells is the main cause of bronchiolitis in children. Excessive mucus secretion is one of the primary symbols in RSV related lower respiratory tract infections (RSV-related LRTI). However, the pathological processes of mucus hypersecretion in RSV-infected airway epithelial cells remains unclear. The current study explores the involvement of miR-34b/miR-34c in mucus hypersecretion in RSV-infected airway epithelial cells by targeting FGFR1. First, miR-34b/miR-34c and FGFR1 mRNA were quantified by qPCR in throat swab samples and cell lines, respectively. Then, the luciferase reporters' assay was designed to verify the direct binding between FGFR1 and miR-34b/miR-34c. Finally, the involvement of AP-1 signalling was assessed by western blot. This study identified that miR-34b/miR-34c was involved in c-Jun-regulated MUC5AC production by targeting FGFR1 in RSV-infected airway epithelial cells. These results provide some useful insights into the molecular mechanisms of mucus hypersecretion which may also bring new potential strategies to improve mucus hypersecretion in RSV disease.
Collapse
Affiliation(s)
- Wenkai Li
- Department of PediatricsHunan Provincial People’s HospitalThe First Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Xizi Du
- Centre for Asthma and Respiratory DiseaseSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of Newcastle and Hunter Medical Research InstituteCallaghanNSWAustralia
- Department of Respiratory MedicineNational Clinical Research Center for Respiratory DiseasesXiangya HospitalCentral South UniversityChangshaChina
| | - Yu Yang
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Lin Yuan
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Ming Yang
- Centre for Asthma and Respiratory DiseaseSchool of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of Newcastle and Hunter Medical Research InstituteCallaghanNSWAustralia
| | - Ling Qin
- Department of Respiratory MedicineNational Clinical Research Center for Respiratory DiseasesXiangya HospitalCentral South UniversityChangshaChina
| | - Leyuan Wang
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Kai Zhou
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Yang Xiang
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Xiangping Qu
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Huijun Liu
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Xiaoqun Qin
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| | - Gelei Xiao
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaChina
| | - Chi Liu
- Department of Respiratory MedicineNational Clinical Research Center for Respiratory DiseasesXiangya HospitalCentral South UniversityChangshaChina
- Department of PhysiologySchool of Basic Medicine ScienceCentral South UniversityChangshaChina
| |
Collapse
|
21
|
Dai P, Tang Z, Ruan P, Bajinka O, Liu D, Tan Y. Gimap5 Inhibits Lung Cancer Growth by Interacting With M6PR. Front Oncol 2021; 11:699847. [PMID: 34604035 PMCID: PMC8479171 DOI: 10.3389/fonc.2021.699847] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Objective Several studies have demonstrated the impacts of GTPases of immunity-associated proteins (GIMAPs) on malignant cells. However, the mechanisms through which Gimap5 regulates lung cancer cells are yet to be thoroughly investigated in the literature. Our study aimed to investigate the function of Gimap5 in the development of lung cancer. Methods The expression levels of the GIMAP family were analyzed in lung cancer patients of various cancer databases and lung cancer cell lines. After the survival rates of the cells were analyzed, we constructed Gimap5 over-expressed lung cancer cell lines and assessed the effects of Gimap5 on cell migration, cell invasion, cell proliferation and the epithelial-mesenchymal transition (EMT). We later screened the interacting proteins of Gimap5 using Co-IP combined with mass spectrometry and then analyzed the expression and distribution of M6PR, including its impacts on protein-arginine deiminase type-4 (PADI4). Results Findings indicated that GIMAP family expression decreased significantly in lung cancer cell lines. We also noticed that the downregulation of the GIMAP family was related to the poor prognosis of lung cancer patients. Our experimental results showed that Gimap5 could inhibit the migration, invasion, proliferation and EMT of lung cancer cell lines. Moreover, we found that Gimap5 promoted the transport of M6PR from the cytoplasm to the cell membrane, thereby inhibiting the enhancement of EMT-related PADI4. Conclusion Our research suggested that Gimap5 could inhibit the growth of lung cancer by interacting with M6PR and that it could be a potential biomarker for the diagnosis and prognosis of lung cancer.
Collapse
Affiliation(s)
- Pei Dai
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhongxiang Tang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Pinglang Ruan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Ousman Bajinka
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China.,China-Africa Research Centre of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Dan Liu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yurong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China.,China-Africa Research Centre of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
22
|
RSV Promotes Epithelial Neuroendocrine Phenotype Differentiation through NODAL Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9956078. [PMID: 34541002 PMCID: PMC8445725 DOI: 10.1155/2021/9956078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 08/16/2021] [Indexed: 01/04/2023]
Abstract
Background Respiratory syncytial virus (RSV) infects infants and children, predisposing them to development of asthma during adulthood. Epithelial neuroendocrine phenotypes may be associated with development of asthma. This study hopes to ascertain if RSV infection promotes epithelial neuroendocrine phenotypes through the NODAL signaling pathway. Methods The GSE6802 data set was obtained from the GEO database, and the differential genes were analyzed using the R language. An in vitro model was constructed with RSV infected human respiratory epithelial cells, and then real-time qPCR and immunofluorescence were used to detect the expression of different epithelial biomarkers and airway neuropeptides. The acute and chronic infection model of RSV infection was established by intranasal injection of RSV into guinea pigs. Immunohistochemistry and Western blot were used to detect the expression of pulmonary neuroendocrine cells markers ENO2 and neuropeptides. Results The expression levels of ENO2, SP, CGRP, and NODAL/ACTRII were significantly higher in the RSV infection group than those of the control group, which were abrogated by siRNA-NODAL. In vivo, we found that the expression levels of ENO2, SP, and CGRP were significantly higher than that of the control group. Conclusion RSV promotes epithelial neuroendocrine phenotypes through the NODAL signaling pathway.
Collapse
|