1
|
Kehs Z, Cross AC, Li YM. From defense to disease: IFITM3 in immunity and Alzheimer's disease. Neurotherapeutics 2025; 22:e00482. [PMID: 39516072 DOI: 10.1016/j.neurot.2024.e00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Innate immunity protein interferon induced transmembrane protein 3 (IFITM3) is a transmembrane protein that has a wide array of functions, including in viral infections, Alzheimer's Disease (AD), and cancer. As an interferon stimulated gene (ISG), IFITM3's expression is upregulated by type-I, II, and III interferons. Moreover, the antiviral activity of IFITM3 is modulated by post-translational modifications. IFITM3 functions in innate immunity to disrupt viral fusion and entry to the plasma membrane as well as prevent viral escape from endosomes. As a γ-secretase modulatory protein, IFITM3 distinctly modulates the processing of amyloid precursor protein (APP) to generate amyloid beta peptides (Aβ) and Notch1 cleavages. Increased IFITM3 expression, which can result from aging, cytokine activation, inflammation, and infection, can lead to an upregulation of γ-secretase for Aβ production that causes a risk of AD. Therefore, the prevention of IFITM3 upregulation has potential in the development of novel therapies for the treatment of AD.
Collapse
Affiliation(s)
- Zoe Kehs
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Abigail C Cross
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA.
| |
Collapse
|
2
|
Yamamura R, Kinoshita M, Yasumizu Y, Yata T, Kihara K, Motooka D, Shiraishi N, Sugiyama Y, Beppu S, Murata H, Koizumi N, Sano I, Koda T, Okuno T, Mochizuki H. Transcriptome signature in the blood of neuromyelitis optica spectrum disorder under steroid tapering. Front Immunol 2025; 16:1508977. [PMID: 39963140 PMCID: PMC11830620 DOI: 10.3389/fimmu.2025.1508977] [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: 10/10/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Background The advent of biologics has significantly transformed treatment strategies for neuromyelitis optica spectrum disorder (NMOSD). However, there are no biomarkers that predict relapses associated with steroid tapering; therefore, it is critical to identify potential indicators of disease activity. In this study, we collected peripheral blood mononuclear cells (PBMCs) from NMOSD patients during steroid tapering and performed bulk RNA sequencing to analyze changes in immune dynamics caused by steroid reduction. Methods PBMCs were collected at 3-5 timepoints from 10 NMOSD patients at our hospital (including one relapse case), and bulk RNA sequencing was performed. All patients were positive for anti-AQP4 antibodies and had no history of biologic use. Results In one relapsed patient, gene groups with decreased expression at relapse were observed predominantly in monocytes, with upregulation in anti-inflammatory pathways such as IL-10, while the upregulated genes were related to interferon signaling. Moreover, after steroid tapering, in non-relapsed patients, genes with increased expression were enriched in inflammatory pathways, represented by interferon signaling, while genes with decreased expression were enriched in pathways related to IL-10 and glucocorticoid receptors. Weighted gene co-expression network analysis identified modules that correlated with steroid dosage, and the modules inversely correlated with steroid dosage were enriched in monocytes, with marked immune signature of interferon pathway. Conclusion This study identified peripheral blood transcriptome signatures that could lead to the identification of clinically relevant NMOSD disease activity biomarkers, and further highlights the pivotal role of interferon and IL-10 signaling in NMOSD.
Collapse
Affiliation(s)
- Ryohei Yamamura
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Makoto Kinoshita
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yoshiaki Yasumizu
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Tomohiro Yata
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Keigo Kihara
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Daisuke Motooka
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Naoyuki Shiraishi
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasuko Sugiyama
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shohei Beppu
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hisashi Murata
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naoshi Koizumi
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Itsuki Sano
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Toru Koda
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
3
|
Hathcock SF, Mamana J, Keyzer TE, Vollmuth N, Shokri MR, Mauser HD, Correll RN, Lam DW, Kim BJ, Sin J. Transcriptomic analysis of coxsackievirus B3 infection in induced pluripotent stem cell-derived brain-like endothelial cells. J Virol 2025; 99:e0182424. [PMID: 39670741 PMCID: PMC11784093 DOI: 10.1128/jvi.01824-24] [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: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 12/14/2024] Open
Abstract
Viral aseptic meningitis is a neuroinflammatory condition that occurs when viruses gain access to the central nervous system (CNS) and induce inflammation. The blood-brain barrier (BBB) is comprised of brain endothelial cells (BECs) that stringently regulate the passage of molecules, toxins, and pathogens from the circulation into the CNS. Through their unique properties, such as complex tight junctions, reduced rates of endocytosis, expression of efflux transporters, and restricted expression of leukocyte adhesion molecules, the BBB is often able to limit pathogen entry into the brain; however, certain neurotropic pathogens, such as coxsackievirus B3 (CVB3) are able to infect the CNS. We have previously demonstrated that CVB3 can infect and disrupt induced pluripotent stem cell-derived brain-like endothelial cells (iBECs), but the host response to this infection remains unknown. Here, we investigate global host transcriptional changes during CVB3 infection of iBECs using RNA sequencing. We validated our data set by exploring pathways altered by CVB3 using quantitative real-time PCR (qPCR) and enzyme-linked immunosorbent assay of upregulated cytokines and interferon signaling molecules. IMPORTANCE Coxsackievirus B3 (CVB3) is a leading cause of viral aseptic meningitis that can produce severe disease in susceptible individuals. To gain access to the central nervous system, CVB3 must cross central nervous system barriers, such as the blood-brain barrier. Previously, we have shown that CVB3 infects a human stem cell-derived brain-like endothelial cell model. Here, we report the global transcriptome of stem cell-derived brain-like endothelial cells to CVB3 infection and provide proof-of-concept validation of the dataset using molecular biology techniques. These data could inform novel mechanisms of CVB3-mediated blood-brain barrier dysfunction.
Collapse
Affiliation(s)
- Sarah F. Hathcock
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Julia Mamana
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Taryn E. Keyzer
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Nadine Vollmuth
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Mohammad-Reza Shokri
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Henry D. Mauser
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Robert N. Correll
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- Center for Convergent Biosciences and Medicine, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Daryl W. Lam
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Brandon J. Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- Center for Convergent Biosciences and Medicine, The University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Jon Sin
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
4
|
Speca DJ, He CW, Meyer CM, Scott EC, Díaz E. Functional characterization of endocytic signals in the SynDIG/PRRT family members SynDIG1 and SynDIG4 in heterologous cells and neurons. Front Cell Neurosci 2025; 18:1526034. [PMID: 39916936 PMCID: PMC11798926 DOI: 10.3389/fncel.2024.1526034] [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: 11/11/2024] [Accepted: 12/24/2024] [Indexed: 02/09/2025] Open
Abstract
The transmembrane protein Synapse Differentiation Induced Gene 4 (SynDIG4), also known as Proline-rich transmembrane protein 1 (PRRT1), is an AMPA-type glutamate receptor (AMPAR) auxiliary factor that is necessary for maintaining extra-synaptic pools of GluA1. Loss of SynDIG4, and the subsequent decrease in extra-synaptic GluA1, has been found to significantly impact synaptic plasticity in the hippocampus. However, how SynDIG4 establishes and maintains these pools is unclear. Previous studies suggested that endocytic machinery is important for maintaining a pool of mobile surface AMPARs, and that proteins associated with such cellular machinery are critical for proper protein trafficking and internalization. Given that SynDIG4 co-localizes with GluA1 in early and recycling endosomes in cultured hippocampal neurons, we sought to identify the sorting signals that target SynDIG4 to endosomes to further elucidate the role of SynDIG4 in GluA1 trafficking. In this study, we report that SynDIG4 possesses a YxxΦ sorting motif, 178-YVPV-181, responsible for binding to the AP-2 complex cargo-sorting subunit μ2. This motif appears critical for proper SynDIG4 internalization, as SynDIG4 mutant 178-AVPA-181, which disrupts binding to μ2, induces aberrant SynDIG4 accumulation at the plasma-membrane of heterologous cells and primary rat hippocampal neurons. We also show that SynDIG4 mutants lacking an endocytic signal co-localize with GluA1 but less so with GluA2 on the surface of heterologous cells. Furthermore, we show that another family member, SynDIG1, is enriched in the trans-Golgi network (TGN) and can traffic between the TGN and plasma membrane. We have identified a non-canonical μ2 binding sequence in SynDIG1 that induces aberrant accumulation at the plasma membrane of heterologous cells and primary rat hippocampal neurons, suggesting a conserved role for μ2-mediated endocytosis within the SynDIG family. These results provide important insight into the mechanisms by which SynDIG proteins are targeted to endosomal compartments as a step in understanding SynDIG-mediated regulation of AMPAR trafficking.
Collapse
Affiliation(s)
- David J. Speca
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Chun-Wei He
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Christina M. Meyer
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Erin C. Scott
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - Elva Díaz
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
5
|
Mak NSC, Liu J, Zhang D, Taylor J, Li X, Rahman K, Chen F, Datta SAK, Lai KK, Shi Z, Temperton N, Irving AT, Compton AA, Sloan RD. Alternative splicing expands the antiviral IFITM repertoire in Chinese rufous horseshoe bats. PLoS Pathog 2024; 20:e1012763. [PMID: 39724110 PMCID: PMC11801718 DOI: 10.1371/journal.ppat.1012763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/06/2025] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Species-specific interferon responses are shaped by the virus-host arms race. The human interferon-induced transmembrane protein (IFITM) family consists of three antiviral IFITM genes that arose by gene duplication. These genes restrict virus entry and are key players in antiviral interferon responses. The unique IFITM repertoires in different species influence their resistance to viral infections, but the role of IFITMs in shaping the enhanced antiviral immunity of reservoir bat species is unclear. Here, we identified an IFITM gene in Chinese rufous horseshoe bat, a natural host of severe acute respiratory syndrome (SARS)-related coronaviruses, that is alternatively spliced to produce two IFITM isoforms in native cells as shown by transcriptomics. These bat IFITMs have conserved structures in vitro as demonstrated by circular dichroism spectroscopy, yet they exhibit distinct antiviral specificities against influenza A virus, Nipah virus and coronaviruses including SARS-CoV, SARS-CoV-2 and MERS-CoV. In parallel with human IFITM1-3, bat IFITM isoforms localize to distinct sites of virus entry which influences their antiviral potency. Further bioinformatic analysis of IFITM repertoires in 206 mammals reveals that alternative splicing is a recurring strategy for IFITM diversification, albeit less widely adopted than gene duplication. These findings demonstrate that alternative splicing is a key strategy for evolutionary diversification in the IFITM family. Our study also highlights an example of convergent evolution where species-specific selection pressures led to expansion of the IFITM family through multiple means, underscoring the importance of IFITM diversity as a component of innate immunity.
Collapse
Affiliation(s)
- Nelly S. C. Mak
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Jingyan Liu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Dan Zhang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Jordan Taylor
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Xiaomeng Li
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Kazi Rahman
- HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Feiyu Chen
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Siddhartha A. K. Datta
- HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Kin Kui Lai
- HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Zhengli Shi
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich Chatham Maritime, Kent, United Kingdom
| | - Aaron T. Irving
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Infection, Immunity & Cancer, Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, China
- College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Biomedical and Health Translational Research Centre of Zhejiang Province, Haining, China
| | - Alex A. Compton
- HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Richard D. Sloan
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| |
Collapse
|
6
|
Xie Q, Wang L, Liao X, Huang B, Luo C, Liao G, Yuan L, Liu X, Luo H, Shu Y. Research Progress into the Biological Functions of IFITM3. Viruses 2024; 16:1543. [PMID: 39459876 PMCID: PMC11512382 DOI: 10.3390/v16101543] [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: 08/15/2024] [Revised: 09/22/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs) are upregulated by interferons. They are not only highly conserved in evolution but also structurally consistent and have almost identical structural domains and functional domains. They are all transmembrane proteins and have multiple heritable variations in genes. The IFITM protein family is closely related to a variety of biological functions, including antiviral immunity, tumor formation, bone metabolism, cell adhesion, differentiation, and intracellular signal transduction. The progress of the research on its structure and related functions, as represented by IFITM3, is reviewed.
Collapse
Affiliation(s)
- Qian Xie
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Liangliang Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China;
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | - Xinzhong Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Bi Huang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Chuming Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Guancheng Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Lifang Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Xuejie Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, China
| |
Collapse
|
7
|
Wu Y, Wang Z, Hu H, Wu T, Alabed AAA, Sun Z, Wang Y, Cui G, Cong W, Li C, Li P. Identification of Immune-Related Gene Signature in Schizophrenia. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:276-288. [PMID: 38863043 PMCID: PMC11190455 DOI: 10.62641/aep.v52i3.1648] [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: 06/13/2024]
Abstract
BACKGROUND Schizophrenia (SCZ) is a type of psychiatric disorder characterized by multiple symptoms. Our aim is to decipher the relevant mechanisms of immune-related gene signatures in SCZ. METHODS The SCZ dataset and its associated immunoregulatory genes were retrieved using Gene Expression Omnibus (GEO) and single-sample gene set enrichment analysis (ssGSEA). Co-expressed gene modules were determined through weighted gene correlation network analysis (WGCNA). To elucidate the functional characteristics of these clusters, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used. Additionally, gene set enrichment analysis (GSEA) and Gene Set Variation Analysis (GSVA) were conducted to identify enriched pathways for the immune subgroups. A protein-protein interaction (PPI) network analysis was performed to identify core genes relevant to SCZ. RESULTS A significantly higher immune score was observed in SCZ compared to control samples. Seven distinct gene modules were identified, with genes highlighted in green selected for further analysis. Using the Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) method, degrees of immune cell adhesion and accumulation related to 22 different immune cell types were calculated. Significantly enriched bioprocesses concerning the immunoregulatory genes with differential expressions included interferon-beta, IgG binding, and response to interferon-gamma, according to GO and KEGG analyses. Eleven hub genes related to immune infiltration emerged as key players among the three top-ranked GO terms. CONCLUSIONS This study underscores the involvement of immunoregulatory reactions in SCZ development. Eleven immune-related genes (IFITM1 (interferon induced transmembrane protein 1), GBP1 (guanylate binding protein 1), BST2 (bone marrow stromal cell antigen 2), IFITM3 (interferon induced transmembrane protein 3), GBP2 (guanylate binding protein 2), CD44 (CD44 molecule), FCER1G (Fc epsilon receptor Ig), HLA-DRA (major histocompatibility complex, class II, DR alpha), FCGR2A (Fc gamma receptor IIa), IFI16 (interferon gamma inducible protein 16), and FCGR3B (Fc gamma receptor IIIb)) were identified as hub genes, representing potential biomarkers and therapeutic targets associated with the immune response in SCZ patients.
Collapse
Affiliation(s)
- Yu Wu
- School of Nursing, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Zhichao Wang
- Department of Academic Research, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Houjia Hu
- School of Basic Medical Sciences, Nanchang University, 330006 Nanchang, Jiangxi, China
| | - Tong Wu
- Department of Psychology, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Alabed Ali A. Alabed
- Community Medicine Department, Faculty of Medicine, Lincoln University College, 47301 Petaling Jaya, Selangor, Malaysia
| | - Zhenghai Sun
- Department of Psychology, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Yuchen Wang
- Department of Psychology, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Guangcheng Cui
- Department of Psychology, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Weiliang Cong
- Department of Anaesthesiology, The Third Affiliated Hospital of Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Chengchong Li
- Department of Psychology, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| | - Ping Li
- Department of Psychology, Qiqihar Medical University, 161000 Qiqihar, Heilongjiang, China
| |
Collapse
|
8
|
Nekulová M, Wyszkowska M, Friedlová N, Uhrík L, Zavadil Kokáš F, Hrabal V, Hernychová L, Vojtěšek B, Hupp TR, Szymański MR. Biochemical evidence for conformational variants in the anti-viral and pro-metastatic protein IFITM1. Biol Chem 2024; 405:311-324. [PMID: 38379409 DOI: 10.1515/hsz-2023-0327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Interferon induced transmembrane proteins (IFITMs) play a dual role in the restriction of RNA viruses and in cancer progression, yet the mechanism of their action remains unknown. Currently, there is no data about the basic biochemical features or biophysical properties of the IFITM1 protein. In this work, we report on description and biochemical characterization of three conformational variants/oligomeric species of recombinant IFITM1 protein derived from an Escherichia coli expression system. The protein was extracted from the membrane fraction, affinity purified, and separated by size exclusion chromatography where two distinct oligomeric species were observed in addition to the expected monomer. These species remained stable upon re-chromatography and were designated as "dimer" and "oligomer" according to their estimated molecular weight. The dimer was found to be less stable compared to the oligomer using circular dichroism thermal denaturation and incubation with a reducing agent. A two-site ELISA and HDX mass spectrometry suggested the existence of structural motif within the N-terminal part of IFITM1 which might be significant in oligomer formation. Together, these data show the unusual propensity of recombinant IFITM1 to naturally assemble into very stable oligomeric species whose study might shed light on IFITM1 anti-viral and pro-oncogenic functions in cells.
Collapse
Affiliation(s)
- Marta Nekulová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Marta Wyszkowska
- Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, University of Gdansk, 80-307 Gdansk, Poland
| | - Nela Friedlová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Lukáš Uhrík
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Filip Zavadil Kokáš
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Václav Hrabal
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Lenka Hernychová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Bořivoj Vojtěšek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Ted R Hupp
- Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XR Edinburgh, UK
| | - Michał R Szymański
- Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, University of Gdansk, 80-307 Gdansk, Poland
| |
Collapse
|
9
|
Li H, Chen M, Zheng T, Lei X, Lin C, Li S, Mo J, Ning Z. IFITM1 and IFITM2 inhibit the replication of senecavirus A by positive feedback with RIG-I signaling pathway. Vet Microbiol 2024; 292:110050. [PMID: 38484578 DOI: 10.1016/j.vetmic.2024.110050] [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: 02/01/2024] [Revised: 03/02/2024] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
The role of host factors in the replication of emerging senecavirus A (SVA) which induced porcine idiopathic vesicular disease (PIVD) distributed worldwide remains obscure. Here, interferon-induced transmembrane (IFITM) protein 1 and 2 inhibit SVA replication by positive feedback with RIG-I signaling pathway was reported. The expression levels of IFITM1 and IFITM2 increased significantly in SVA infected 3D4/21 cells. Infection experiments of cells with over and interference expression of IFITM1 and IFITM2 showed that these two proteins inhibit SVA replication by regulating the expression of interferon beta (IFN-β), IFN-stimulated gene 15 (ISG-15), interleukin 6 (IL-6), IL-8, tumor necrosis factor alpha (TNF-α), IFN regulatory factor-3 (IRF3), and IRF7. Further results showed that antiviral responses of IFITM1 and IFITM2 were achieved by activating retinoic acid-inducible gene I (RIG-I) signaling pathway which in turn enhanced the expression of IFITM1 and IFITM2. It is noteworthy that conserved domains of these two proteins also paly the similar role. These findings provide new data on the role of host factors in infection and replication of SVA and help to develop new agents against the virus.
Collapse
Affiliation(s)
- Huizi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ming Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Tingting Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoling Lei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Cunhao Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Shuo Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jiacong Mo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhangyong Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China.
| |
Collapse
|
10
|
Yang Y, Ding T, Cong Y, Luo X, Liu C, Gong T, Zhao M, Zheng X, Li C, Zhang Y, Zhou J, Ni C, Zhang X, Ji Z, Wu T, Yang S, Zhou Q, Wu D, Gong X, Zheng Q, Li X. Interferon-induced transmembrane protein-1 competitively blocks Ephrin receptor A2-mediated Epstein-Barr virus entry into epithelial cells. Nat Microbiol 2024; 9:1256-1270. [PMID: 38649412 PMCID: PMC11087256 DOI: 10.1038/s41564-024-01659-0] [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: 03/06/2023] [Accepted: 03/04/2024] [Indexed: 04/25/2024]
Abstract
Epstein-Barr virus (EBV) can infect both B cells and epithelial cells (ECs), causing diseases such as mononucleosis and cancer. It enters ECs via Ephrin receptor A2 (EphA2). The function of interferon-induced transmembrane protein-1 (IFITM1) in EBV infection of ECs remains elusive. Here we report that IFITM1 inhibits EphA2-mediated EBV entry into ECs. RNA-sequencing and clinical sample analysis show reduced IFITM1 in EBV-positive ECs and a negative correlation between IFITM1 level and EBV copy number. IFITM1 depletion increases EBV infection and vice versa. Exogenous soluble IFITM1 effectively prevents EBV infection in vitro and in vivo. Furthermore, three-dimensional structure prediction and site-directed mutagenesis demonstrate that IFITM1 interacts with EphA2 via its two specific residues, competitively blocking EphA2 binding to EBV glycoproteins. Finally, YTHDF3, an m6A reader, suppresses IFITM1 via degradation-related DEAD-box protein 5 (DDX5). Thus, this study underscores IFITM1's crucial role in blocking EphA2-mediated EBV entry into ECs, indicating its potential in preventing EBV infection.
Collapse
Affiliation(s)
- Yinggui Yang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Tengteng Ding
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Cong
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaomin Luo
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Changlin Liu
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Gong
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Min Zhao
- PANACRO(Hefei) Pharmaceutical Technology Co. Ltd., Hefei, China
| | - Xichun Zheng
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Chenglin Li
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Yuanbin Zhang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiayi Zhou
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuping Ni
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xueyu Zhang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziliang Ji
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Tao Wu
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Shaodong Yang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Qingchun Zhou
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Dinglan Wu
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China.
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Xinqi Gong
- Mathematical Intelligence Application LAB, Institute for Mathematical Sciences, Renmin University of China, Beijing, China.
| | - Qingyou Zheng
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China.
| | - Xin Li
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China.
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Elessawy SM, Shehab A, Soliman DA, Eldeeb MA, Taha SI. Interferon-Induced Transmembrane Protein-3 Rs12252-G Variant Increases COVID-19 Mortality Potential in Egyptian Population. Viral Immunol 2024; 37:186-193. [PMID: 38717821 DOI: 10.1089/vim.2024.0015] [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] [Indexed: 05/14/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) represented an international health risk. Variants of the interferon-induced transmembrane protein-3 (IFITM3) gene can increase the risk of developing severe viral infections. This cross-sectional study investigated the association between IFITM3 rs12252A>G single nucleotide polymorphism (SNP) and COVID-19 severity and mortality in 100 Egyptian patients. All participants were subjected to serum interleukin-6 (IL-6) determination by ELISA and IFITM3 rs12252 genotyping by real-time polymerase chain reaction. Of all participants, 85.0% had the IFITM3 rs12252 homozygous AA genotype, whereas 15.0% had the heterozygous AG genotype. None of our participants had the homozygous GG genotype. The IFITM3 rs12252A allele was found in 92.5% and the G allele in only 7.5%. There was no significant association (p > 0.05) between the IFITM3 rs12252 SNP and COVID-19 severity, intensive care unit (ICU) admission, or IL-6 serum levels. The heterozygous AG genotype frequency showed a significant increase among participants who died (32.0%) compared with those who had been cured (9.3%). The mutant G allele was associated with patients' death. Its frequency among cured participants was 8.5%, whereas in those who died was 24.2% (p = 0.024) with 3.429 odds ratio [95% confidence interval: 1.1-10.4]. In conclusion, this study revealed a significant association between the G allele variant of IFITM3 rs12252 and COVID-19 mortality. However, results were unable to establish a significant link between rs12252 polymorphism, disease severity, ICU admission, or serum IL-6 levels.
Collapse
Affiliation(s)
- Sara M Elessawy
- Department of Clinical Pathology, Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Abeer Shehab
- Department of Clinical Pathology, Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Dina A Soliman
- Department of Clinical Pathology, Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mai A Eldeeb
- Department of Internal Medicine, Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sara I Taha
- Department of Clinical Pathology, Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
Du S, Wang Y, Wang J, Ma Y, Xu W, Shi X, Li L, Hao P, Liu Q, Liao M, Zhou B, Jin N, Wong YK, Hu L, Wang J, Liu W, Li C. IFITM3 inhibits severe fever with thrombocytopenia syndrome virus entry and interacts with viral Gc protein. J Med Virol 2024; 96:e29491. [PMID: 38402626 DOI: 10.1002/jmv.29491] [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: 10/20/2023] [Revised: 01/17/2024] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne hemorrhagic fever disease with high fatality rate of 10%-20%. Vaccines or specific therapeutic measures remain lacking. Human interferon inducible transmembrane protein 3 (hIFITM3) is a broad-spectrum antiviral factor targeting viral entry. However, the antiviral activity of hIFITM3 against SFTS virus (SFTSV) and the functional mechanism of IFITM3 remains unclear. Here we demonstrate that endogenous IFITM3 provides protection against SFTSV infection and participates in the anti-SFTSV effect of type Ⅰ and Ⅲ interferons (IFNs). IFITM3 overexpression exhibits anti-SFTSV function by blocking Gn/Gc-mediated viral entry and fusion. Further studies showed that IFITM3 binds SFTSV Gc directly and its intramembrane domain (IMD) is responsible for this interaction and restriction of SFTSV entry. Mutation of two neighboring cysteines on IMD weakens IFITM3-Gc interaction and attenuates the antiviral activity of IFITM3, suggesting that IFITM3-Gc interaction may partly mediate the inhibition of SFTSV entry. Overall, our data demonstrate for the first time that hIFITM3 plays a critical role in the IFNs-mediated anti-SFTSV response, and uncover a novel mechanism of IFITM3 restriction of SFTSV infection, highlighting the potential of clinical intervention on SFTS disease.
Collapse
Affiliation(s)
- Shouwen Du
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangdong Academy of Agricultural Sciences, Guangzhou, P.R. China
- Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, P.R. China
| | - Yuhang Wang
- Shenzhen Bay Laboratory, Pingshan Translational Medicine Center, Shenzhen, P.R. China
| | - Jiamin Wang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Yidan Ma
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Wang Xu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Xiaoshuang Shi
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Pengfei Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Quan Liu
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangdong Academy of Agricultural Sciences, Guangzhou, P.R. China
| | - Ming Liao
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Guangdong Academy of Agricultural Sciences, Guangzhou, P.R. China
| | - Boping Zhou
- Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, P.R. China
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| | - Yin K Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lifen Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
| | - Jigang Wang
- Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, P.R. China
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, P.R. China
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, P.R. China
| |
Collapse
|
13
|
Zhang Y, Li X, Zhang S, Li J, Liu M, Lu Y, Han J. Role of IFITM2 in osteogenic differentiation of C3H10T1/2 mesenchymal stem cells. Intractable Rare Dis Res 2024; 13:42-50. [PMID: 38404731 PMCID: PMC10883848 DOI: 10.5582/irdr.2023.01108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/27/2024] Open
Abstract
Interferon-inducible transmembrane (IFITM) are a family of small proteins localized to plasma and endolysosomal membranes. Their functions beyond restricting viral entry and replication have been revealed in recent years. IFITM5 is involved in bone mineralization and is an osteogenic cell surface marker. IFITM1 and 3 interact with desmin and myosin, and are involved in myogenic differentiation. This study found upregulation of Ifitm2 during osteogenic differentiation of C3H10T1/2 cells. This positively correlated to the expression of osteogenic differentiation markers Col1a1, Alp, Runx2, and Ocn. Knockdown of Ifitm2 by siRNAs inhibited osteogenic differentiation, calcium deposition, and osteogenic marker expression of C3H10T1/2 cells. The osteoblast transcriptome revealed that knocking down Ifitm2 affected the expression Wnt signaling pathway-related genes, including Wnt family members, their receptors Lrp, Frizzled, and Lgr, and transmembrane molecule Rnf43 that suppresses the Wnt signaling pathway. Luciferase assays indicated enhancement of canonical Wnt signaling pathways by Ifitm2 overexpression. Furthermore, IFITM2 was colocalized in the metaphyseal bone and growth plate of the mouse tibial bone with SP7, a transcription factor essential for osteoblast differentiation and bone formation. These findings reveal a possible novel function and potential mechanisms of Ifitm2 in osteogenic differentiation.
Collapse
Affiliation(s)
- Yongtao Zhang
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Xiangdong Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Shanshan Zhang
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Junfeng Li
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Meilin Liu
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Yanqin Lu
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Jinxiang Han
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| |
Collapse
|
14
|
Sievers BL, Cheng MTK, Csiba K, Meng B, Gupta RK. SARS-CoV-2 and innate immunity: the good, the bad, and the "goldilocks". Cell Mol Immunol 2024; 21:171-183. [PMID: 37985854 PMCID: PMC10805730 DOI: 10.1038/s41423-023-01104-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023] Open
Abstract
An ancient conflict between hosts and pathogens has driven the innate and adaptive arms of immunity. Knowledge about this interplay can not only help us identify biological mechanisms but also reveal pathogen vulnerabilities that can be leveraged therapeutically. The humoral response to SARS-CoV-2 infection has been the focus of intense research, and the role of the innate immune system has received significantly less attention. Here, we review current knowledge of the innate immune response to SARS-CoV-2 infection and the various means SARS-CoV-2 employs to evade innate defense systems. We also consider the role of innate immunity in SARS-CoV-2 vaccines and in the phenomenon of long COVID.
Collapse
Affiliation(s)
| | - Mark T K Cheng
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kata Csiba
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Bo Meng
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Ravindra K Gupta
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
15
|
López-Domínguez R, Villatoro-García JA, Marañón C, Goldman D, Petri M, Carmona-Sáez P, Alarcón-Riquelme M, Toro-Dominguez D. Immune and molecular landscape behind non-response to Mycophenolate Mofetil and Azathioprine in lupus nephritis therapy. RESEARCH SQUARE 2024:rs.3.rs-3783877. [PMID: 38260685 PMCID: PMC10802741 DOI: 10.21203/rs.3.rs-3783877/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Lupus nephritis (LN) represents one of the most severe complications of systemic lupus erythematosus, leading to end-stage kidney disease in worst cases. Current first-line therapies for LN, including mycophenolate mofetil (MMF) and azathioprine (AZA), fail to induce long-term remission in 60-70% of the patients, evidencing the urgent need to delve into the molecular knowledge-gap behind the non-response to these therapies. A longitudinal cohort of treated LN patients including clinical, cellular and transcriptomic data, was analyzed. Gene-expression signatures behind non-response to different drugs were revealed by differential expression analysis. Drug-specific non-response mechanisms and cell proportion differences were identified. Blood cell subsets mediating non-response were described using single-cell RNASeq data. We show that AZA and MMF non-response implicates different cells and regulatory functions. Mechanistic models were used to suggest add-on therapies to improve their current performance. Our results provide new insights into the molecular mechanisms associated with treatment failures in LN.
Collapse
Affiliation(s)
- Raúl López-Domínguez
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, PTS Granada
| | - Juan Antonio Villatoro-García
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, PTS Granada
| | - Concepción Marañón
- Department of Medical Genomics, Center for Genomics and Oncological Research (GENYO)
| | | | | | - Pedro Carmona-Sáez
- Department of Bioinformatics, Center for Genomics and Oncological Research (GENYO)
| | | | - Daniel Toro-Dominguez
- GENYO. Centre for Genomics and Oncological Research: Pfizer, University of Granada, Andalusian Regional Government, PTS Granada
| |
Collapse
|
16
|
Eshchenko N, Sergeeva M, Zhuravlev E, Kudria K, Goncharova E, Komissarov A, Stepanov G. A Knockout of the IFITM3 Gene Increases the Sensitivity of WI-38 VA13 Cells to the Influenza A Virus. Int J Mol Sci 2024; 25:625. [PMID: 38203797 PMCID: PMC10778886 DOI: 10.3390/ijms25010625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
One of the ways to regulate the sensitivity of human cells to the influenza virus is to knock out genes of the innate immune response. Promising targets for the knockout are genes of the interferon-inducible transmembrane protein (IFITM) family, in particular the IFITM3 gene, whose product limits the entry of a virus into the cell by blocking the fusion of the viral and endosomal membranes. In this study, by means of genome-editing system CRISPR/Cas9, monoclonal cell lines with an IFITM3 knockout were obtained based on WI-38 VA13 cells (human origin). It was found that such cell lines are more sensitive to infection by influenza A viruses of various subtypes. Nevertheless, this feature is not accompanied by an increased titer of newly formed viral particles in a culture medium.
Collapse
Affiliation(s)
- Natalya Eshchenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| | - Mariia Sergeeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg 197376, Russia;
| | - Evgenii Zhuravlev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| | - Kira Kudria
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg 197376, Russia;
| | - Elena Goncharova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| | - Andrey Komissarov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg 197376, Russia;
| | - Grigory Stepanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| |
Collapse
|
17
|
Vetters J, van Helden M, De Nolf C, Rennen S, Cloots E, Van De Velde E, Fayazpour F, Van Moorleghem J, Vanheerswynghels M, Vergote K, Boon L, Vivier E, Lambrecht BN, Janssens S. Canonical IRE1 function needed to sustain vigorous natural killer cell proliferation during viral infection. iScience 2023; 26:108570. [PMID: 38162021 PMCID: PMC10755724 DOI: 10.1016/j.isci.2023.108570] [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: 08/25/2023] [Revised: 10/16/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
The unfolded protein response (UPR) aims to restore ER homeostasis under conditions of high protein folding load, a function primarily serving secretory cells. Additional, non-canonical UPR functions have recently been unraveled in immune cells. We addressed the function of the inositol-requiring enzyme 1 (IRE1) signaling branch of the UPR in NK cells in homeostasis and microbial challenge. Cell-intrinsic compound deficiency of IRE1 and its downstream transcription factor XBP1 in NKp46+ NK cells, did not affect basal NK cell homeostasis, or overall outcome of viral MCMV infection. However, mixed bone marrow chimeras revealed a competitive advantage in the proliferation of IRE1-sufficient Ly49H+ NK cells after viral infection. CITE-Seq analysis confirmed strong induction of IRE1 early upon infection, concomitant with the activation of a canonical UPR signature. Therefore, we conclude that IRE1/XBP1 activation is required during vigorous NK cell proliferation early upon viral infection, as part of a canonical UPR response.
Collapse
Affiliation(s)
- Jessica Vetters
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Mary van Helden
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Byondis B.V., Nijmegen, the Netherlands
| | - Clint De Nolf
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Barriers in Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sofie Rennen
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Eva Cloots
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Evelien Van De Velde
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Farzaneh Fayazpour
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Justine Van Moorleghem
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Manon Vanheerswynghels
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Karl Vergote
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | | | - Eric Vivier
- Aix Marseille University, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- AP-HM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Bart N. Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Sophie Janssens
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
18
|
Yu C, Wang G, Liu Q, Zhai J, Xue M, Li Q, Xian Y, Zheng C. Host antiviral factors hijack furin to block SARS-CoV-2, ebola virus, and HIV-1 glycoproteins cleavage. Emerg Microbes Infect 2023; 12:2164742. [PMID: 36591809 PMCID: PMC9897805 DOI: 10.1080/22221751.2022.2164742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Viral envelope glycoproteins are crucial for viral infections. In the process of enveloped viruses budding and release from the producer cells, viral envelope glycoproteins are presented on the viral membrane surface as spikes, promoting the virus's next-round infection of target cells. However, the host cells evolve counteracting mechanisms in the long-term virus-host co-evolutionary processes. For instance, the host cell antiviral factors could potently suppress viral replication by targeting their envelope glycoproteins through multiple channels, including their intracellular synthesis, glycosylation modification, assembly into virions, and binding to target cell receptors. Recently, a group of studies discovered that some host antiviral proteins specifically recognized host proprotein convertase (PC) furin and blocked its cleavage of viral envelope glycoproteins, thus impairing viral infectivity. Here, in this review, we briefly summarize several such host antiviral factors and analyze their roles in reducing furin cleavage of viral envelope glycoproteins, aiming at providing insights for future antiviral studies.
Collapse
Affiliation(s)
- Changqing Yu
- School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People’s Republic of China
| | - Guosheng Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Qiang Liu
- Nanchong Key Laboratory of Disease Prevention, Control and Detection in Livestock and Poultry, Nanchong Vocational and Technical College, Nanchong, People’s Republic of China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, People’s Republic of China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China,Mengzhou Xue
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China,Qiang Li
| | - Yuanhua Xian
- School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People’s Republic of China,Yuanhua Xian
| | - Chunfu Zheng
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China,Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Canada, Chunfu Zheng
| |
Collapse
|
19
|
Pecoraro V, Cuccorese M, Trenti T. Genetic polymorphisms of ACE1, ACE2, IFTM3, TMPRSS2 and TNFα genes associated with susceptibility and severity of SARS-CoV-2 infection: a systematic review and meta-analysis. Clin Exp Med 2023; 23:3251-3264. [PMID: 37055652 PMCID: PMC10101542 DOI: 10.1007/s10238-023-01038-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/01/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Some human polymorphisms of ACE1, ACE2, IFITM3, TMPRSS2 and TNFα genes may have an effect on the susceptibility to SARS-CoV-2 infection and increase the risk to develop severe COVID-19. We conducted a systematic review of current evidence to investigate the association of genetic variants of these genes with the susceptibility to virus infection and patient prognosis. METHODS We systematically searched Medline, Embase and The Cochrane Library for articles published until May 2022, and included observational studies covering genetic association of ACE1, ACE2, IFITM3, TMPRSS2 and TNFα genes with COVID-19 susceptibility or prognosis. We evaluated the methodological quality of included studies, and pooled data as convenient in meta-analysis (MA). Odds ratio (OR) values and 95% confidence intervals were calculated. RESULTS We included 35 studies (20 on ACE, 5 each on IFITM3, TMPRSS2, TNFα), enrolling 21,452 participants, of them 9401 were COVID-19 confirmed cases. ACE1 rs4646994 and rs1799752, ACE2 rs2285666, TMPRSS2 rs12329760, IFITM3 rs12252 and TNFα rs1800629 were identifies as common polymorphisms. Our MA showed an association between genetic polymorphisms and susceptibility to SARS-CoV-2 infection for IFITM3 rs12252 CC (OR 5.67) and CT (OR 1.64) genotypes. Furthermore, MA uncovered that both ACE DD (OR 1.27) and IFITM3 CC (OR 2.26) genotypes carriers had a significantly increased risk of developing severe COVID-19. DISCUSSION These results provide a critical evaluation of genetic polymorphisms as predictors in SARS-CoV-2 infection. ACE1 DD and IFITM3 CC polymorphisms would lead to a genetic predisposition for severe lung injury in patients with COVID-19.
Collapse
Affiliation(s)
- Valentina Pecoraro
- Department of Laboratory Medicine and Pathology, Azienda USL of Modena, Modena, Italy
| | - Michela Cuccorese
- Department of Laboratory Medicine and Pathology, Azienda USL of Modena, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Azienda USL of Modena, Modena, Italy
| |
Collapse
|
20
|
Zhao Q, Zhang R, Qiao C, Miao Y, Yuan Y, Zheng H. Ubiquitination network in the type I IFN-induced antiviral signaling pathway. Eur J Immunol 2023; 53:e2350384. [PMID: 37194705 DOI: 10.1002/eji.202350384] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/14/2023] [Accepted: 05/16/2023] [Indexed: 05/18/2023]
Abstract
Type I IFN (IFN-I) is the body's first line of defense against pathogen infection. IFN-I can induce cellular antiviral responses and therefore plays a key role in driving antiviral innate and adaptive immunity. Canonical IFN-I signaling activates the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, which induces the expression of IFN-stimulated genes and eventually establishes a complex antiviral state in the cells. Ubiquitin is a ubiquitous cellular molecule for protein modifications, and the ubiquitination modifications of protein have been recognized as one of the key modifications that regulate protein levels and/or signaling activation. Despite great advances in understanding the ubiquitination regulation of many signaling pathways, the mechanisms by which protein ubiquitination regulates IFN-I-induced antiviral signaling have not been explored until very recently. This review details the current understanding of the regulatory network of ubiquitination that critically controls the IFN-I-induced antiviral signaling pathway from three main levels, including IFN-I receptors, IFN-I-induced cascade signals, and effector IFN-stimulated genes.
Collapse
Affiliation(s)
- Qian Zhao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Renxia Zhang
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Caixia Qiao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Ying Miao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Yukang Yuan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Hui Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| |
Collapse
|
21
|
Kaur R, Tada T, Landau NR. Restriction of SARS-CoV-2 replication by receptor transporter protein 4 (RTP4). mBio 2023; 14:e0109023. [PMID: 37382452 PMCID: PMC10470548 DOI: 10.1128/mbio.01090-23] [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: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 06/30/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is subject to restriction by several interferon-inducible host proteins. To identify novel factors that limit replication of the virus, we tested a panel of genes that we found were induced by interferon treatment of primary human monocytes by RNA sequencing. Further analysis showed that one of the several candidates genes tested, receptor transporter protein 4 (RTP4), that had previously been shown to restrict flavivirus replication, prevented the replication of the human coronavirus HCoV-OC43. Human RTP4 blocked the replication of SARS-CoV-2 in susceptible ACE2.CHME3 cells and was active against SARS-CoV-2 Omicron variants. The protein prevented the synthesis of viral RNA, resulting in the absence of detectable viral protein synthesis. RTP4 bound the viral genomic RNA and the binding was dependent on the conserved zinc fingers in the amino-terminal domain. Expression of the protein was strongly induced in SARS-CoV-2-infected mice although the mouse homolog was inactive against the virus, suggesting that the protein is active against another virus that remains to be identified. IMPORTANCE The rapid spread of a pathogen of human coronavirus (HCoV) family member, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), around the world has led to a coronavirus disease 2019 (COVID-19) pandemic. The COVID-19 pandemic spread highlights the need for rapid identification of new broad-spectrum anti-coronavirus drugs and screening of antiviral host factors capable of inhibiting coronavirus infection. In the present work, we identify and characterize receptor transporter protein 4 (RTP4) as a host restriction factor that restricts coronavirus infection. We examined the antiviral role of hRTP4 toward the coronavirus family members including HCoV-OC43, SARS-CoV-2, Omicron BA.1, and BA.2. Molecular and biochemical analysis showed that hRTP4 binds to the viral RNA and targets the replication phase of viral infection and is associated with reduction of nucleocapsid protein. Significant higher levels of ISGs were observed in SARS-CoV-2 mouse model, suggesting the role of RTP4 in innate immune regulation in coronavirus infection. The identification of RTP4 reveals a potential target for therapy against coronavirus infection.
Collapse
Affiliation(s)
- Ramanjit Kaur
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Takuya Tada
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Nathaniel Roy Landau
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
22
|
Zhang Y, Lu Y, Li X, Zhang S, Liu P, Hao X, Han J. The novel role of IFITM1-3 in myogenic differentiation of C2C12 cells. Intractable Rare Dis Res 2023; 12:180-190. [PMID: 37662621 PMCID: PMC10468414 DOI: 10.5582/irdr.2023.01050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/15/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs 1, 2, and 3) play a critical role in preventing pathogen infection in vertebrates. They are also involved in the occurrence and prognosis of cancer. Myogenesis is a complex process regulated by several factors. This study disclosed that Ifitm1-3 were upregulated in the process of myogenic differentiation of C2C12 myoblasts on days 3, 5, and 7. This positively correlated with the expression of differentiation factors MyoD, myogenin, Mrf5, and desmin. Furthermore, knockdown of Ifitm1-3 by their individual siRNAs inhibited myogenesis of C2C12 myoblasts, with relative downregulation of MyoD, myogenin, Mrf5, and desmin. Subsequently, myotube formation and fusion percentage decreased. Co-immunoprecipitation combined with LC-MS/MS analysis uncovered the interaction proteins of IFITM1 and IFITM3 in C2C12 myoblasts. A total of 84 overlapped interaction proteins of IFITM1 and IFITM3 were identified, and one of the clusters was engaged in cytoskeletal and sarcomere proteins, including desmin, myosin, actin, vimentin, nestin, ankycorbin, and nucleolin. Hence, we hypothesize that these interacting proteins may function as scaffolds for IFITM1-3, possibly through the interaction protein desmin to initiate further interaction with other proteins to participate in myogenesis; however, the molecular mechanisms remain unclear. Our study may contribute to the development of novel therapeutics for myopathic diseases.
Collapse
Affiliation(s)
- Yongtao Zhang
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Science College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Yanqin Lu
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Science College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Xianxian Li
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Science College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Shanshan Zhang
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Science College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Pengchao Liu
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Science College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Xiaoyang Hao
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Science College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Jinxiang Han
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Science College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| |
Collapse
|
23
|
Schelle L, Abrantes J, Baldauf HM, Esteves PJ. Evolution of primate interferon-induced transmembrane proteins (IFITMs): a story of gain and loss with a differentiation into a canonical cluster and IFITM retrogenes. Front Microbiol 2023; 14:1213685. [PMID: 37577422 PMCID: PMC10415907 DOI: 10.3389/fmicb.2023.1213685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/06/2023] [Indexed: 08/15/2023] Open
Abstract
Interferon-inducible transmembrane proteins (IFITMs) are a family of transmembrane proteins. The subgroup of immunity-related (IR-)IFITMs is involved in adaptive and innate immune responses, being especially active against viruses. Here, we suggest that IFITMs should be classified as (1) a canonical IFITM gene cluster, which is located on the same chromosome, and (2) IFITM retrogenes, with a random and unique location at different positions within the genome. Phylogenetic analyses of the canonical cluster revealed the existence of three novel groups of primate IFITMs (pIFITM) in the IR-IFITM clade: the prosimian pIFITMs(pro), the new world monkey pIFITMs(nwm) and the old world monkey pIFITMs(owm). Therefore, we propose a new nomenclature: IR-pIFITM1, IR-pIFITM2, IR-pIFITM3, IR-pIFITMnwm, IR-pIFITMowm, and IR-pIFITMpro. We observed divergent evolution for pIFITM5 and pIFITM10, and evidence for concerted evolution and a mechanism of birth-and-death evolution model for the IR-pIFITMs. In contrast, the IFITMs scattered throughout the genomes possessed features of retrogenes retrotransposed by class 1 transposable elements. The origin of the IFITM retrogenes correspond to more recent events. We hypothesize that the transcript of a canonical IFITM3 has been constantly retrotransposed using class 1 transposable elements resulting in the IFITM retro(pseudo)genes. The unique pattern of each species has most likely been caused by constant pseudogenization and loss of the retro(pseudo)genes. This suggests a third mechanism of evolution for the IR-IFITMs in primates, similar to the birth-and-death model of evolution, but via a transposable element mechanism, which resulted in retro(pseudo)genes.
Collapse
Affiliation(s)
- Luca Schelle
- Faculty of Medicine, Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU München, Munich, Germany
| | - Joana Abrantes
- CIBIO-InBIO, Research Center in Biodiversity and Genetic Resources, University of Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Hanna-Mari Baldauf
- Faculty of Medicine, Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU München, Munich, Germany
| | - Pedro José Esteves
- CIBIO-InBIO, Research Center in Biodiversity and Genetic Resources, University of Porto, Vairão, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- CITS - Center of Investigation in Health Technologies, CESPU, Gandra, Portugal
| |
Collapse
|
24
|
Lu Y, Saibro-Girardi C, Fitz NF, McGuire MR, Ostach MA, Mamun-Or-Rashid ANM, Lefterov I, Koldamova R. Multi-transcriptomics reveals brain cellular responses to peripheral infection in Alzheimer's disease model mice. Cell Rep 2023; 42:112785. [PMID: 37436901 PMCID: PMC10530196 DOI: 10.1016/j.celrep.2023.112785] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/03/2023] [Accepted: 06/24/2023] [Indexed: 07/14/2023] Open
Abstract
Peripheral inflammation has been linked to various neurodegenerative disorders, including Alzheimer's disease (AD). Here we perform bulk, single-cell, and spatial transcriptomics in APP/PS1 mice intranasally exposed to Staphylococcus aureus to determine how low-grade peripheral infection affects brain transcriptomics and AD-like pathology. Chronic exposure led to increased amyloid plaque burden and plaque-associated microglia, significantly affecting the transcription of brain barrier-associated cells, which resulted in barrier leakage. We reveal cell-type- and spatial-specific transcriptional changes related to brain barrier function and neuroinflammation during the acute infection. Both acute and chronic exposure led to brain macrophage-associated responses and detrimental effects in neuronal transcriptomics. Finally, we identify unique transcriptional responses at the amyloid plaque niches following acute infection characterized by higher disease-associated microglia gene expression and a larger effect on astrocytic or macrophage-associated genes, which could facilitate amyloid and related pathologies. Our findings provide important insights into the mechanisms linking peripheral inflammation to AD pathology.
Collapse
Affiliation(s)
- Yi Lu
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Carolina Saibro-Girardi
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nicholas Francis Fitz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mikayla Ranae McGuire
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mary Ann Ostach
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - A N M Mamun-Or-Rashid
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Iliya Lefterov
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Radosveta Koldamova
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
25
|
Malarmathi M, Murali N, Selvaraju M, Sivakumar K, Gowthaman V, Raghavendran VB, Raja A, Peters SO, Thiruvenkadan AK. In Vitro Characterization of chIFITMs of Aseel and Kadaknath Chicken Breeds against Newcastle Disease Virus Infection. BIOLOGY 2023; 12:919. [PMID: 37508350 PMCID: PMC10376314 DOI: 10.3390/biology12070919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/14/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023]
Abstract
Newcastle disease (ND) is highly contagious and usually causes severe illness that affects Aves all over the world, including domestic poultry. Depending on the virus's virulence, it can impact the nervous, respiratory, and digestive systems and cause up to 100% mortality. The chIFITM genes are activated in response to viral infection. The current study was conducted to quantify the mRNA of chIFITM genes in vitro in response to ND viral infection. It also examined its ability to inhibit ND virus replication in chicken embryo fibroblast (CEF) cells of the Aseel and Kadaknath breeds. Results from the study showed that the expression of all chIFITM genes was significantly upregulated throughout the period in the infected CEF cells of both breeds compared to uninfected CEF cells. In CEF cells of the Kadaknath breed, elevated levels of expression of the chIFITM3 gene dramatically reduced ND viral growth, and the viral load was 60% lower than in CEF cells of the Aseel breed. The expression level of the chIFITMs in Kadaknath ranged from 2.39 to 11.68 log2 folds higher than that of control CEFs and was consistently (p < 0.01) higher than Aseel CEFs. Similar to this, theIFN-γ gene expresses strongly quickly and peaks at 13.9 log2 fold at 48 hpi. Based on these cellular experiments, the Kadaknath breed exhibits the potential for greater disease tolerance than Aseel. However, to gain a comprehensive understanding of disease resistance mechanisms in chickens, further research involving in vivo investigations is crucial.
Collapse
Affiliation(s)
- Muthusamy Malarmathi
- Veterinary College and Research Institute, Tamil Nadu Veterinary and Animal Sciences University, Namakkal 637 002, India
| | - Nagarajan Murali
- Veterinary College and Research Institute, Tamil Nadu Veterinary and Animal Sciences University, Namakkal 637 002, India
| | - Mani Selvaraju
- Veterinary College and Research Institute, Tamil Nadu Veterinary and Animal Sciences University, Namakkal 637 002, India
| | - Karuppusamy Sivakumar
- Faculty of Food and Agriculture, The University of the West Indies, St Augustine 999183, Trinidad and Tobago
| | - Vasudevan Gowthaman
- Poultry Disease Diagnosis and Surveillance Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Namakkal 637 002, India
| | | | - Angamuthu Raja
- Veterinary College and Research Institute, Tamil Nadu Veterinary and Animal Sciences University, Namakkal 637 002, India
| | - Sunday O Peters
- Department of Animal Science, Berry College, Mount Berry, GA 30149, USA
| | | |
Collapse
|
26
|
Gholami M, Sakhaee F, Mirzaei Gheinari F, Sotoodehnejadnematalahi F, Ghazanfari Jajin M, Zamani MS, Ahmadi I, Anvari E, Fateh A. Interferon-Induced Transmembrane Protein 3 rs34481144 C/T Genotype and Clinical Parameters Related to Progression of COVID-19. J Immunol Res 2023; 2023:2345062. [PMID: 37323564 PMCID: PMC10266908 DOI: 10.1155/2023/2345062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/04/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Recent research has associated the interferon-induced transmembrane protein 3 gene (IFITM3) with the outcomes of coronavirus disease 2019 (COVID-19), although the findings are contradictory. This study aimed to determine the relationship between IFITM3 gene rs34481144 polymorphism and clinical parameters with COVID-19 mortality. The tetra-primer amplification refractory mutation system-polymerase chain reaction assay was used to analyze IFITM3 rs34481144 polymorphism in 1,149 deceased and 1,342 recovered patients. The clinical parameters were extracted from the patients' medical records. In this study, the frequency of IFITM3 rs34481144 CT genotypes (OR 1.47, 95% CI 1.23-1.76, P < 0.0001) in both sexes was significantly higher in deceased patients than in recovered patients. Moreover, IFITM3 rs34481144 TT genotypes (OR 3.38, 95% CI 1.05-10.87, P < 0.0001) in women were significantly associated with COVID-19 mortality. The multivariable logistic regression model results indicated that mean age (P < 0.001), alkaline phosphatase (P = 0.005), alanine aminotransferase (P < 0.001), low-density lipoprotein (P < 0.001), high-density lipoprotein (P < 0.001), fasting blood glucose (P = 0.010), creatinine (P < 0.001), uric acid (P < 0.001), C-reactive protein (P = 0.004), 25-hydroxyvitamin D (P < 0.001), erythrocyte sedimentation rate (P < 0.001), and real-time PCR Ct values (P < 0.001) were linked with increased COVID-19 death rates. In conclusion, IFITM3 rs34481144 gene polymorphism was linked to the mortality of COVID-19, with the rs34481144-T allele being especially important for mortality. Further studies are needed to confirm the results of this study.
Collapse
Affiliation(s)
- Melika Gholami
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Sakhaee
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | | | | | | | | | - Iraj Ahmadi
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Enayat Anvari
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
27
|
Prikryl D, Marin M, Desai TM, Du Y, Fu H, Melikyan GB. Cyclosporines Antagonize the Antiviral Activity of IFITMProteins by Redistributing Them toward the Golgi Apparatus. Biomolecules 2023; 13:937. [PMID: 37371517 PMCID: PMC10296495 DOI: 10.3390/biom13060937] [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: 05/17/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs) block the fusion of diverse enveloped viruses, likely through increasing the cell membrane's rigidity. Previous studies have reported that the antiviral activity of the IFITM family member, IFITM3, is antagonized by cell pretreatment with rapamycin derivatives and cyclosporines A and H (CsA and CsH) that promote the degradation of IFITM3. Here, we show that CsA and CsH potently enhance virus fusion with IFITM1- and IFITM3-expressing cells by inducing their rapid relocalization from the plasma membrane and endosomes, respectively, towards the Golgi. This relocalization is not associated with a significant degradation of IFITMs. Although prolonged exposure to CsA induces IFITM3 degradation in cells expressing low endogenous levels of this protein, its levels remain largely unchanged in interferon-treated cells or cells ectopically expressing IFITM3. Importantly, the CsA-mediated redistribution of IFITMs to the Golgi occurs on a much shorter time scale than degradation and thus likely represents the primary mechanism of enhancement of virus entry. We further show that rapamycin also induces IFITM relocalization toward the Golgi, albeit less efficiently than cyclosporines. Our findings highlight the importance of regulation of IFITM trafficking for its antiviral activity and reveal a novel mechanism of the cyclosporine-mediated modulation of cell susceptibility to enveloped virus infection.
Collapse
Affiliation(s)
- David Prikryl
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mariana Marin
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Tanay M. Desai
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
- Carl Zeiss Microscopy, White Plains, NY 10601, USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Atlanta, GA 30322, USA
| | - Gregory B. Melikyan
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
28
|
Unali G, Crivicich G, Pagani I, Abou‐Alezz M, Folchini F, Valeri E, Matafora V, Reisz JA, Giordano AMS, Cuccovillo I, Butta GM, Donnici L, D'Alessandro A, De Francesco R, Manganaro L, Cittaro D, Merelli I, Petrillo C, Bachi A, Vicenzi E, Kajaste‐Rudnitski A. Interferon-inducible phospholipids govern IFITM3-dependent endosomal antiviral immunity. EMBO J 2023; 42:e112234. [PMID: 36970857 PMCID: PMC10183820 DOI: 10.15252/embj.2022112234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
The interferon-induced transmembrane proteins (IFITM) are implicated in several biological processes, including antiviral defense, but their modes of action remain debated. Here, taking advantage of pseudotyped viral entry assays and replicating viruses, we uncover the requirement of host co-factors for endosomal antiviral inhibition through high-throughput proteomics and lipidomics in cellular models of IFITM restriction. Unlike plasma membrane (PM)-localized IFITM restriction that targets infectious SARS-CoV2 and other PM-fusing viral envelopes, inhibition of endosomal viral entry depends on lysines within the conserved IFITM intracellular loop. These residues recruit Phosphatidylinositol 3,4,5-trisphosphate (PIP3) that we show here to be required for endosomal IFITM activity. We identify PIP3 as an interferon-inducible phospholipid that acts as a rheostat for endosomal antiviral immunity. PIP3 levels correlated with the potency of endosomal IFITM restriction and exogenous PIP3 enhanced inhibition of endocytic viruses, including the recent SARS-CoV2 Omicron variant. Together, our results identify PIP3 as a critical regulator of endosomal IFITM restriction linking it to the Pi3K/Akt/mTORC pathway and elucidate cell-compartment-specific antiviral mechanisms with potential relevance for the development of broadly acting antiviral strategies.
Collapse
Affiliation(s)
- Giulia Unali
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| | - Giovanni Crivicich
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Isabel Pagani
- Viral Pathogenesis and Biosafety UnitIRCCS Ospedale San RaffaeleMilanItaly
| | - Monah Abou‐Alezz
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Filippo Folchini
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Erika Valeri
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| | | | - Julie A Reisz
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraCOUSA
| | - Anna Maria Sole Giordano
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Giacomo M Butta
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi"MilanItaly
- Department of Pharmacological and Biomolecular Sciences (DiSFeB)University of MilanMilanItaly
| | - Lorena Donnici
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi"MilanItaly
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraCOUSA
| | - Raffaele De Francesco
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi"MilanItaly
- Department of Pharmacological and Biomolecular Sciences (DiSFeB)University of MilanMilanItaly
| | - Lara Manganaro
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi"MilanItaly
- Department of Pharmacological and Biomolecular Sciences (DiSFeB)University of MilanMilanItaly
| | - Davide Cittaro
- Center for Omics SciencesIRCCS Ospedale San RaffaeleMilanItaly
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Carolina Petrillo
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Angela Bachi
- FIRC Institute of Molecular Oncology (IFOM)MilanItaly
| | - Elisa Vicenzi
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| | - Anna Kajaste‐Rudnitski
- San Raffaele Telethon Institute for Gene Therapy (SR‐TIGET), IRCCS Ospedale San RaffaeleMilanItaly
| |
Collapse
|
29
|
Son Y, Lee CG, Kim JS, Lee HJ. Low-dose-rate ionizing radiation affects innate immunity protein IFITM3 in a mouse model of Alzheimer's disease. Int J Radiat Biol 2023; 99:1649-1659. [PMID: 37162420 DOI: 10.1080/09553002.2023.2211142] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/17/2023] [Indexed: 05/11/2023]
Abstract
PURPOSE Although the adverse health risks associated with low-dose radiation (LDR) are highly debated, relevant data on neuronal function following chronic LDR exposure are still lacking. MATERIALS AND METHODS To confirm the effect of chronic LDR on the progression of Alzheimer's disease (AD), we investigated changes in behavior and neuroinflammation after radiation exposure in wild-type (WT) and 5xFAD (TG) mice, an animal model of AD. WT and TG mice, classified by genotyping, were exposed to low-dose-rate radiation for 112 days, with cumulative doses of 0, 0.1, and 0.3 Gy, then evaluated using the open-field and Y-maze behavioral function tests. Changes in the levels of APP processing- and neuroinflammation-related genes were also investigated. RESULTS No apparent change was evident in either non-spatial memory function or locomotor activity, as examined by the Y-maze and open field tests, respectively. Although chronic LDR did not affect the levels of APP processing, gliosis (Iba1 and GFAP), or inflammatory cytokines (IL-1β, IL-6, and TNF-α), the levels of IFN-γ were significantly downregulated in TG mice following LDR exposure. In an additional analysis, we examined the genes related to IFN signaling and found that the levels of interferon induced transmembrane protein 3 (IFITM3) were decreased significantly in TG mice following LDR with 0.1 or 0.3 Gy. CONCLUSIONS Therefore, this study revealed the possibility that LDR could affect the progression of AD, which may be associated with decreased IFN-related signaling, especially IFITM3. Our findings suggest that further studies are required regarding the potential role of LDR in the progression of AD.
Collapse
Affiliation(s)
- Yeonghoon Son
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Chang Geun Lee
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Joong Sun Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences (KIRAMS), Seoul, Republic of Korea
| |
Collapse
|
30
|
Taurodeoxycholic acid-YAP1 upregulates OTX1 in promoting gallbladder cancer malignancy through IFITM3-dependent AKT activation. Oncogene 2023; 42:1466-1477. [PMID: 36928361 DOI: 10.1038/s41388-023-02660-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
Orthodenticle homeobox (OTX1) is reported to be involved in numerous cancers, but the expression level and molecular function of OTX1 in gallbladder cancer (GBC) remain unknown. Here, we found the elevated level of OTX1 associated with poor prognosis in human gallbladder cancer. In vitro and in vivo studies of human gallbladder cancer cell lines demonstrated that overexpression of OTX1 promoted cell proliferation, whereas the downregulation inhibited it. Additionally, we found a tight correlation between the serum level of taurodeoxycholic acid (TDCA) and OTX1 expression. TDCA-induced activation of YAP1 by phosphorylation inhibition contributed to the transcriptional activation of OTX1. Mechanistically, we identified that OTX1 activated AKT signaling pathway by transactivating the expression of IFITM3 and thus promoted the proliferation of GBC cells. Taken together, our results showed that TDCA-YAP1-dependent expression of OTX1 regulated IFITM3 and affected GBC proliferation via the AKT signaling pathway. Our experiments also suggested that OTX1 is a novel therapeutic target for GBC.
Collapse
|
31
|
Basile A, Zannella C, De Marco M, Sanna G, Franci G, Galdiero M, Manzin A, De Laurenzi V, Chetta M, Rosati A, Turco MC, Marzullo L. Spike-mediated viral membrane fusion is inhibited by a specific anti-IFITM2 monoclonal antibody. Antiviral Res 2023; 211:105546. [PMID: 36669656 DOI: 10.1016/j.antiviral.2023.105546] [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: 10/13/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
The early steps of viral infection involve protein complexes and structural lipid rearrangements which characterize the peculiar strategies of each virus to invade permissive host cells. Members of the human immune-related interferon-induced transmembrane (IFITM) protein family have been described as inhibitors of the entry of a broad range of viruses into the host cells. Recently, it has been shown that SARS-CoV-2 is able to hijack IFITM2 for efficient infection. Here, we report the characterization of a newly generated specific anti-IFITM2 mAb able to impair Spike-mediated internalization of SARS-CoV-2 in host cells and, consequently, to reduce the SARS-CoV-2 cytopathic effects and syncytia formation. Furthermore, the anti-IFITM2 mAb reduced HSVs- and RSV-dependent cytopathic effects, suggesting that the IFITM2-mediated mechanism of host cell invasion might be shared with other viruses besides SARS-CoV-2. These results show the specific role of IFITM2 in mediating viral entry into the host cell and its candidacy as a cell target for antiviral therapeutic strategies.
Collapse
Affiliation(s)
- Anna Basile
- Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, Baronissi, SA, 84081, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
| | - Margot De Marco
- Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, Baronissi, SA, 84081, Italy; FIBROSYS s.r.l., University of Salerno, Baronissi, SA, 84081, Italy
| | - Giuseppina Sanna
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, Baronissi, SA, 84081, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
| | - Aldo Manzin
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, 09042, Italy
| | - Vincenzo De Laurenzi
- FIBROSYS s.r.l., University of Salerno, Baronissi, SA, 84081, Italy; Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, Chieti, Italy
| | - Massimiliano Chetta
- U.O.C. Medical and Laboratory Genetics, A.O.R.N, Cardarelli, Naples, 80131, Italy
| | - Alessandra Rosati
- Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, Baronissi, SA, 84081, Italy; FIBROSYS s.r.l., University of Salerno, Baronissi, SA, 84081, Italy.
| | - Maria Caterina Turco
- Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, Baronissi, SA, 84081, Italy; FIBROSYS s.r.l., University of Salerno, Baronissi, SA, 84081, Italy
| | - Liberato Marzullo
- Department of Medicine, Surgery and Dentistry "Schola Medica Salernitana", University of Salerno, Baronissi, SA, 84081, Italy; FIBROSYS s.r.l., University of Salerno, Baronissi, SA, 84081, Italy
| |
Collapse
|
32
|
Escudero-Pérez B, Lalande A, Mathieu C, Lawrence P. Host–Pathogen Interactions Influencing Zoonotic Spillover Potential and Transmission in Humans. Viruses 2023; 15:v15030599. [PMID: 36992308 PMCID: PMC10060007 DOI: 10.3390/v15030599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Emerging infectious diseases of zoonotic origin are an ever-increasing public health risk and economic burden. The factors that determine if and when an animal virus is able to spill over into the human population with sufficient success to achieve ongoing transmission in humans are complex and dynamic. We are currently unable to fully predict which pathogens may appear in humans, where and with what impact. In this review, we highlight current knowledge of the key host–pathogen interactions known to influence zoonotic spillover potential and transmission in humans, with a particular focus on two important human viruses of zoonotic origin, the Nipah virus and the Ebola virus. Namely, key factors determining spillover potential include cellular and tissue tropism, as well as the virulence and pathogenic characteristics of the pathogen and the capacity of the pathogen to adapt and evolve within a novel host environment. We also detail our emerging understanding of the importance of steric hindrance of host cell factors by viral proteins using a “flytrap”-type mechanism of protein amyloidogenesis that could be crucial in developing future antiviral therapies against emerging pathogens. Finally, we discuss strategies to prepare for and to reduce the frequency of zoonotic spillover occurrences in order to minimize the risk of new outbreaks.
Collapse
Affiliation(s)
- Beatriz Escudero-Pérez
- WHO Collaborating Centre for Arbovirus and Haemorrhagic Fever Reference and Research, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, 38124 Braunschweig, Germany
| | - Alexandre Lalande
- CIRI (Centre International de Recherche en Infectiologie), Team Neuro-Invasion, TROpism and VIRal Encephalitis, INSERM U1111, CNRS UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007 Lyon, France
| | - Cyrille Mathieu
- CIRI (Centre International de Recherche en Infectiologie), Team Neuro-Invasion, TROpism and VIRal Encephalitis, INSERM U1111, CNRS UMR5308, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, 69007 Lyon, France
| | - Philip Lawrence
- CONFLUENCE: Sciences et Humanités (EA 1598), Université Catholique de Lyon (UCLy), 69002 Lyon, France
- Correspondence:
| |
Collapse
|
33
|
Villanueva-Aguilar ME, Rizo-de-la-Torre LDC, Granados-Muñiz MDP, Montoya-Fuentes A, Montoya-Fuentes H. The Genetic Variant TNFA (rs361525) Is Associated with Increased Susceptibility to Developing Dengue Symptoms. Viral Immunol 2023; 36:229-237. [PMID: 36730734 DOI: 10.1089/vim.2022.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Dengue virus (DENV) is the causal agent of dengue fever. The symptoms and signs of dengue vary from febrile illness to hemorrhagic syndrome. IFITM3 and TNFA are genes of the innate immune system. Variants IFITM3 (rs12252 T>C) and TNFA (rs1800629 G > A and rs361525 G>A) might alter gene expression and change the course of the disease. Our first objective was to determine whether these variants were associated with the susceptibility and severity of dengue. The second was to assess the association of these variants with each symptom. We studied 272 cases with suspected dengue infection, of which 102 were confirmed dengue cases (DENV+) and 170 were dengue-like cases without DENV infection (DENV-). Samples of 201 individuals from the general population of Mexico were included as a reference. Genotyping was performed by the polymerase chain reaction-restriction fragment length polymorphism technique. Odds ratios and confidence intervals were calculated using Pearson's chi-square test and later adjusted for age and sex with a binary logistic regression model. Haldane correction is applied when necessary. We found a significantly higher frequency of the A allele of TNFA rs361525 in both the DENV+ and DENV- groups compared with the general population. Focusing on DENV+ and DENV-, the frequency of the A allele of TNFA rs361525 was higher in the DENV+ group. A broad spectrum of symptoms was related to the A allele of both TNFA variants. We conclude that TNFA rs361525 increases the susceptibility to symptomatic dengue but can also be associated with susceptibility to other dengue-like symptoms from unknown causes.
Collapse
Affiliation(s)
- Mónica Edith Villanueva-Aguilar
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México.,Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UDG), Guadalajara, Jalisco, México
| | - Lourdes Del Carmen Rizo-de-la-Torre
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| | - María Del Pilar Granados-Muñiz
- Laboratorio de Apoyo a la Vigilancia e Investigación Epidemiológica (LAVIE), Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| | - Andrea Montoya-Fuentes
- Laboratorio de Apoyo a la Vigilancia e Investigación Epidemiológica (LAVIE), Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| | - Héctor Montoya-Fuentes
- División de Medicina Molecular, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México.,Laboratorio de Apoyo a la Vigilancia e Investigación Epidemiológica (LAVIE), Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, México
| |
Collapse
|
34
|
Confort MP, Duboeuf M, Thiesson A, Pons L, Marziali F, Desloire S, Ratinier M, Cimarelli A, Arnaud F. IFITMs from Naturally Infected Animal Species Exhibit Distinct Restriction Capacities against Toscana and Rift Valley Fever Viruses. Viruses 2023; 15:v15020306. [PMID: 36851520 PMCID: PMC9965546 DOI: 10.3390/v15020306] [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: 01/06/2023] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Rift Valley Fever virus (RVFV) and Toscana virus (TOSV) are two pathogenic arthropod-borne viruses responsible for zoonotic infections in both humans and animals; as such, they represent a growing threat to public and veterinary health. Interferon-induced transmembrane (IFITM) proteins are broad inhibitors of a large panel of viruses belonging to various families and genera. However, little is known on the interplay between RVFV, TOSV, and the IFITM proteins derived from their naturally infected host species. In this study, we investigated the ability of human, bovine, and camel IFITMs to restrict RVFV and TOSV infection. Our results indicated that TOSV was extremely sensitive to inhibition by all the animal IFITMs tested, while RVFV was inhibited by human IFITM-2 and IFITM-3, but not IFITM-1, and exhibited a more heterogeneous resistance phenotype towards the individual bovine and camel IFITMs tested. Overall, our findings shed some light on the complex and differential interplay between two zoonotic viruses and IFITMs from their naturally infected animal species.
Collapse
Affiliation(s)
- Marie-Pierre Confort
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, PSL University, F-69007 Lyon, France
| | - Maëva Duboeuf
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, PSL University, F-69007 Lyon, France
| | - Adrien Thiesson
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, PSL University, F-69007 Lyon, France
| | - Léa Pons
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Nationale Supérieure de Lyon, F-69342 Lyon, France
| | - Federico Marziali
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Nationale Supérieure de Lyon, F-69342 Lyon, France
| | - Sophie Desloire
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, PSL University, F-69007 Lyon, France
| | - Maxime Ratinier
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, PSL University, F-69007 Lyon, France
| | - Andrea Cimarelli
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Nationale Supérieure de Lyon, F-69342 Lyon, France
- Correspondence: (A.C.); (F.A.); Tel.: +33-(0)-4-7272-8696 (A.C.); +33-4-3728-7612 (F.A.); Fax: +33-(0)-4-7272-8137 (A.C.); +33-4-3728-7605 (F.A.)
| | - Frédérick Arnaud
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, PSL University, F-69007 Lyon, France
- Correspondence: (A.C.); (F.A.); Tel.: +33-(0)-4-7272-8696 (A.C.); +33-4-3728-7612 (F.A.); Fax: +33-(0)-4-7272-8137 (A.C.); +33-4-3728-7605 (F.A.)
| |
Collapse
|
35
|
Extracellular Vesicles and Viruses: Two Intertwined Entities. Int J Mol Sci 2023; 24:ijms24021036. [PMID: 36674550 PMCID: PMC9861478 DOI: 10.3390/ijms24021036] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Viruses share many attributes in common with extracellular vesicles (EVs). The cellular machinery that is used for EV production, packaging of substrates and secretion is also commonly manipulated by viruses for replication, assembly and egress. Viruses can increase EV production or manipulate EVs to spread their own genetic material or proteins, while EVs can play a key role in regulating viral infections by transporting immunomodulatory molecules and viral antigens to initiate antiviral immune responses. Ultimately, the interactions between EVs and viruses are highly interconnected, which has led to interesting discoveries in their associated roles in the progression of different diseases, as well as the new promise of combinational therapeutics. In this review, we summarize the relationships between viruses and EVs and discuss major developments from the past five years in the engineering of virus-EV therapies.
Collapse
|
36
|
Lou K, Wassarman DR, Yang T, Paung Y, Zhang Z, O’Loughlin TA, Moore MK, Egan RK, Greninger P, Benes CH, Seeliger MA, Taunton J, Gilbert LA, Shokat KM. IFITM proteins assist cellular uptake of diverse linked chemotypes. Science 2022; 378:1097-1104. [PMID: 36480603 PMCID: PMC9924227 DOI: 10.1126/science.abl5829] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The search for cell-permeable drugs has conventionally focused on low-molecular weight (MW), nonpolar, rigid chemical structures. However, emerging therapeutic strategies break traditional drug design rules by employing flexibly linked chemical entities composed of more than one ligand. Using complementary genome-scale chemical-genetic approaches we identified an endogenous chemical uptake pathway involving interferon-induced transmembrane proteins (IFITMs) that modulates the cell permeability of a prototypical biopic inhibitor of MTOR (RapaLink-1, MW: 1784 g/mol). We devised additional linked inhibitors targeting BCR-ABL1 (DasatiLink-1, MW: 1518 g/mol) and EIF4A1 (BisRoc-1, MW: 1466 g/mol), uptake of which was facilitated by IFITMs. We also found that IFITMs moderately assisted some proteolysis-targeting chimeras and examined the physicochemical requirements for involvement of this uptake pathway.
Collapse
Affiliation(s)
- Kevin Lou
- Department of Cellular and Molecular Pharmacology,
University of California, San Francisco, San Francisco, CA 94158, United
States
- Howard Hughes Medical Institute, University of California,
San Francisco, San Francisco, CA 94158, United States
| | - Douglas R. Wassarman
- Department of Cellular and Molecular Pharmacology,
University of California, San Francisco, San Francisco, CA 94158, United
States
- Howard Hughes Medical Institute, University of California,
San Francisco, San Francisco, CA 94158, United States
| | - Tangpo Yang
- Department of Cellular and Molecular Pharmacology,
University of California, San Francisco, San Francisco, CA 94158, United
States
| | - YiTing Paung
- Department of Pharmacological Sciences, Stony Brook
University, Stony Brook, New York 11794-8651, United States
| | - Ziyang Zhang
- Department of Cellular and Molecular Pharmacology,
University of California, San Francisco, San Francisco, CA 94158, United
States
- Howard Hughes Medical Institute, University of California,
San Francisco, San Francisco, CA 94158, United States
- Department of Chemistry, University of California,
Berkeley, Berkeley, 94720, CA, United States
| | - Thomas A. O’Loughlin
- Helen Diller Family Comprehensive Cancer Center, University
of California, San Francisco, San Francisco, CA 94158, United States
- Department of Urology, University of California, San
Francisco, San Francisco, CA 94158, United States
| | - Megan K. Moore
- Department of Cellular and Molecular Pharmacology,
University of California, San Francisco, San Francisco, CA 94158, United
States
- Howard Hughes Medical Institute, University of California,
San Francisco, San Francisco, CA 94158, United States
| | - Regina K. Egan
- Center for Cancer Research, Massachusetts General Hospital
Cancer Center, Charlestown, MA 02129, United States
| | - Patricia Greninger
- Center for Cancer Research, Massachusetts General Hospital
Cancer Center, Charlestown, MA 02129, United States
| | - Cyril H. Benes
- Center for Cancer Research, Massachusetts General Hospital
Cancer Center, Charlestown, MA 02129, United States
- Department of Medicine, Harvard Medical School, Boston, MA
02115, United States
| | - Markus A. Seeliger
- Department of Pharmacological Sciences, Stony Brook
University, Stony Brook, New York 11794-8651, United States
| | - Jack Taunton
- Department of Cellular and Molecular Pharmacology,
University of California, San Francisco, San Francisco, CA 94158, United
States
| | - Luke A. Gilbert
- Helen Diller Family Comprehensive Cancer Center, University
of California, San Francisco, San Francisco, CA 94158, United States
- Department of Urology, University of California, San
Francisco, San Francisco, CA 94158, United States
- Innovative Genomics Institute, University of California,
San Francisco, San Francisco, CA 94158, United States
- Arc Institute, Palo Alto, CA, 94304, United States
| | - Kevan M. Shokat
- Department of Cellular and Molecular Pharmacology,
University of California, San Francisco, San Francisco, CA 94158, United
States
- Howard Hughes Medical Institute, University of California,
San Francisco, San Francisco, CA 94158, United States
- Department of Chemistry, University of California,
Berkeley, Berkeley, 94720, CA, United States
| |
Collapse
|
37
|
Braun AH, Frank AM, Ho N, Buchholz CJ. Dasatinib is a potent enhancer for CAR T cell generation by CD3-targeted lentiviral vectors. Mol Ther Methods Clin Dev 2022; 28:90-98. [PMID: 36620073 PMCID: PMC9801082 DOI: 10.1016/j.omtm.2022.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
CD3-targeted lentiviral vectors (CD3-LVs) mediate selective transduction of human T lymphocytes in vitro and in vivo while simultaneously activating the targeted cells. Previously, we have demonstrated that CD3-LV leads to downmodulation of the CD3:T cell receptor (TCR) complex. We therefore hypothesized that inhibition of CD3 phosphorylation by Src/Abl tyrosine kinase inhibitors such as dasatinib results in enhancement of gene delivery by T cell-targeted LVs. Indeed, dasatinib treatment of T cells prior to incubation with CD3-LV increased reporter gene delivery by 3- to 10-fold. Moreover, the presence of dasatinib enhanced selective transduction into non-activated target cells present in whole blood. When combined with delivery of the CD19-chimeric antigen receptor (CAR) gene, dasatinib increased CAR T cell numbers by close to 10-fold. Importantly, the short-term exposure of T cells to dasatinib during vector incubation did not interfere with tumor cell killing by the resulting CAR T cells and rather came along with less upregulated exhaustion markers and a more naive phenotype. Our data suggest that dasatinib prevents CD3-LV-induced phosphorylation and CD3:TCR intake, thereby increasing the amount of CD3-LV bound to the cell surface. This is the first description of dasatinib as transduction enhancer, an activity particularly relevant for CAR T cell generation with CD3-LV.
Collapse
Affiliation(s)
- Angela H. Braun
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Annika M. Frank
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Naphang Ho
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Christian J. Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany,Corresponding author Christian J. Buchholz, Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany.
| |
Collapse
|
38
|
Transcriptomic analysis of the innate immune response to in vitro transfection of plasmid DNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 31:43-56. [PMID: 36618265 PMCID: PMC9800263 DOI: 10.1016/j.omtn.2022.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
The innate immune response to cytosolic DNA is intended to protect the host from viral infections, but it can also inhibit the delivery and expression of therapeutic transgenes in gene and cell therapies. The goal of this work was to use mRNA sequencing to identify genes that may influence transfection efficiency in four different cell types (PC-3, Jurkat, HEK-293T, and primary T cells). The highest transfection efficiency was observed in HEK-293T cells, which upregulated only 142 genes with no known antiviral functions after transfection with lipofectamine. Lipofection upregulated 1,057 cytokine-stimulated genes (CSGs) in PC-3 cells, which exhibited a significantly lower transfection efficiency. However, when PC-3 cells were transfected in serum-containing media or electroporated, the observed transfection efficiencies were significantly higher while the expression levels of cytokines and CSGs decreased. In contrast, lipofection of Jurkat and primary T cells only upregulated a few genes, but several of the antiviral CSGs that were absent in HEK-293T cells and upregulated in PC-3 cells were observed to be constitutively expressed in T cells, which may explain the relatively low Lipofection efficiencies observed with T cells (8%-21% GFP+). Indeed, overexpression of one CSG (IFI16) significantly decreased transfection efficiency in HEK-293T cells.
Collapse
|
39
|
Kehn-Hall K, Bradfute SB. Understanding host responses to equine encephalitis virus infection: implications for therapeutic development. Expert Rev Anti Infect Ther 2022; 20:1551-1566. [PMID: 36305549 DOI: 10.1080/14787210.2022.2141224] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV) are mosquito-borne New World alphaviruses that cause encephalitis in equids and humans. These viruses can cause severe disease and death, as well as long-term severe neurological symptoms in survivors. Despite the pathogenesis and weaponization of these viruses, there are no approved therapeutics for treating infection. AREAS COVERED In this review, we describe the molecular pathogenesis of these viruses, discuss host-pathogen interactions needed for viral replication, and highlight new avenues for drug development with a focus on host-targeted approaches. EXPERT OPINION Current approaches have yielded some promising therapeutics, but additional emphasis should be placed on advanced development of existing small molecules and pursuit of pan-encephalitic alphavirus drugs. More research should be conducted on EEEV and WEEV, given their high lethality rates.
Collapse
Affiliation(s)
- Kylene Kehn-Hall
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.,Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Steven B Bradfute
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
40
|
IFITM proteins: Understanding their diverse roles in viral infection, cancer, and immunity. J Biol Chem 2022; 299:102741. [PMID: 36435199 PMCID: PMC9800550 DOI: 10.1016/j.jbc.2022.102741] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/27/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs) are broad spectrum antiviral factors that inhibit the entry of a wide range of clinically important pathogens including influenza A virus, HIV-1, and Dengue virus. IFITMs are thought to act primarily by antagonizing virus-cell membrane fusion in this regard. However, recent work on these proteins has uncovered novel post-entry viral restriction mechanisms. IFITMs are also increasingly thought to have a role regulating immune responses, including innate antiviral and inflammatory responses as well as adaptive T-cell and B-cell responses. Further, IFITMs may have pathological activities in cancer, wherein IFITM expression can be a marker of therapeutically resistant and aggressive disease courses. In this review, we summarize the respective literatures concerning these apparently diverse functions with a view to identifying common themes and potentially yielding a more unified understanding of IFITM biology.
Collapse
|
41
|
Wang Z, Tuo X, Zhang J, Chai K, Tan J, Qiao W. Antiviral role of IFITM3 in prototype foamy virus infection. Virol J 2022; 19:195. [PMID: 36419065 PMCID: PMC9682733 DOI: 10.1186/s12985-022-01931-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Foamy viruses (FVs) are retroviruses with unique replication strategies that cause lifelong latent infections in their hosts. FVs can also produce foam-like cytopathic effects in vitro. However, the effect of host cytokines on FV replication requires further investigation. Although interferon induced transmembrane (IFITMs) proteins have become the focus of antiviral immune response research due to their broad-spectrum antiviral ability, it remains unclear whether IFITMs can affect FV replication. METHOD In this study, the PFV virus titer was characterized by measuring luciferase activity after co-incubation of PFVL cell lines with the cell culture supernatants (cell-free PFV) or the cells transfected with pcPFV plasmid/infected with PFV (cell-associated PFV). The foam-like cytopathic effects of PFV infected cells was observed to reflect the virus replication. The total RNA of PFV infected cells was extracted, and the viral genome was quantified by Quantitative reverse transcription PCR to detect the PFV entry into target cells. RESULTS In the present study, we demonstrated that IFITM1-3 overexpression inhibited prototype foamy virus (PFV) replication. In addition, an IFITM3 knockdown by small interfering RNA increased PFV replication. We further demonstrated that IFITM3 inhibited PFV entry into host cells. Moreover, IFITM3 also reduced the number of PFV envelope proteins, which was related to IFITM3 promoted envelope degradation through the lysosomal pathway. CONCLUSIONS Taken together, these results demonstrate that IFITM3 inhibits PFV replication by inhibiting PFV entry into target cells and reducing the number of PFV envelope.
Collapse
Affiliation(s)
- Zhaohuan Wang
- grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Xiaopeng Tuo
- grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071 China ,Present Address: Merck Sharp & Dohme Corp, Building 21, Rongda Road, Chaoyang District, Beijing, 1000102 People’s Republic of China
| | - Junshi Zhang
- grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071 China ,grid.417031.00000 0004 1799 2675Present Address: Department of Hematology, Oncology Centrer, Tianjin People’s Hospital, No. 190 Jieyuan Road, Hongqiao District, Tianjin, 300121 People’s Republic of China
| | - Keli Chai
- grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071 China ,grid.417303.20000 0000 9927 0537Present Address: Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221002 Jiangsu China
| | - Juan Tan
- grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071 China
| | - Wentao Qiao
- grid.216938.70000 0000 9878 7032Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071 China
| |
Collapse
|
42
|
Friedlová N, Zavadil Kokáš F, Hupp TR, Vojtěšek B, Nekulová M. IFITM protein regulation and functions: Far beyond the fight against viruses. Front Immunol 2022; 13:1042368. [PMID: 36466909 PMCID: PMC9716219 DOI: 10.3389/fimmu.2022.1042368] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Interferons (IFNs) are important cytokines that regulate immune responses through the activation of hundreds of genes, including interferon-induced transmembrane proteins (IFITMs). This evolutionarily conserved protein family includes five functionally active homologs in humans. Despite the high sequence homology, IFITMs vary in expression, subcellular localization and function. The initially described adhesive and antiproliferative or pro-oncogenic functions of IFITM proteins were diluted by the discovery of their antiviral properties. The large set of viruses that is inhibited by these proteins is constantly expanding, as are the possible mechanisms of action. In addition to their beneficial antiviral effects, IFITM proteins are often upregulated in a broad spectrum of cancers. IFITM proteins have been linked to most hallmarks of cancer, including tumor cell proliferation, therapeutic resistance, angiogenesis, invasion, and metastasis. Recent studies have described the involvement of IFITM proteins in antitumor immunity. This review summarizes various levels of IFITM protein regulation and the physiological and pathological functions of these proteins, with an emphasis on tumorigenesis and antitumor immunity.
Collapse
Affiliation(s)
- Nela Friedlová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Filip Zavadil Kokáš
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Ted R. Hupp
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bořivoj Vojtěšek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Marta Nekulová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
43
|
Li H, Ni R, Wang K, Tian Y, Gong H, Yan W, Tang Y, Lei C, Wang H, Yang X. Chicken interferon-induced transmembrane protein 1 promotes replication of coronavirus infectious bronchitis virus in a cell-specific manner. Vet Microbiol 2022; 275:109597. [PMCID: PMC9616511 DOI: 10.1016/j.vetmic.2022.109597] [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: 06/29/2022] [Revised: 10/06/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2022]
Abstract
Interferon-induced transmembrane proteins (IFITMs) are broad-spectrum antiviral proteins that inhibit numerous virus infections by impeding viral entry into target cells. However, increasing evidence suggests diverse functions of IFITMs in virus infection, especially with the coronavirus. We analyzed the effect of chicken interferon-induced transmembrane proteins (chIFITMs) on coronavirus infectious bronchitis virus (IBV) infection in vitro. We demonstrated that the antiviral effects of IFITMs are dependent on cell and virus types. The overexpression of chIFITM1 dramatically promoted the replication of IBV Beaudette strain in the chicken hepatocellular carcinoma cell line, LMH. Mechanistically, chIFITMs share roughly the same subcellular localization in different host cells, and overexpressed of chIFITM1 have no effect of viral attachment and entry. Further studies revealed that mutations of amino acids at key positions (60KSRD63, 68KDFV71) in the intracellular loop domain (CIL) caused loss of the promoted function. Interaction with downstream proteins in co-response to viral infection could be the primary reason behind variable functions of chIFITM1 in different cells. In all, our study explored the functions of chIFITMs in viral infection from a new perspective.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Ruiqi Ni
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Kailu Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yiming Tian
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, China
| | - Huilin Gong
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Wenjun Yan
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yizhi Tang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Changwei Lei
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hongning Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Xin Yang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China,Corresponding author at: Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China
| |
Collapse
|
44
|
Peters BA, Pass HI, Burk RD, Xue X, Goparaju C, Sollecito CC, Grassi E, Segal LN, Tsay JCJ, Hayes RB, Ahn J. The lung microbiome, peripheral gene expression, and recurrence-free survival after resection of stage II non-small cell lung cancer. Genome Med 2022; 14:121. [PMID: 36303210 PMCID: PMC9609265 DOI: 10.1186/s13073-022-01126-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Cancer recurrence after tumor resection in early-stage non-small cell lung cancer (NSCLC) is common, yet difficult to predict. The lung microbiota and systemic immunity may be important modulators of risk for lung cancer recurrence, yet biomarkers from the lung microbiome and peripheral immune environment are understudied. Such markers may hold promise for prediction as well as improved etiologic understanding of lung cancer recurrence. METHODS In tumor and distant normal lung samples from 46 stage II NSCLC patients with curative resection (39 tumor samples, 41 normal lung samples), we conducted 16S rRNA gene sequencing. We also measured peripheral blood immune gene expression with nanoString®. We examined associations of lung microbiota and peripheral gene expression with recurrence-free survival (RFS) and disease-free survival (DFS) using 500 × 10-fold cross-validated elastic-net penalized Cox regression, and examined predictive accuracy using time-dependent receiver operating characteristic (ROC) curves. RESULTS Over a median of 4.8 years of follow-up (range 0.2-12.2 years), 43% of patients experienced a recurrence, and 50% died. In normal lung tissue, a higher abundance of classes Bacteroidia and Clostridia, and orders Bacteroidales and Clostridiales, were associated with worse RFS, while a higher abundance of classes Alphaproteobacteria and Betaproteobacteria, and orders Burkholderiales and Neisseriales, were associated with better RFS. In tumor tissue, a higher abundance of orders Actinomycetales and Pseudomonadales were associated with worse DFS. Among these taxa, normal lung Clostridiales and Bacteroidales were also related to worse survival in a previous small pilot study and an additional independent validation cohort. In peripheral blood, higher expression of genes TAP1, TAPBP, CSF2RB, and IFITM2 were associated with better DFS. Analysis of ROC curves revealed that lung microbiome and peripheral gene expression biomarkers provided significant additional recurrence risk discrimination over standard demographic and clinical covariates, with microbiome biomarkers contributing more to short-term (1-year) prediction and gene biomarkers contributing to longer-term (2-5-year) prediction. CONCLUSIONS We identified compelling biomarkers in under-explored data types, the lung microbiome, and peripheral blood gene expression, which may improve risk prediction of recurrence in early-stage NSCLC patients. These findings will require validation in a larger cohort.
Collapse
Affiliation(s)
- Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, #1315AB, The Bronx, New York, NY, 10461, USA.
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
- NYU Perlmutter Cancer Center, New York, NY, USA
| | - Robert D Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, #1315AB, The Bronx, New York, NY, 10461, USA
- Department of Pediatrics, Albert Einstein College of Medicine, The Bronx, New York, NY, USA
- Department of Microbiology & Immunology, and Obstetrics & Gynecology & Women's Health, Albert Einstein College of Medicine, The Bronx, New York, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, #1315AB, The Bronx, New York, NY, 10461, USA
| | - Chandra Goparaju
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | | | - Evan Grassi
- Department of Pediatrics, Albert Einstein College of Medicine, The Bronx, New York, NY, USA
| | | | | | - Richard B Hayes
- NYU Perlmutter Cancer Center, New York, NY, USA
- Department of Population Health, NYU Langone Health, New York, NY, USA
| | - Jiyoung Ahn
- NYU Perlmutter Cancer Center, New York, NY, USA
- Department of Population Health, NYU Langone Health, New York, NY, USA
| |
Collapse
|
45
|
Shilagardi K, Spear ED, Abraham R, Griffin DE, Michaelis S. The Integral Membrane Protein ZMPSTE24 Protects Cells from SARS-CoV-2 Spike-Mediated Pseudovirus Infection and Syncytia Formation. mBio 2022; 13:e0254322. [PMID: 36197088 PMCID: PMC9601121 DOI: 10.1128/mbio.02543-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/26/2022] Open
Abstract
COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has had a devastating impact on global public health, emphasizing the importance of understanding innate immune mechanisms and cellular restriction factors that cells can harness to fight viral infections. The multimembrane-spanning zinc metalloprotease ZMPSTE24 is one such restriction factor. ZMPSTE24 has a well-characterized proteolytic role in the maturation of prelamin A, precursor of the nuclear scaffold protein lamin A. An apparently unrelated role for ZMPSTE24 in viral defense involves its interaction with the interferon-inducible membrane proteins (IFITMs), which block virus-host cell fusion by rigidifying cellular membranes and thereby prevent viral infection. ZMPSTE24, like the IFITMs, defends cells against a broad spectrum of enveloped viruses. However, its ability to protect against coronaviruses has never been examined. Here, we show that overexpression of ZMPSTE24 reduces the efficiency of cellular infection by SARS-CoV-2 Spike-pseudotyped lentivirus and that genetic knockout or small interfering RNA-mediated knockdown of endogenous ZMPSTE24 enhances infectivity. We further demonstrate a protective role for ZMPSTE24 in a Spike-ACE2-dependent cell-cell fusion assay. In both assays, a catalytic dead version of ZMPSTE24 is equally as protective as the wild-type protein, indicating that ZMPSTE24's proteolytic activity is not required for defense against SARS-CoV-2. Finally, we demonstrate by plaque assays that Zmpste24-/- mouse cells show enhanced infection by a genuine coronavirus, mouse hepatitis virus (MHV). This study extends the range of viral protection afforded by ZMPSTE24 to include coronaviruses and suggests that targeting ZMPSTE24's mechanism of viral defense could have therapeutic benefit. IMPORTANCE The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 has underscored the importance of understanding intrinsic cellular components that can be harnessed as the cell's first line of defense to fight against viral infection. Our paper focuses on one such protein, the integral membrane protease ZMPSTE24, which interacts with interferon-inducible transmembrane proteins (IFITMs). IFITMs interfere with virus entry by inhibiting fusion between viral and host cell membranes, and ZMPSTE24 appears to contribute to this inhibitory activity. ZMPSTE24 has been shown to defend cells against several, but not all, enveloped viruses. In this study, we extend ZMPSTE24's reach to include coronaviruses, by showing that ZMPSTE24 protects cells from SARS-CoV-2 pseudovirus infection, Spike protein-mediated cell-cell fusion, and infection by the mouse coronavirus MHV. This work lays the groundwork for further studies to decipher the mechanistic role of ZMPSTE24 in blocking the entry of SARS-CoV-2 and other viruses into cells.
Collapse
Affiliation(s)
- Khurts Shilagardi
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Eric D. Spear
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Rachy Abraham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Diane E. Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Susan Michaelis
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Lang R, Li H, Luo X, Liu C, Zhang Y, Guo S, Xu J, Bao C, Dong W, Yu Y. Expression and mechanisms of interferon-stimulated genes in viral infection of the central nervous system (CNS) and neurological diseases. Front Immunol 2022; 13:1008072. [PMID: 36325336 PMCID: PMC9618809 DOI: 10.3389/fimmu.2022.1008072] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/28/2022] [Indexed: 09/16/2023] Open
Abstract
Interferons (IFNs) bind to cell surface receptors and activate the expression of interferon-stimulated genes (ISGs) through intracellular signaling cascades. ISGs and their expression products have various biological functions, such as antiviral and immunomodulatory effects, and are essential effector molecules for IFN function. ISGs limit the invasion and replication of the virus in a cell-specific and region-specific manner in the central nervous system (CNS). In addition to participating in natural immunity against viral infections, studies have shown that ISGs are essential in the pathogenesis of CNS disorders such as neuroinflammation and neurodegenerative diseases. The aim of this review is to present a macroscopic overview of the characteristics of ISGs that restrict viral neural invasion and the expression of the ISGs underlying viral infection of CNS cells. Furthermore, we elucidate the characteristics of ISGs expression in neurological inflammation, neuropsychiatric disorders such as depression as well as neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Finally, we summarize several ISGs (ISG15, IFIT2, IFITM3) that have been studied more in recent years for their antiviral infection in the CNS and their research progress in neurological diseases.
Collapse
Affiliation(s)
- Rui Lang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huiting Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiaoqin Luo
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Cencen Liu
- Department of Pathology, People’s Hospital of Zhongjiang County, DeYang, China
| | - Yiwen Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - ShunYu Guo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingyi Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Changshun Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurological diseases and brain function laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
47
|
Rahman K, Datta SA, Beaven AH, Jolley AA, Sodt AJ, Compton AA. Cholesterol Binds the Amphipathic Helix of IFITM3 and Regulates Antiviral Activity. J Mol Biol 2022; 434:167759. [PMID: 35872070 PMCID: PMC9342930 DOI: 10.1016/j.jmb.2022.167759] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023]
Abstract
The interferon-induced transmembrane (IFITM) proteins broadly inhibit the entry of diverse pathogenic viruses, including Influenza A virus (IAV), Zika virus, HIV-1, and SARS coronaviruses by inhibiting virus-cell membrane fusion. IFITM3 was previously shown to disrupt cholesterol trafficking, but the functional relationship between IFITM3 and cholesterol remains unclear. We previously showed that inhibition of IAV entry by IFITM3 is associated with its ability to promote cellular membrane rigidity, and these activities are functionally linked by a shared requirement for the amphipathic helix (AH) found in the intramembrane domain (IMD) of IFITM3. Furthermore, it has been shown that the AH of IFITM3 alters lipid membranes in vitro in a cholesterol-dependent manner. Therefore, we aimed to elucidate the relationship between IFITM3 and cholesterol in more detail. Using a fluorescence-based in vitro binding assay, we found that a peptide derived from the AH of IFITM3 directly interacted with the cholesterol analog, NBD-cholesterol, while other regions of the IFITM3 IMD did not, and native cholesterol competed with this interaction. In addition, recombinant full-length IFITM3 protein also exhibited NBD-cholesterol binding activity. Importantly, previously characterized mutations within the AH of IFITM3 that strongly inhibit antiviral function (F63Q and F67Q) disrupted AH structure in solution, inhibited cholesterol binding in vitro, and restricted bilayer insertion in silico. Our data suggest that direct interactions with cholesterol may contribute to the inhibition of membrane fusion pore formation by IFITM3. These findings may facilitate the design of therapeutic peptides for use in broad-spectrum antiviral therapy.
Collapse
Affiliation(s)
- Kazi Rahman
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Siddhartha A.K. Datta
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Andrew H. Beaven
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States,National Institute of General Medical Sciences, Bethesda, MD 20892, United States
| | - Abigail A. Jolley
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Alexander J. Sodt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States
| | - Alex A. Compton
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States,Corresponding author
| |
Collapse
|
48
|
Tiwari V, Viswanath S. Identification of potential modulators of IFITM3 by in-silico modeling and virtual screening. Sci Rep 2022; 12:15952. [PMID: 36153346 PMCID: PMC9509314 DOI: 10.1038/s41598-022-20259-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractIFITM3 is a transmembrane protein that confers innate immunity. It has been established to restrict entry of multiple viruses. Overexpression of IFITM3 has been shown to be associated with multiple cancers, implying IFITM3 to be good therapeutic target. The regulation of IFITM3 activity is mediated by multiple post-translational modifications (PTM). In this study, we have modelled the structure of IFITM3, consistent with experimental predictions on its membrane topology. MD simulation in membrane-aqueous environment revealed the stability of the model. Ligand binding sites on the IFITM3 surface were predicted and it was observed that the best site includes important residues involved in PTM and has good druggable score. Molecular docking was performed using FDA approved ligands and natural ligands from Super Natural II database. The ligands were re-ranked by calculating binding free energy. Select docking complexes were simulated again to substantiate the binding between ligand and IFITM3. We observed that known drugs like Eluxadoline and natural products like SN00224572 and Parishin A have good binding affinity against IFITM3. These ligands form persistent interactions with key lysine residues (Lys83, Lys104) and hence can potentially alter the activity of IFITM3. The results of this computational study can provide a starting point for experimental investigations on IFITM3 modulators.
Collapse
|
49
|
Hepatitis Viruses Control Host Immune Responses by Modifying the Exosomal Biogenesis Pathway and Cargo. Int J Mol Sci 2022; 23:ijms231810862. [PMID: 36142773 PMCID: PMC9505460 DOI: 10.3390/ijms231810862] [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: 08/11/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The development of smart immune evasion mechanisms is crucial for the establishment of acute and chronic viral hepatitis. Hepatitis is a major health problem worldwide arising from different causes, such as pathogens, metabolic disorders, and xenotoxins, with the five hepatitis viruses A, B, C, D, and E (HAV, HBV, HCV, HDV, and HEV) representing the majority of the cases. Most of the hepatitis viruses are considered enveloped. Recently, it was reported that the non-enveloped HAV and HEV are, in reality, quasi-enveloped viruses exploiting exosomal-like biogenesis mechanisms for budding. Regardless, all hepatitis viruses use exosomes to egress, regulate, and eventually escape from the host immune system, revealing another key function of exosomes apart from their recognised role in intercellular communication. This review will discuss how the hepatitis viruses exploit exosome biogenesis and transport capacity to establish successful infection and spread. Then, we will outline the contribution of exosomes in viral persistence and liver disease progression.
Collapse
|
50
|
Titanji BK, Wang Z, Chen J, Hui Q, So-Armah K, Freiberg M, Justice AC, Ke X, Marconi VC, Sun YV. Soluble CD14-associated DNA methylation sites predict mortality among men with HIV infection. AIDS 2022; 36:1563-1571. [PMID: 35979830 PMCID: PMC9394925 DOI: 10.1097/qad.0000000000003279] [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] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Elevated plasma levels of sCD14 predict all-cause mortality in people with HIV (PWH). Epigenetic regulation plays a key role in infection and inflammation. To reveal the epigenetic relationships between sCD14, immune function and disease progression among PWH, we conducted an epigenome-wide association study (EWAS) of sCD14 and investigated the relationship with mortality. DESIGN AND METHODS DNA methylation (DNAm) levels of peripheral blood samples from PWH in the Veterans Aging Cohort Study (VACS) were measured using the Illumina Infinium Methylation 450K (n = 549) and EPIC (850K) BeadChip (n = 526). Adjusted for covariates and multiple testing, we conducted an epigenome-wide discovery, replication, and meta-analysis to identify significant associations with sCD14. We then examined and replicated the relationship between the principal epigenetic sites and survival using Cox regression models. FINDINGS We identified 118 DNAm sites significantly associated with sCD14 in the meta-analysis of 1075 PWH. The principal associated DNAm sites mapped to genes (e.g. STAT1, PARP9, IFITM1, MX1, and IFIT1) related to inflammation and antiviral response. Adjusting for multiple testing, 10 of 118 sCD14-associated DNAm sites significantly predicted survival time conditional on sCD14 levels. CONCLUSION The identification of DNAm sites independently predicting survival may improve our understanding of prognosis and potential therapeutic targets among PWH.
Collapse
Affiliation(s)
- Boghuma K Titanji
- Division of Infectious Diseases, Emory University School of Medicine
| | - Zeyuan Wang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Qin Hui
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | | | - Matthew Freiberg
- Cardiovascular Medicine Division and Tennessee Valley Healthcare System, Vanderbilt University Medical Center, Nashville, TN
| | - Amy C Justice
- Yale University School of Medicine, New Haven
- Connecticut Veteran Health System, West Haven, CT
| | - Xu Ke
- Yale University School of Medicine, New Haven
- Connecticut Veteran Health System, West Haven, CT
| | - Vincent C Marconi
- Division of Infectious Diseases, Emory University School of Medicine
- Atlanta Veterans Affairs Healthcare System, Decatur
- Hubert Department of Global Health, Rollins School of Public Health
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
- Atlanta Veterans Affairs Healthcare System, Decatur
| |
Collapse
|