51
|
Janssen E, Tohme M, Butts J, Giguere S, Sage PT, Velázquez FE, Kam C, Milin E, Das M, Sobh A, Al-Tamemi S, Luscinskas FW, Batista F, Geha RS. DOCK8 is essential for LFA-1-dependent positioning of T follicular helper cells in germinal centers. JCI Insight 2020; 5:134508. [PMID: 32573493 DOI: 10.1172/jci.insight.134508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 06/18/2020] [Indexed: 01/07/2023] Open
Abstract
T follicular helper (Tfh) cell migration into germinal centers (GCs) is essential for the generation of GC B cells and antibody responses to T cell-dependent (TD) antigens. This process requires interactions between lymphocyte function-associated antigen 1 (LFA-1) on Tfh cells and ICAMs on B cells. The mechanisms underlying defective antibody responses to TD antigens in DOCK8 deficiency are incompletely understood. We show that mice selectively lacking DOCK8 in T cells had impaired IgG antibody responses to TD antigens, decreased GC size, and reduced numbers of GC B cells. However, they developed normal numbers of Tfh cells with intact capacity for driving B cell differentiation into a GC phenotype in vitro. Notably, migration of DOCK8-deficient T cells into GCs was defective. Following T cell receptor (TCR)/CD3 ligation, DOCK8-deficient T cells had impaired LFA-1 activation and reduced binding to ICAM-1. Our results therefore indicate that DOCK8 is important for LFA-1-dependent positioning of Tfh cells in GCs, and thereby the generation of GC B cells and IgG antibody responses to TD antigen.
Collapse
Affiliation(s)
- Erin Janssen
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mira Tohme
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jordan Butts
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sophie Giguere
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Peter T Sage
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Francisco E Velázquez
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Departments of Pathology and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Christy Kam
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Elena Milin
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mrinmoy Das
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ali Sobh
- Department of Pediatrics, Mansoura University Children's Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Francis W Luscinskas
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Departments of Pathology and Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Facundo Batista
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
52
|
Prinz M, Jung S, Priller J. Microglia Biology: One Century of Evolving Concepts. Cell 2020; 179:292-311. [PMID: 31585077 DOI: 10.1016/j.cell.2019.08.053] [Citation(s) in RCA: 801] [Impact Index Per Article: 200.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/08/2019] [Accepted: 08/27/2019] [Indexed: 01/05/2023]
Abstract
Microglia were first recognized as a distinct cell population in the CNS one century ago. For a long time, they were primarily considered to be phagocytes responsible for removing debris during CNS development and disease. More recently, advances in imaging and genetics and the advent of single-cell technologies provided new insights into the much more complex and fascinating biology of microglia. The ontogeny of microglia was identified, and their functions in health and disease were better defined. Although many questions about microglia and their roles in human diseases remain unanswered, the prospect of targeting microglia for the treatment of neurological and psychiatric disorders is tantalizing.
Collapse
Affiliation(s)
- Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany; DZNE and BIH, Berlin, Germany; University of Edinburgh and UK DRI, Edinburgh, UK.
| |
Collapse
|
53
|
Topology, Antiviral Functional Residues and Mechanism of IFITM1. Viruses 2020; 12:v12030295. [PMID: 32182730 PMCID: PMC7150853 DOI: 10.3390/v12030295] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
Interferon-inducible transmembrane proteins (IFITM1/2/3) have been reported to suppress the entry of a wide range of viruses. However, their antiviral functional residues and specific mechanisms are still unclear. Here, we firstly resolved the topology of IFITM1 on the plasma membrane where N-terminus points into the cytoplasm and C-terminus resides extracellularly. Further, KRRK basic residues of IFITM1 locating at 62–67 of the conserved intracellular loop (CIL) were found to play a key role in the restriction on the Zika virus (ZIKV) and dengue virus (DENV). Similarly, KRRK basic residues of IFITM2/3 also contributed to suppressing ZIKV replication. Finally, IFITM1 was revealed to be capable of restricting the release of ZIKV particles from endosome to cytosol so as to impede the entry of ZIKV into host cells, which was tightly related with the inhibition of IFITM1 on the acidification of organelles. Overall, our study provided topology, antiviral functional residues and the mechanism of interferon-inducible transmembrane proteins.
Collapse
|
54
|
Resveratrol trimer enhances gene delivery to hematopoietic stem cells by reducing antiviral restriction at endosomes. Blood 2020; 134:1298-1311. [PMID: 31416800 DOI: 10.1182/blood.2019000040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022] Open
Abstract
Therapeutic gene delivery to hematopoietic stem cells (HSCs) holds great potential as a life-saving treatment of monogenic, oncologic, and infectious diseases. However, clinical gene therapy is severely limited by intrinsic HSC resistance to modification with lentiviral vectors (LVs), thus requiring high doses or repeat LV administration to achieve therapeutic gene correction. Here we show that temporary coapplication of the cyclic resveratrol trimer caraphenol A enhances LV gene delivery efficiency to human and nonhuman primate hematopoietic stem and progenitor cells with integrating and nonintegrating LVs. Although significant ex vivo, this effect was most dramatically observed in human lineages derived from HSCs transplanted into immunodeficient mice. We further show that caraphenol A relieves restriction of LV transduction by altering the levels of interferon-induced transmembrane (IFITM) proteins IFITM2 and IFITM3 and their association with late endosomes, thus augmenting LV core endosomal escape. Caraphenol A-mediated IFITM downregulation did not alter the LV integration pattern or bias lineage differentiation. Taken together, these findings compellingly demonstrate that the pharmacologic modification of intrinsic immune restriction factors is a promising and nontoxic approach for improving LV-mediated gene therapy.
Collapse
|
55
|
Benfield CT, MacKenzie F, Ritzefeld M, Mazzon M, Weston S, Tate EW, Teo BH, Smith SE, Kellam P, Holmes EC, Marsh M. Bat IFITM3 restriction depends on S-palmitoylation and a polymorphic site within the CD225 domain. Life Sci Alliance 2020; 3:e201900542. [PMID: 31826928 PMCID: PMC6907390 DOI: 10.26508/lsa.201900542] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022] Open
Abstract
Host interferon-induced transmembrane proteins (IFITMs) are broad-spectrum antiviral restriction factors. Of these, IFITM3 potently inhibits viruses that enter cells through acidic endosomes, many of which are zoonotic and emerging viruses with bats (order Chiroptera) as their natural hosts. We previously demonstrated that microbat IFITM3 is antiviral. Here, we show that bat IFITMs are characterized by strong adaptive evolution and identify a highly variable and functionally important site-codon 70-within the conserved CD225 domain of IFITMs. Mutation of this residue in microbat IFITM3 impairs restriction of representatives of four different virus families that enter cells via endosomes. This mutant shows altered subcellular localization and reduced S-palmitoylation, a phenotype copied by mutation of conserved cysteine residues in microbat IFITM3. Furthermore, we show that microbat IFITM3 is S-palmitoylated on cysteine residues C71, C72, and C105, mutation of each cysteine individually impairs virus restriction, and a triple C71A-C72A-C105A mutant loses all restriction activity, concomitant with subcellular re-localization of microbat IFITM3 to Golgi-associated sites. Thus, we propose that S-palmitoylation is critical for Chiropteran IFITM3 function and identify a key molecular determinant of IFITM3 S-palmitoylation.
Collapse
Affiliation(s)
- Camilla To Benfield
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, UK
| | - Farrell MacKenzie
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | | | - Michela Mazzon
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Stuart Weston
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, UK
| | - Boon Han Teo
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, UK
| | - Sarah E Smith
- Kymab Ltd, The Bennet Building (B930), Babraham Research Campus, Cambridge, UK
| | - Paul Kellam
- Department of Infectious Disease, Imperial College Faculty of Medicine, Wright Fleming Institute, St Mary's Campus, London, UK
- Kymab Ltd, The Bennet Building (B930), Babraham Research Campus, Cambridge, UK
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, School of Life and Environmental Sciences and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Mark Marsh
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
56
|
Aiello A, Giannessi F, Percario ZA, Affabris E. An emerging interplay between extracellular vesicles and cytokines. Cytokine Growth Factor Rev 2019; 51:49-60. [PMID: 31874738 DOI: 10.1016/j.cytogfr.2019.12.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are small membrane-bound particles that are naturally released from cells. They are recognized as potent vehicles of intercellular communication both in prokaryotes and eukaryotes. Because of their capacity to carry biological macromolecules such as proteins, lipids and nucleic acids, EVs influence different physiological and pathological functions of both parental and recipient cells. Although multiple pathways have been proposed for cytokine secretion beyond the classical ER/Golgi route, EVs have recently recognized as an alternative secretory mechanism. Interestingly, cytokines/chemokines exploit these vesicles to be released into the extracellular milieu, and also appear to modulate their release, trafficking and/or content. In this review, we provide an overview of the cytokines/chemokines that are known to be associated with EVs or their regulation with a focus on TNFα, IL-1β and IFNs.
Collapse
|
57
|
Porcine IFITM1 is a host restriction factor that inhibits pseudorabies virus infection. Int J Biol Macromol 2019; 151:1181-1193. [PMID: 31743714 PMCID: PMC7102536 DOI: 10.1016/j.ijbiomac.2019.10.162] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/26/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022]
Abstract
Interferon-inducible transmembrane proteins (IFITMs) restrict infection by several viruses, such as influenza A virus, West Nile virus and dengue virus. It has not been determined whether porcine IFITMs (pIFITMs) inhibit infection by pseudorabies virus (PRV), an enveloped, double-stranded DNA virus, which is the etiological agent of Aujeszky's disease in pigs. Here, we report that PRV infection elicited pIFITM1 expression in PK15 porcine kidney epithelial cells and 3D4/21 alveolar macrophages. pIFITM2 and pIFITM3 expression was only elevated in PK15 cells during PRV infection. Depletion of pIFITM1 using RNA interference, either in PK15 or in 3D4/21 cells, enhanced PRV infection while overexpression of pIFITM1 had the opposite effect. Knockdown of pIFITM2 and pIFITM3 did not influence PRV infection, suggesting that pIFITM2 and pIFITM3 are independent of PRV infection. PRV-induced pIFITM1 expression was dependent on the cGAS/STING/TBK1/IRF3 innate immune pathway and interferon-alpha receptor-1, suggesting that pIFITM1 is up-regulated by the type I interferon signaling pathway. The anti-PRV role of pIFITM1 was inhibited upon PRV entry. Our data demonstrate that pIFITM1 is a host restriction factor that inhibits PRV entry that may shed light on a strategy for prevention of PRV infection.
Collapse
|
58
|
Interplay between Intrinsic and Innate Immunity during HIV Infection. Cells 2019; 8:cells8080922. [PMID: 31426525 PMCID: PMC6721663 DOI: 10.3390/cells8080922] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023] Open
Abstract
Restriction factors are antiviral components of intrinsic immunity which constitute a first line of defense by blocking different steps of the human immunodeficiency virus (HIV) replication cycle. In immune cells, HIV infection is also sensed by several pattern recognition receptors (PRRs), leading to type I interferon (IFN-I) and inflammatory cytokines production that upregulate antiviral interferon-stimulated genes (ISGs). Several studies suggest a link between these two types of immunity. Indeed, restriction factors, that are generally interferon-inducible, are able to modulate immune responses. This review highlights recent knowledge of the interplay between restriction factors and immunity inducing antiviral defenses. Counteraction of this intrinsic and innate immunity by HIV viral proteins will also be discussed.
Collapse
|
59
|
Gan CP, Sam KK, Yee PS, Zainal NS, Lee BKB, Abdul Rahman ZA, Patel V, Tan AC, Zain RB, Cheong SC. IFITM3 knockdown reduces the expression of CCND1 and CDK4 and suppresses the growth of oral squamous cell carcinoma cells. Cell Oncol (Dordr) 2019; 42:477-490. [PMID: 30949979 PMCID: PMC7771307 DOI: 10.1007/s13402-019-00437-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2019] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Oral squamous cell carcinoma (OSCC) is a challenging disease to treat. Up to 50% of OSCC patients with advanced disease develop recurrences. Elucidation of key molecular mechanisms underlying OSCC development may provide opportunities to target specific genes and, thus, to improve patient survival. In this study, we examined the expression and functional role of interferon transmembrane protein 3 (IFITM3) in OSCC development. METHODS The expression of IFITM3 in OSCC and normal oral mucosal tissues was assessed by qRT-PCR and immunohistochemistry. The role of IFITM3 in driving OSCC cell proliferation and survival was examined using siRNA-mediated gene knockdown, and the role of IFITM3 in driving cell cycle regulators was examined using Western blotting. RESULTS We found that IFITM3 is overexpressed in more than 79% of primary OSCCs. We also found that IFITM3 knockdown led to impaired OSCC cell growth through inhibition of cell proliferation, induction of cell cycle arrest, senescence and apoptosis. In addition, we found that IFITM3 knockdown led to reduced expressions of CCND1 and CDK4 and reduced RB phosphorylation, leading to inhibition of OSCC cell growth. This information may be instrumental for the design of novel targeted therapeutic strategies. CONCLUSIONS From our data we conclude that IFITM3 is overexpressed in OSCC and may regulate the CCND1-CDK4/6-pRB axis to mediate OSCC cell growth.
Collapse
Affiliation(s)
- Chai Phei Gan
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 2nd Floor, Outpatient Centre, Subang Jaya Medical Centre, 47500, Subang Jaya, Selangor, Malaysia
| | - Kin Kit Sam
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 2nd Floor, Outpatient Centre, Subang Jaya Medical Centre, 47500, Subang Jaya, Selangor, Malaysia
| | - Pei San Yee
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 2nd Floor, Outpatient Centre, Subang Jaya Medical Centre, 47500, Subang Jaya, Selangor, Malaysia
| | - Nur Syafinaz Zainal
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 2nd Floor, Outpatient Centre, Subang Jaya Medical Centre, 47500, Subang Jaya, Selangor, Malaysia
| | - Bernard Kok Bang Lee
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 2nd Floor, Outpatient Centre, Subang Jaya Medical Centre, 47500, Subang Jaya, Selangor, Malaysia
| | - Zainal Ariff Abdul Rahman
- Department of Oral & Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Vyomesh Patel
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 2nd Floor, Outpatient Centre, Subang Jaya Medical Centre, 47500, Subang Jaya, Selangor, Malaysia
| | - Aik Choon Tan
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rosnah Binti Zain
- Oral Cancer Research & Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Sok Ching Cheong
- Head and Neck Cancer Research Team, Cancer Research Malaysia, 2nd Floor, Outpatient Centre, Subang Jaya Medical Centre, 47500, Subang Jaya, Selangor, Malaysia.
- Department of Oral & Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
60
|
Liao Y, Goraya MU, Yuan X, Zhang B, Chiu SH, Chen JL. Functional Involvement of Interferon-Inducible Transmembrane Proteins in Antiviral Immunity. Front Microbiol 2019; 10:1097. [PMID: 31156602 PMCID: PMC6532022 DOI: 10.3389/fmicb.2019.01097] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/30/2019] [Indexed: 01/03/2023] Open
Abstract
Interferons (IFNs) play crucial roles in host defense against viral infections by inducing the expression of numerous IFN-stimulated genes (ISGs) that can activate host antiviral immunity. Interferon-inducible transmembrane proteins (IFITMs), a family of small transmembrane proteins, are critical ISG products. Compelling evidence has implicated that IFITMs can establish an innate immune state to eliminate pathogens efficiently. IFITM proteins can impede broad-spectrum viral infection through various mechanisms. It is generally believed that IFITMs can block the viral entry by suppressing viral membrane fusion. However, some findings indicated that IFITMs might also inhibit viral gene expression and viral protein synthesis and thereby impair viral replication. IFITMs may incorporate into virions during viral assembly and thus reduce the infectivity of nascent virions. The precise inhibitory mechanism of IFITMs on viral infection and replication still requires further exploration. In this review, we highlight the recent findings regarding critical roles of IFITMs in host-virus interaction. We also discuss the molecular mechanisms underlying their functions in antiviral responses.
Collapse
Affiliation(s)
- Yuan Liao
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mohsan Ullah Goraya
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xu Yuan
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Baoge Zhang
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shih-Hsin Chiu
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
61
|
Yan J, Jiang Y, Lu J, Wu J, Zhang M. Inhibiting of Proliferation, Migration, and Invasion in Lung Cancer Induced by Silencing Interferon-Induced Transmembrane Protein 1 (IFITM1). BIOMED RESEARCH INTERNATIONAL 2019; 2019:9085435. [PMID: 31205947 PMCID: PMC6530206 DOI: 10.1155/2019/9085435] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/27/2019] [Accepted: 04/18/2019] [Indexed: 01/09/2023]
Abstract
Interferon-induced transmembrane protein 1 (IFITM1), a 17-kDa membrane protein, is generally known as a modulator in many cellular functions. Recent studies showed overexpression of IFITM1 in cancers and relationship between IFITM1 overexpression and tumor progression. However, the role of IFITM1 in lung cancer remains unclear. Here, we presented the overexpression of IFITM1 in lung cancer tissues and cell lines A549 and H460 using quantitative Real-Time RT-PCR. In vitro assay indicated IFITM1 silencing inhibited lung cancer cell proliferation, migration, and invasion. Further, in vivo assay showed that IFITM1 silencing markedly suppressed cell growth and metastasis of lung cancer in tumor-bearing BALB/c nude mice. Mechanistically, we found that IFITM1 silencing significantly alleviated the protein levels of β-catenin, cyclin D1, and c-Mycin lung cancer cells and tumor samples. Taken together, our study revealed the role of IFITM1 as a tumor promoter during lung cancer development and the possible molecular mechanism.
Collapse
Affiliation(s)
- Jun Yan
- Department of Pathology, Tianjin First Center Hospital, Tianjin 300192, China
| | - Ying Jiang
- Department of Pathology, Tianjin First Center Hospital, Tianjin 300192, China
| | - Jianfeng Lu
- Department of Pathology, Tianjin First Center Hospital, Tianjin 300192, China
| | - Jianhui Wu
- Department of Pathology, Tianjin First Center Hospital, Tianjin 300192, China
| | - Mingfang Zhang
- Department of Pathology, Tianjin First Center Hospital, Tianjin 300192, China
| |
Collapse
|
62
|
D Urbano V, De Crignis E, Re MC. Host Restriction Factors and Human Immunodeficiency Virus (HIV-1): A Dynamic Interplay Involving All Phases of the Viral Life Cycle. Curr HIV Res 2019; 16:184-207. [PMID: 30117396 DOI: 10.2174/1570162x16666180817115830] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 02/08/2023]
Abstract
Mammalian cells have evolved several mechanisms to prevent or block lentiviral infection and spread. Among the innate immune mechanisms, the signaling cascade triggered by type I interferon (IFN) plays a pivotal role in limiting the burden of HIV-1. In the presence of IFN, human cells upregulate the expression of a number of genes, referred to as IFN-stimulated genes (ISGs), many of them acting as antiviral restriction factors (RFs). RFs are dominant proteins that target different essential steps of the viral cycle, thereby providing an early line of defense against the virus. The identification and characterization of RFs have provided unique insights into the molecular biology of HIV-1, further revealing the complex host-pathogen interplay that characterizes the infection. The presence of RFs drove viral evolution, forcing the virus to develop specific proteins to counteract their activity. The knowledge of the mechanisms that prevent viral infection and their viral counterparts may offer new insights to improve current antiviral strategies. This review provides an overview of the RFs targeting HIV-1 replication and the mechanisms that regulate their expression as well as their impact on viral replication and the clinical course of the disease.
Collapse
Affiliation(s)
- Vanessa D Urbano
- Retrovirus Laboratory, Operative Unit of Clinical Microbiology, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Elisa De Crignis
- Retrovirus Laboratory, Operative Unit of Clinical Microbiology, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Maria Carla Re
- Retrovirus Laboratory, Operative Unit of Clinical Microbiology, S. Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
63
|
Wellington D, Laurenson-Schafer H, Abdel-Haq A, Dong T. IFITM3: How genetics influence influenza infection demographically. Biomed J 2019; 42:19-26. [PMID: 30987701 PMCID: PMC6468115 DOI: 10.1016/j.bj.2019.01.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/06/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022] Open
Abstract
The role of host genetics in influenza infection is unclear despite decades of interest. Confounding factors such as age, sex, ethnicity and environmental factors have made it difficult to assess the role of genetics without influence. In recent years a single nucleotide polymorphism, interferon-induced transmembrane protein 3 (IFITM3) rs12252, has been shown to alter the severity of influenza infection in Asian populations. In this review we investigate this polymorphism as well as several others suggested to alter the host's defence against influenza infection. In addition, we highlight the open questions surrounding the viral restriction protein IFITM3 with the hope that by answering some of these questions we can elucidate the mechanism of IFITM3 viral restriction and therefore how this restriction is altered due to the rs12252 polymorphism.
Collapse
Affiliation(s)
- Dannielle Wellington
- MRC Human Immunology Unit, WIMM, University of Oxford, OX3 9DS, UK; CAMS Oxford Institute, Nuffield Department of Medicine, Oxford University, OX3 9FZ, UK.
| | - Henry Laurenson-Schafer
- MRC Human Immunology Unit, WIMM, University of Oxford, OX3 9DS, UK; CAMS Oxford Institute, Nuffield Department of Medicine, Oxford University, OX3 9FZ, UK
| | - Adi Abdel-Haq
- MRC Human Immunology Unit, WIMM, University of Oxford, OX3 9DS, UK; Martin-Luther-University, Halle-Wittenberg, Germany
| | - Tao Dong
- MRC Human Immunology Unit, WIMM, University of Oxford, OX3 9DS, UK; CAMS Oxford Institute, Nuffield Department of Medicine, Oxford University, OX3 9FZ, UK.
| |
Collapse
|
64
|
Zhao X, Li J, Winkler CA, An P, Guo JT. IFITM Genes, Variants, and Their Roles in the Control and Pathogenesis of Viral Infections. Front Microbiol 2019; 9:3228. [PMID: 30687247 PMCID: PMC6338058 DOI: 10.3389/fmicb.2018.03228] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/12/2018] [Indexed: 01/01/2023] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs) are a family of small proteins that localize in the plasma and endolysosomal membranes. IFITMs not only inhibit viral entry into host cells by interrupting the membrane fusion between viral envelope and cellular membranes, but also reduce the production of infectious virions or infectivity of progeny virions. Not surprisingly, some viruses can evade the restriction of IFITMs and even hijack the antiviral proteins to facilitate their infectious entry into host cells or promote the assembly of virions, presumably by modulating membrane fusion. Similar to many other host defense genes that evolve under the selective pressure of microorganism infection, IFITM genes evolved in an accelerated speed in vertebrates and many single-nucleotide polymorphisms (SNPs) have been identified in the human population, some of which have been associated with severity and prognosis of viral infection (e.g., influenza A virus). Here, we review the function and potential impact of genetic variation for IFITM restriction of viral infections. Continuing research efforts are required to decipher the molecular mechanism underlying the complicated interaction among IFITMs and viruses in an effort to determine their pathobiological roles in the context of viral infections in vivo.
Collapse
Affiliation(s)
- Xuesen Zhao
- Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Disease, Beijing, China
| | - Jiarui Li
- Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Disease, Beijing, China
| | - Cheryl A Winkler
- Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Ping An
- Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Ju-Tao Guo
- Baruch S. Blumberg Institute, Hepatitis B Foundation, Doylestown, PA, United States
| |
Collapse
|
65
|
Xie S, Luo H, Zhang H, Zhu H, Zuo X, Liu S. Discovery of Key Genes in Dermatomyositis Based on the Gene Expression Omnibus Database. DNA Cell Biol 2018; 37:982-992. [PMID: 30383435 DOI: 10.1089/dna.2018.4256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The aim of this study was to identify biomarkers of dermatomyositis (DM). The analysis was conducted by retrieving DM-related cDNA microarray data sets from public databases. Gene ontology, Kyoto encyclopedia of genes and genomes, and protein-protein interaction analyses were performed, together with quantitative PCR-based detection of biomarkers in muscle tissue after stimulation with serum from patients with DM or healthy controls. Our analysis of five microarray data sets identified 20 common differentially expressed genes that are closely associated with DM. PCR analysis showed that mRNAs of IFITM2, LY6E, DDX58, and IFI6 were expressed at significantly higher levels in the muscle tissue of patients with DM than in normal muscle tissues. These mRNAs were also upregulated in human skeletal muscle cells stimulated with the serum from patients with DM. The results of integrated analyses of the DM microarray data and the mRNA levels of genes showed significant differences between the muscle tissues of DM patients and controls, which could indicate key pathogenic genes and novel therapeutic targets for DM.
Collapse
Affiliation(s)
- Shuoshan Xie
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Hui Luo
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Huali Zhang
- 2 Department of Pathophysiology, Xiangya School of Medicine, Central South University , Changsha, China
| | - Honglin Zhu
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Xiaoxia Zuo
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Sijia Liu
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| |
Collapse
|
66
|
Abstract
Rapamycin and its derivatives are specific inhibitors of mammalian target of rapamycin (mTOR) kinase and, as a result, are well-established immunosuppressants and antitumorigenic agents. Additionally, this class of drug promotes gene delivery by facilitating lentiviral vector entry into cells, revealing its potential to improve gene therapy efforts. However, the precise mechanism was unknown. Here, we report that mTOR inhibitor treatment results in down-regulation of the IFN-induced transmembrane (IFITM) proteins. IFITM proteins, especially IFITM3, are potent inhibitors of virus-cell fusion and are broadly active against a range of pathogenic viruses. We found that the effect of rapamycin treatment on lentiviral transduction is diminished upon IFITM silencing or knockout in primary and transformed cells, and the extent of transduction enhancement depends on basal expression of IFITM proteins, with a major contribution from IFITM3. The effect of rapamycin treatment on IFITM3 manifests at the level of protein, but not mRNA, and is selective, as many other endosome-associated transmembrane proteins are unaffected. Rapamycin-mediated degradation of IFITM3 requires endosomal trafficking, ubiquitination, endosomal sorting complex required for transport (ESCRT) machinery, and lysosomal acidification. Since IFITM proteins exhibit broad antiviral activity, we show that mTOR inhibition also promotes infection by another IFITM-sensitive virus, Influenza A virus, but not infection by Sendai virus, which is IFITM-resistant. Our results identify the molecular basis by which mTOR inhibitors enhance virus entry into cells and reveal a previously unrecognized immunosuppressive feature of these clinically important drugs. In addition, this study uncovers a functional convergence between the mTOR pathway and IFITM proteins at endolysosomal membranes.
Collapse
|
67
|
Evolution-Guided Structural and Functional Analyses of the HERC Family Reveal an Ancient Marine Origin and Determinants of Antiviral Activity. J Virol 2018; 92:JVI.00528-18. [PMID: 29669830 PMCID: PMC6002735 DOI: 10.1128/jvi.00528-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/10/2018] [Indexed: 01/24/2023] Open
Abstract
In humans, homologous to the E6-AP carboxyl terminus (HECT) and regulator of chromosome condensation 1 (RCC1)-like domain-containing protein 5 (HERC5) is an interferon-induced protein that inhibits replication of evolutionarily diverse viruses, including human immunodeficiency virus type 1 (HIV-1). To better understand the origin, evolution, and function of HERC5, we performed phylogenetic, structural, and functional analyses of the entire human small-HERC family, which includes HERC3, HERC4, HERC5, and HERC6. We demonstrated that the HERC family emerged >595 million years ago and has undergone gene duplication and gene loss events throughout its evolution. The structural topology of the RCC1-like domain and HECT domains from all HERC paralogs is highly conserved among evolutionarily diverse vertebrates despite low sequence homology. Functional analyses showed that the human small HERCs exhibit different degrees of antiviral activity toward HIV-1 and that HERC5 provides the strongest inhibition. Notably, coelacanth HERC5 inhibited simian immunodeficiency virus (SIV), but not HIV-1, particle production, suggesting that the antiviral activity of HERC5 emerged over 413 million years ago and exhibits species- and virus-specific restriction. In addition, we showed that both HERC5 and HERC6 are evolving under strong positive selection, particularly blade 1 of the RCC1-like domain, which we showed is a key determinant of antiviral activity. These studies provide insight into the origin, evolution, and biological importance of the human restriction factor HERC5 and the other HERC family members. IMPORTANCE Intrinsic immunity plays an important role as the first line of defense against viruses. Studying the origins, evolution, and functions of proteins responsible for effecting this defense will provide key information about virus-host relationships that can be exploited for future drug development. We showed that HERC5 is one such antiviral protein that belongs to an evolutionarily conserved family of HERCs with an ancient marine origin. Not all vertebrates possess all HERC members, suggesting that different HERCs emerged at different times during evolution to provide the host with a survival advantage. Consistent with this, two of the more recently emerged HERC members, HERC5 and HERC6, displayed strong signatures of having been involved in an ancient evolutionary battle with viruses. Our findings provide new insights into the evolutionary origin and function of the HERC family in vertebrate evolution, identifying HERC5 and possibly HERC6 as important effectors of intrinsic immunity in vertebrates.
Collapse
|
68
|
Wang Q, Yuan X, Li C, Deng H, Wang C. Distribution of Ifitm3 in Yellow-Feathered Broilers and Inhibition of Avian Reovirus Multiplication by Ifitm3. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2018. [DOI: 10.1590/1806-9061-2017-0662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Q Wang
- Fujian agriculture and forestry university, China; Fujian Agriculture and Forestry University, China
| | - X Yuan
- Fujian agriculture and forestry university, China
| | - C Li
- Fujian agriculture and forestry university, China
| | - H Deng
- Fujian agriculture and forestry university, China
| | - C Wang
- Fujian agriculture and forestry university, China
| |
Collapse
|
69
|
Stankovic ND, Hoppmann N, Teodorczyk M, Kim EL, Bros M, Giese A, Zipp F, Schmidt MHH. No role of IFITM3 in brain tumor formation in vivo. Oncotarget 2018; 7:86388-86405. [PMID: 27835870 PMCID: PMC5349921 DOI: 10.18632/oncotarget.13199] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 10/29/2016] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal solid tumors in adults. Despite aggressive treatment approaches for patients, GBM recurrence is inevitable, in part due to the existence of stem-like brain tumor-propagating cells (BTPCs), which produce factors rendering them resistant to radio- and chemotherapy. Comparative transcriptome analysis of irradiated, patient-derived BTPCs revealed a significant upregulation of the interferon-inducible transmembrane protein 3 (IFITM3), suggesting the protein as a factor mediating radio resistance. Previously, IFITM3 has been described to affect glioma cells; therefore, the role of IFITM3 in the formation and progression of brain tumors has been investigated in vivo. Intracranial implantation studies using radio-selected BTPCs alongside non-irradiated parental BTPCs in immunodeficient mice displayed no influence of irradiation on animal survival. Furthermore, gain and loss of function studies using BTPCs ectopically expressing IFITM3 or having IFITM3 down-modulated by a shRNA approach, did affect neither tumor growth nor animal survival. Additionally, a syngeneic model based on the mouse glioma cell line GL261 was applied in order to consider the possibility that IFITM3 relies on an intact immune system to unfold its tumorigenic potential. GL261 cells ectopically expressing IFITM3 were implanted into the striatum of immunocompetent mice without influencing the survival of glioma-bearing animals. Lastly, the vasculature and the extent of microglia/macrophage invasion into the tumor were studied in BTPC and GL261 tumors but neither parameter was altered by IFITM3. This report presents for the first time that IFITM3 is upregulated in patient-derived BTPCs upon irradiation but does not affect brain tumor formation or progression in vivo.
Collapse
Affiliation(s)
- Nevenka Dudvarski Stankovic
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), Johannes Gutenberg University, School of Medicine, Mainz, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicola Hoppmann
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Research Center for Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), Johannes Gutenberg University, School of Medicine, Mainz, Germany
| | - Marcin Teodorczyk
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), Johannes Gutenberg University, School of Medicine, Mainz, Germany
| | - Ella L Kim
- Translational Oncology Research Group, Department of Neurosurgery, Johannes Gutenberg University, School of Medicine, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, Johannes Gutenberg University, School of Medicine, Mainz, Germany
| | - Alf Giese
- Translational Oncology Research Group, Department of Neurosurgery, Johannes Gutenberg University, School of Medicine, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Research Center for Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), Johannes Gutenberg University, School of Medicine, Mainz, Germany
| | - Mirko H H Schmidt
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), Johannes Gutenberg University, School of Medicine, Mainz, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
70
|
Alteber Z, Sharbi-Yunger A, Pevsner-Fischer M, Blat D, Roitman L, Tzehoval E, Elinav E, Eisenbach L. The anti-inflammatory IFITM genes ameliorate colitis and partially protect from tumorigenesis by changing immunity and microbiota. Immunol Cell Biol 2018; 96:284-297. [PMID: 29356071 DOI: 10.1111/imcb.12000] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022]
Abstract
Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.
Collapse
Affiliation(s)
- Zoya Alteber
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Adi Sharbi-Yunger
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | - Dan Blat
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lior Roitman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Esther Tzehoval
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Eran Elinav
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Lea Eisenbach
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
71
|
Shan J, Zhao B, Shan Z, Nie J, Deng R, Xiong R, Tsun A, Pan W, Zhao H, Chen L, Jin Y, Qian Z, Lui K, Liang R, Li D, Sun B, Lavillette D, Xu K, Li B. Histone demethylase LSD1 restricts influenza A virus infection by erasing IFITM3-K88 monomethylation. PLoS Pathog 2017; 13:e1006773. [PMID: 29281729 PMCID: PMC5760097 DOI: 10.1371/journal.ppat.1006773] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 01/09/2018] [Accepted: 11/27/2017] [Indexed: 11/30/2022] Open
Abstract
The histone demethylase LSD1 has been known as a key transcriptional coactivator for DNA viruses such as herpes virus. Inhibition of LSD1 was found to block viral genome transcription and lytic replication of DNA viruses. However, RNA virus genomes do not rely on chromatin structure and histone association, and the role of demethylase activity of LSD1 in RNA virus infections is not anticipated. Here, we identify that, contrary to its role in enhancing DNA virus replication, LSD1 limits RNA virus replication by demethylating and activating IFITM3 which is a host restriction factor for many RNA viruses. We have found that LSD1 is recruited to demethylate IFITM3 at position K88 under IFNα treatment. However, infection by either Vesicular Stomatitis Virus (VSV) or Influenza A Virus (IAV) triggers methylation of IFITM3 by promoting its disassociation from LSD1. Accordingly, inhibition of the enzymatic activity of LSD1 by Trans-2-phenylcyclopropylamine hydrochloride (TCP) increases IFITM3 monomethylation which leads to more severe disease outcomes in IAV-infected mice. In summary, our findings highlight the opposite role of LSD1 in fighting RNA viruses comparing to DNA viruses infection. Our data suggest that the demethylation of IFITM3 by LSD1 is beneficial for the host to fight against RNA virus infection. The viral genomes of DNA viruses but not RNA viruses form chromatin structure during infection. Thus, epigenetic modulators are not expected to have crucial roles in RNA viral infection. However, here, we identify for the first time, that, opposite to its role in enhancing DNA virus replication, LSD1, a histone demethylase, limits RNA virus replication. We show that, under IFNα treatment, LSD1 is involved in the demethylation of IFITM3, a well-known host restriction factor for many RNA viruses. To counteract IFITM3 activation by demethylation, several RNA viruses, such as VSV and IAV, but not Zika virus, have developed strategy to inactive IFITM3 by promoting its dissociation from LSD1. In agreement with our findings, the inhibition of the enzymatic activity of LSD1 by small molecule leads to more severe disease outcomes in IAV-infected mice. Our data suggest that although LSD1 inhibitor is beneficial for treating DNA virus infection, it could be harmful to the host suffering from RNA virus infection. On the contrary, developing strategies to stimulate LSD1 activity to demethylate of IFITM3 is essential to fight RNA viruses.
Collapse
Affiliation(s)
- Jiaoyu Shan
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Human Parasitology Department of Basic Medicine College, Xinjiang Medical University, Urumqi, China
| | - Binbin Zhao
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Zhao Shan
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia Nie
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rong Deng
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Xiong
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of interspecies transmission of arboviruses and therapeutics research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Andy Tsun
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weiqi Pan
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hanzhi Zhao
- CAS Center for Excellence in Molecular Cell Science, Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ling Chen
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ying Jin
- CAS Center for Excellence in Molecular Cell Science, Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhikang Qian
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of Herpesvirus and Molecular Virology Research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kawing Lui
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Liang
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Bing Sun
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Virology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dimitri Lavillette
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of interspecies transmission of arboviruses and therapeutics research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ke Xu
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of interspecies transmission of arboviruses and therapeutics research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (BL); (KX)
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
- * E-mail: (BL); (KX)
| |
Collapse
|
72
|
Shi G, Schwartz O, Compton AA. More than meets the I: the diverse antiviral and cellular functions of interferon-induced transmembrane proteins. Retrovirology 2017; 14:53. [PMID: 29162141 PMCID: PMC5697417 DOI: 10.1186/s12977-017-0377-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/13/2017] [Indexed: 01/14/2023] Open
Abstract
The first responders of human antiviral immunity are components of the intrinsic immune response that reside within each and every one of our cells. This cell-autonomous arsenal consists of nucleic acid sensors and antiviral effectors strategically placed by evolution to detect and restrict invading viruses. While some factors are present at baseline to allow for constant surveillance of the cell interior, others are upregulated by cytokines (such as interferons) that signal a viral infection underway in neighboring cells. In this review, we highlight the multiple roles played by the interferon-induced transmembrane (IFITM) proteins during viral infection, with focuses on IFITM3 and HIV-1. Moreover, we discuss the cellular pathways in which IFITM proteins are intertwined and the various functions they have been ascribed outside the context of infection. While appreciated as broadly-acting, potent restriction factors that prevent virus infection and pathogenesis in cell culture and in vivo, questions remain regarding their precise mode of action and importance in certain viral contexts. Continued efforts to study IFITM protein function will further cement their status as critical host determinants of virus susceptibility and prioritize them in the development of new antiviral therapies.
Collapse
Affiliation(s)
- Guoli Shi
- Antiviral Immunity and Resistance Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Paris, France.,UMR CNRS 3569, Paris, France
| | - Alex A Compton
- Antiviral Immunity and Resistance Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
73
|
Randolph AG, Yip WK, Allen EK, Rosenberger CM, Agan AA, Ash SA, Zhang Y, Bhangale TR, Finkelstein D, Cvijanovich NZ, Mourani PM, Hall MW, Su HC, Thomas PG. Evaluation of IFITM3 rs12252 Association With Severe Pediatric Influenza Infection. J Infect Dis 2017; 216:14-21. [PMID: 28531322 DOI: 10.1093/infdis/jix242] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/15/2017] [Indexed: 02/03/2023] Open
Abstract
Background Interferon-induced transmembrane protein 3 (IFITM3) restricts endocytic fusion of influenza virus. IFITM3 rs12252_C, a putative alternate splice site, has been associated with influenza severity in adults. IFITM3 has not been evaluated in pediatric influenza. Methods The Pediatric Influenza (PICFLU) study enrolled children with suspected influenza infection across 38 pediatric intensive care units during November 2008 to April 2016. IFITM3 was sequenced in patients and parents were genotyped for specific variants for family-based association testing. rs12252 was genotyped in 54 African-American pediatric outpatients with influenza (FLU09), included in the population-based comparisons with 1000 genomes. Splice site analysis of rs12252_C was performed using PICFLU and FLU09 patient RNA. Results In PICFLU, 358 children had influenza infection. We identified 22 rs12252_C homozygotes in 185 white non-Hispanic children. rs12252_C was not associated with influenza infection in population or family-based analyses. We did not identify the Δ21 IFITM3 isoform in RNAseq data. The rs12252 genotype was not associated with IFITM3 expression levels, nor with critical illness severity. No novel rare IFITM3 functional variants were identified. Conclusions rs12252 was not associated with susceptibility to influenza-related critical illness in children or with critical illness severity. Our data also do not support it being a splice site.
Collapse
Affiliation(s)
- Adrienne G Randolph
- Department of Anesthesia, Perioperative and Pain Medicine, Boston Children's Hospital
- Departments of Anaesthesia and Pediatrics, Harvard Medical School, Boston
| | - Wai-Ki Yip
- Foundation Medicine Inc, Cambridge, Massachusetts
| | - Emma Kaitlynn Allen
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Anna A Agan
- Department of Anesthesia, Perioperative and Pain Medicine, Boston Children's Hospital
| | - Stephanie A Ash
- Department of Anesthesia, Perioperative and Pain Medicine, Boston Children's Hospital
| | - Yu Zhang
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - David Finkelstein
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Peter M Mourani
- Section of Critical Care, Department of Pediatrics, University of Colorado School of Medicine and Research Institute, Children's Hospital Colorado, Aurora
| | - Mark W Hall
- Critical Care Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Helen C Su
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
74
|
The osteogenic cell surface marker BRIL/IFITM5 is dispensable for bone development and homeostasis in mice. PLoS One 2017; 12:e0184568. [PMID: 28880886 PMCID: PMC5589259 DOI: 10.1371/journal.pone.0184568] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/26/2017] [Indexed: 02/07/2023] Open
Abstract
BRIL (bone-restricted IFITM-like), is a short transmembrane protein expressed almost exclusively in osteoblasts. Although much is known about its bone-restricted gene expression pattern and protein biochemical and topological features, little information is available for BRIL physiological function. Two autosomal dominant forms of osteogenesis imperfecta (OI) are caused by distinct, but recurrent mutations in the BRIL gene. Yet, the underlying mechanisms by which those mutations lead to OI are still poorly understood. A previous report indicated that BRIL knockout (KO) mice had bone deformities, shortened long bones, and reproductive problems. Here we generated and systematically analyzed the skeletal phenotype of a new global Bril KO/LacZ knockin mouse model. KO mice reproduced and thrived normally up to 12 month of age. The skeletal phenotype of KO and WT littermates was assessed at embryonic (E13.5 to E18.5) and postnatal (2 days, 3 weeks, 3 months and 8 months) time-points. Embryos from E13.5 through to E18.5 showed significant X-Gal staining in all skeletal elements without any apparent patterning anomalies. Although bone deformities were never observed at any postnatal ages, minor and transient differences were noted in terms of bone length and static uCT parameters, but not systematically across all ages and genders. These changes, however, were not accompanied by significant alteration in bone material properties as assessed by a 3-point bending test. In addition, no changes were detected in circulating serum markers of bone turnover (P1NP, CTX-I, and osteocalcin). Gene expression monitoring also revealed no major impact of the loss of BRIL. Further, when mice were challenged with a surgically-induced fracture in tibia, bones repaired equally well in the KO mice as compared to WT. Finally, we showed that BRIL C-terminus is not a bona fide binding site for calcium. In conclusion, our in depth analysis suggest that skeletal patterning, bone mass accrual and remodeling in mice proceeded independent of BRIL.
Collapse
|
75
|
Li HP, Chen PG, Liu FT, Zhu HS, Jiao XQ, Zhong K, Guo YJ, Zha GM, Han LQ, Lu WF, Wang YY, Yang GY. Characterization and anti-inflammation role of swine IFITM3 gene. Oncotarget 2017; 8:73579-73589. [PMID: 29088728 PMCID: PMC5650283 DOI: 10.18632/oncotarget.20568] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/09/2017] [Indexed: 12/30/2022] Open
Abstract
IFITM3 is involved in cell adhesion, apoptosis, immune, and antivirus activity. Furthermore, IFITM3 gene has been considered as a preferential marker for inflammatory diseases, and positive correlation to pathological grades. Therefore, we assumed that IFITM3 was regulated by different signal pathways. To better understand IFITM3 function in inflammatory response, we cloned swine IFITM3 gene, and detected IFITM3 distribution in tissues, as well as characterized this gene. Results indicated that the length of swine IFITM3 gene was 438 bp, encoding 145 amino acids. IFITM3 gene expression abundance was higher in spleen and lungs. Moreover, we next constructed the eukaryotic expression vector PBIFM3 and transfected into PK15 cells, finally obtained swine IFITM3 gene stable expression cell line. Meanwhile, we explored the effects of LPS on swine IFITM3 expression. Results showed that LPS increased IFITM3 mRNA abundance and exhibited time-dependent effect for LPS treatment. To further demonstrate the mechanism that IFITM3 regulated type I IFNs production, we also detected the important molecules expression of TLR4 signaling pathway. In transfected and non-transfected IFITM3 PK15 cells, LPS exacerbated the relative expression of TLR4-NFκB signaling molecules. However, the IFITM3 overexpression suppressed the inflammatory development of PK15 cells. In conclusion, these data indicated that the overexpression of swine IFITM3 could decrease the inflammatory response through TLR4 signaling pathway, and participate in type I interferon production. These findings may lead to an improved understanding of the biological function of IFITM3 in inflammation.
Collapse
Affiliation(s)
- He-Ping Li
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Pei-Ge Chen
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Fu-Tao Liu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - He-Shui Zhu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Xian-Qin Jiao
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Kai Zhong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yu-Jie Guo
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Guang-Ming Zha
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Li-Qiang Han
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Wei-Fei Lu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yue-Ying Wang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou, Henan, China
| |
Collapse
|
76
|
Zhang D, Wang H, He H, Niu H, Li Y. Interferon induced transmembrane protein 3 regulates the growth and invasion of human lung adenocarcinoma. Thorac Cancer 2017; 8:337-343. [PMID: 28544512 PMCID: PMC5494463 DOI: 10.1111/1759-7714.12451] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/30/2017] [Accepted: 04/05/2017] [Indexed: 01/16/2023] Open
Abstract
Background Interferon induced transmembrane protein 3 (IFITM3) plays an important role in the tumorigenesis and progression of multiple cancers. This study investigated the expression and function of IFITM3 in human lung adenocarcinoma. Methods Fifty human lung adenocarcinoma tissues were collected. IFITM3 expression was assessed by immunohistochemical staining. The clinicopathologic characteristics of all patients were analyzed. Results IFITM3 was mainly detected in the cytoplasm of advanced cancer tissues and its expression was correlated with tumor malignancy grade. Knockdown of IFITM3 in vitro markedly inhibited the proliferation and invasion of lung adenocarcinoma cells. Conclusion IFITM3 represents a potential therapeutic target for the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, China.,Department of Pulmonary Medicine, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Huimin Wang
- Department of Pulmonary Medicine, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Huijie He
- Department of Pulmonary Medicine, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Haiying Niu
- Department of Pulmonary Medicine, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Yu Li
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
77
|
Identification of IFITM1 and IFITM3 in Goose: Gene Structure, Expression Patterns, and Immune Reponses against Tembusu Virus Infection. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5149062. [PMID: 28386554 PMCID: PMC5366768 DOI: 10.1155/2017/5149062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/27/2016] [Accepted: 02/07/2017] [Indexed: 11/17/2022]
Abstract
As interferon-stimulated genes (ISGs), interferon-inducible transmembrane proteins 1 and 3 (IFITM1 and IFITM3) can effectively inhibit the replication of multiple viruses. Here, goose IFITM1 and IFITM3 were cloned and identified for the first time. The two proteins share the same topological structure and several important sites critical for the antiviral functions in other species are conserved in the goose. Goose IFITM1 and IFITM3 are most closely related to their respective orthologs in ducks; these proteins exhibited high mRNA transcript levels in immune-related tissues, including the thymus, bursa of Fabricius, and Harderian gland, compared to other tissues. Moreover, goose IFITM1 was highly constitutively expressed in gastrointestinal tract tissues, while goose IFITM3 was expressed in respiratory organs. Furthermore, goose IFITM3 was activated in goose peripheral blood mononuclear cells (PBMCs) infected with Tembusu virus (TMUV) or treated with Toll-like receptors (TLRs) agonists, while only the R848 and Poly (I:C) agonists induced significant upregulation of goose IFITM1. Furthermore, goose IFITM1 and IFITM3 were upregulated in the sampled tissues, to some extent, after TMUV infection. Notably, significant upregulation of goose IFITM1 and IFITM3 was detected in the cecum and cecal tonsil, where TMUV was primarily distributed. These data provide new insights into the immune effectors in geese and promote our understanding of the role of IFITM1 and IFITM3 in the defense against TMUV.
Collapse
|
78
|
Secreted Interferon-Inducible Factors Restrict Hepatitis B and C Virus Entry In Vitro. J Immunol Res 2017; 2017:4828936. [PMID: 28367455 PMCID: PMC5358466 DOI: 10.1155/2017/4828936] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/24/2017] [Accepted: 02/06/2017] [Indexed: 02/06/2023] Open
Abstract
Interferon-α (IFN-α) has been used for more than 20 years as the first-line therapy for hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, because it has a number of antiviral effects. In this study, we describe a novel mode of its antiviral action. We demonstrate that the supernatant from IFN-α-treated cultured cells restricted HBV and HCV infection by inhibiting viral entry into hepatoma cells. The factors contained in the supernatant competed with the virus for binding to heparan glycosaminoglycans—the nonspecific attachment step shared by HBV and HCV. Secreted factors of high molecular mass that bind to heparin columns elicited the antiviral effect. In conclusion, IFN-α is able to induce soluble factors that can bind to heparan glycosaminoglycans thus leading to the inhibition of viral binding.
Collapse
|
79
|
IGF1/IGF1R/STAT3 signaling-inducible IFITM2 promotes gastric cancer growth and metastasis. Cancer Lett 2017; 393:76-85. [PMID: 28223169 DOI: 10.1016/j.canlet.2017.02.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/20/2017] [Accepted: 02/11/2017] [Indexed: 01/06/2023]
Abstract
Interferon-induced transmembrane proteins (IFITMs) are expressed in some types of cancer. However, their precise roles in tumor progression remain unclear. The present study investigated the function of IFITM2 in gastric cancer (GC) progression. A retrospective analysis of a public database and 167 GC patients revealed that IFITM2 expression was upregulated in gastric tumor samples, which was positively correlated with disease progression, more frequent postoperative recurrence, and higher mortality rate. IFITM2 knockdown decreased GC cell proliferation, migration, invasion, and epithelial-to-mesenchymal transition in vitro. We also found that IFITM2 expression was in part induced by insulin-like growth factor (IGF) 1 via IGF1 receptor/signal transducer and activator of transcription 3 signaling. Furthermore, IFITM2 regulated interleukin-6 expression and secretion, which in turn increased IFITM2 expression. Silencing of IFITM2 expression suppressed tumor growth and lung metastasis in vivo. These results suggest that IFITM2 is a novel prognostic biomarker and regulator of GC progression.
Collapse
|
80
|
Goffinet C. Cellular Antiviral Factors that Target Particle Infectivity of HIV-1. Curr HIV Res 2016; 14:211-6. [PMID: 26674651 PMCID: PMC5403965 DOI: 10.2174/1570162x14666151216145521] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/30/2015] [Accepted: 12/14/2015] [Indexed: 11/22/2022]
Abstract
Background: In the past decade, the identification and characterization of antiviral genes with the ability to interfere with virus replication has established cell-intrinsic innate immunity as a third line of antiviral defense in addition to adaptive and classical innate immunity. Understanding how cellular factors have evolved to inhibit HIV-1 reveals particularly vulnerable points of the viral replication cycle. Many, but not all, antiviral proteins share type I interferon-upregulated expression and sensitivity to viral counteraction or evasion measures. Whereas well-established restriction factors interfere with early post-entry steps and release of HIV-1, recent research has revealed a diverse set of proteins that reduce the infectious quality of released particles using individual, to date poorly understood modes of action. These include induction of paucity of mature glycoproteins in nascent virions or self-incorporation into the virus particle, resulting in poor infectiousness of the virion and impaired spread of the infection. Conclusion: A better understanding of these newly discovered antiviral factors may open new avenues towards the design of drugs that repress the spread of viruses whose genomes have already integrated.
Collapse
Affiliation(s)
- Christine Goffinet
- Institute of Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany.
| |
Collapse
|
81
|
Liu BY, Lu YQ, Han F, Wang Y, Mo XK, Han JX. Effects of the overexpression of IFITM5 and IFITM5 c.-14C>T mutation on human osteosarcoma cells. Oncol Lett 2016; 13:111-118. [PMID: 28123530 PMCID: PMC5244967 DOI: 10.3892/ol.2016.5426] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/19/2016] [Indexed: 11/18/2022] Open
Abstract
The present study aimed to investigate the effects of overexpression of interferon-induced transmembrane protein 5 (IFITM5) and IFITM5 c.-14C>T mutation on osteogenic differentiation, and the proliferation, migration and invasion of SaOS2 cells. SaOS2 cells were transfected with plasmids containing wild type IFITM5 (W) or IFITM5 containing the c.-14C>T mutation (MU). The mRNA and protein expression levels of IFITM5 in SaOS2 cells were respectively detected by reverse transcription quantitative polymerase chain reaction and western blotting. The proliferative, migratory and invasive ability of SaOS2 cells was also examined. In addition, the expression levels of osteogenic differentiation markers alkaline phosphatase (ALP), osteocalcin (OCN) and runt-related transcription factor 2 (Runx2) were detected. Mineralized nodules were detected by Alizarin Red S staining and were quantified by measuring absorbance. The mRNA and protein expression levels of IFITM5 were high in cells transfected with IFITM5 and IFITM5 c.-14C>T mutation, and were higher in cells transfected with IFITM5 c.-14C>T mutation. There was no difference in proliferation between the control group (C) and the W and MU groups. However, overexpression of IFITM5 and IFITM5 c.-14C>T mutation increased apoptotic rate, decreased invasive capacity, increased the expression of ALP, OCN and Runx2, and increased the number of mineralized nodules following osteogenic induction. In addition, compared with C and W groups, cells transfected with IFITM5 c.-14C>T mutation exhibited decreased migratory ability. In conclusion, overexpression of IFITM5 and IFITM5 c.-14C>T mutation promotes tumor cell apoptosis, inhibits tumor invasion and promotes osteogenic differentiation. These findings may provide a theoretical basis for the development of a novel treatment method that targets IFITM5, and provides a platform for the potential treatment of human osteosarcoma.
Collapse
Affiliation(s)
- Bao-Yan Liu
- Shandong Medical Biotechnological Center, School of Medicine and Life Science, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250062, P.R. China
| | - Yan-Qin Lu
- Key Laboratory for Rare Disease Research of Shandong, Shandong Medical Biotechnological Center, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Feng Han
- Shandong Medical Biotechnological Center, School of Medicine and Life Science, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250062, P.R. China
| | - Yong Wang
- Department of Neurosurgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xin-Kai Mo
- Shandong Medical Biotechnological Center, School of Medicine and Life Science, Shandong Academy of Medical Sciences, University of Jinan, Jinan, Shandong 250062, P.R. China; Clinical Laboratory, Shandong Cancer Hospital and Institution, Jinan, Shandong 250117, P.R. China
| | - Jin-Xiang Han
- Key Laboratory for Rare Disease Research of Shandong, Shandong Medical Biotechnological Center, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| |
Collapse
|
82
|
Caba O, Irigoyen A, Jimenez-Luna C, Benavides M, Ortuño FM, Gallego J, Rojas I, Guillen-Ponce C, Torres C, Aranda E, Prados J. Identification of gene expression profiling associated with erlotinib-related skin toxicity in pancreatic adenocarcinoma patients. Toxicol Appl Pharmacol 2016; 311:113-116. [PMID: 27720938 DOI: 10.1016/j.taap.2016.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/21/2016] [Accepted: 10/05/2016] [Indexed: 01/26/2023]
|
83
|
Shen C, Li YJ, Yin QQ, Jiao WW, Li QJ, Xiao J, Sun L, Xu F, Li JQ, Qi H, Shen AD. Identification of differentially expressed transcripts targeted by the knockdown of endogenous IFITM3. Mol Med Rep 2016; 14:4367-4373. [PMID: 27667301 DOI: 10.3892/mmr.2016.5777] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/24/2016] [Indexed: 11/05/2022] Open
Abstract
Interferon inducible transmembrane protein 3 (IFITM3) is a double transmembrane protein. As a member of the IFITM family, IFITM3 can be upregulated by interferon (IFN) to be involved in various biological processes. In order to determine whether gene expression profiles can be altered by a lack of IFITM3, the present study used shRNAs lentivirus for knocking down the endogenous expression of IFITM3 in human HeLa cells and human whole genome microarrays to obtain gene expression profiles. A total of 1,011 downregulated transcripts and 615 upregulated transcripts were identified using the Agilent expression platform. The identified transcripts were involved in multiple pathways, including the complement pathways, and the antigen processing and presentation pathway. The present study identified the transcripts, which were affected by the downregulation of endogenous IFITM3 and the pathways they were involved in. These findings may lead to an improved understanding of the biological functions of IFITM3.
Collapse
Affiliation(s)
- Chen Shen
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Ying-Jia Li
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Qing-Qin Yin
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Wei-Wei Jiao
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Qin-Jing Li
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jing Xiao
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Lin Sun
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Fang Xu
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jie-Qiong Li
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Hui Qi
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - A-Dong Shen
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, National Clinical Research Center for Respiratory Diseases, Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| |
Collapse
|
84
|
Park HJ, Lee WY, Jeong HY, Kang HS, Kim JB, Song H. Mitochondrial interferon-induced transmembrane protein-1 is a critical regulator of cell death in MPRO cells. BIOTECHNOL BIOPROC E 2016. [DOI: 10.1007/s12257-016-0253-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
85
|
The Interferon-Stimulated Gene IFITM3 Restricts Infection and Pathogenesis of Arthritogenic and Encephalitic Alphaviruses. J Virol 2016; 90:8780-94. [PMID: 27440901 DOI: 10.1128/jvi.00655-16] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/17/2016] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Host cells respond to viral infections by producing type I interferon (IFN), which induces the expression of hundreds of interferon-stimulated genes (ISGs). Although ISGs mediate a protective state against many pathogens, the antiviral functions of the majority of these genes have not been identified. IFITM3 is a small transmembrane ISG that restricts a broad range of viruses, including orthomyxoviruses, flaviviruses, filoviruses, and coronaviruses. Here, we show that alphavirus infection is increased in Ifitm3(-/-) and Ifitm locus deletion (Ifitm-del) fibroblasts and, reciprocally, reduced in fibroblasts transcomplemented with Ifitm3. Mechanistic studies showed that Ifitm3 did not affect viral binding or entry but inhibited pH-dependent fusion. In a murine model of chikungunya virus arthritis, Ifitm3(-/-) mice sustained greater joint swelling in the ipsilateral ankle at days 3 and 7 postinfection, and this correlated with higher levels of proinflammatory cytokines and viral burden. Flow cytometric analysis suggested that Ifitm3(-/-) macrophages from the spleen were infected at greater levels than observed in wild-type (WT) mice, results that were supported by experiments with Ifitm3(-/-) bone marrow-derived macrophages. Ifitm3(-/-) mice also were more susceptible than WT mice to lethal alphavirus infection with Venezuelan equine encephalitis virus, and this was associated with greater viral burden in multiple organs. Collectively, our data define an antiviral role for Ifitm3 in restricting infection of multiple alphaviruses. IMPORTANCE The interferon-induced transmembrane protein 3 (IFITM3) inhibits infection of multiple families of viruses in cell culture. Compared to other viruses, much less is known about the antiviral effect of IFITM3 on alphaviruses. In this study, we characterized the antiviral activity of mouse Ifitm3 against arthritogenic and encephalitic alphaviruses using cells and animals with a targeted gene deletion of Ifitm3 as well as deficient cells transcomplemented with Ifitm3. Based on extensive virological analysis, we demonstrate greater levels of alphavirus infection and disease pathogenesis when Ifitm3 expression is absent. Our data establish an inhibitory role for Ifitm3 in controlling infection of alphaviruses.
Collapse
|
86
|
Abstract
Osteogenesis imperfecta is a phenotypically and molecularly heterogeneous group of inherited connective tissue disorders that share similar skeletal abnormalities causing bone fragility and deformity. Previously, the disorder was thought to be an autosomal dominant bone dysplasia caused by defects in type I collagen, but in the past 10 years discoveries of novel (mainly recessive) causative genes have lent support to a predominantly collagen-related pathophysiology and have contributed to an improved understanding of normal bone development. Defects in proteins with very different functions, ranging from structural to enzymatic and from intracellular transport to chaperones, have been described in patients with osteogenesis imperfecta. Knowledge of the specific molecular basis of each form of the disorder will advance clinical diagnosis and potentially stimulate targeted therapeutic approaches. In this Seminar, together with diagnosis, management, and treatment, we describe the defects causing osteogenesis imperfecta and their mechanism and interrelations, and classify them into five groups on the basis of the metabolic pathway compromised, specifically those related to collagen synthesis, structure, and processing; post-translational modification; folding and cross-linking; mineralisation; and osteoblast differentiation.
Collapse
Affiliation(s)
- Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Joan C Marini
- Bone and Extracellular Matrix Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
87
|
Ling S, Zhang C, Wang W, Cai X, Yu L, Wu F, Zhang L, Tian C. Combined approaches of EPR and NMR illustrate only one transmembrane helix in the human IFITM3. Sci Rep 2016; 6:24029. [PMID: 27046158 PMCID: PMC4820770 DOI: 10.1038/srep24029] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/18/2016] [Indexed: 11/09/2022] Open
Abstract
Interferon-inducible transmembrane protein IFITM3 was known to restrict the entry of a wide spectrum of viruses to the cytosol of the host. The mechanism used by the protein to restrict viral entry is unclear given the unavailability of the membrane topology and structures of the IFITM family proteins. Systematic site-directed spin labeling (SDSL) and electron paramagnetic resonance (EPR) studies of IFITM3 in detergent micelles identified a single, long transmembrane helix in the C-terminus and an intramembrane segment in the N-terminal hydrophobic region. Solution NMR studies of the same sample verified the secondary structure distribution and demonstrated two rigid regions interacting with the micellar surface. The resulting membrane topology of IFITM3 supports the mechanism of an enhanced restricted membrane hemi-fusion.
Collapse
Affiliation(s)
- Shenglong Ling
- Hefei National Laboratory of Microscale Physical Sciences, School of Life Science, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Chengwei Zhang
- Hefei National Laboratory of Microscale Physical Sciences, School of Life Science, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Wei Wang
- Hefei National Laboratory of Microscale Physical Sciences, School of Life Science, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Xiaoying Cai
- Hefei National Laboratory of Microscale Physical Sciences, School of Life Science, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Lu Yu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, 230031, P. R. China
| | - Fangming Wu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, 230031, P. R. China
| | - Longhua Zhang
- Hefei National Laboratory of Microscale Physical Sciences, School of Life Science, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Changlin Tian
- Hefei National Laboratory of Microscale Physical Sciences, School of Life Science, University of Science and Technology of China, Hefei, 230027, P. R. China.,High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, 230031, P. R. China
| |
Collapse
|
88
|
Londrigan SL, Tate MD, Job ER, Moffat JM, Wakim LM, Gonelli CA, Purcell DFJ, Brooks AG, Villadangos JA, Reading PC, Mintern JD. Endogenous Murine BST-2/Tetherin Is Not a Major Restriction Factor of Influenza A Virus Infection. PLoS One 2015; 10:e0142925. [PMID: 26566124 PMCID: PMC4643895 DOI: 10.1371/journal.pone.0142925] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/28/2015] [Indexed: 01/23/2023] Open
Abstract
BST-2 (tetherin, CD317, HM1.24) restricts virus growth by tethering enveloped viruses to the cell surface. The role of BST-2 during influenza A virus infection (IAV) is controversial. Here, we assessed the capacity of endogenous BST-2 to restrict IAV in primary murine cells. IAV infection increased BST-2 surface expression by primary macrophages, but not alveolar epithelial cells (AEC). BST-2-deficient AEC and macrophages displayed no difference in susceptibility to IAV infection relative to wild type cells. Furthermore, BST-2 played little role in infectious IAV release from either AEC or macrophages. To examine BST-2 during IAV infection in vivo, we infected BST-2-deficient mice. No difference in weight loss or in viral loads in the lungs and/or nasal tissues were detected between BST-2-deficient and wild type animals. This study rules out a major role for endogenous BST-2 in modulating IAV in the mouse model of infection.
Collapse
Affiliation(s)
- Sarah L. Londrigan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Michelle D. Tate
- Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Monash University, Clayton, Victoria, 3168, Australia
| | - Emma R. Job
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Jessica M. Moffat
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Linda M. Wakim
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Christopher A. Gonelli
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Damien F. J. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Andrew G. Brooks
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Jose A. Villadangos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia
| | - Patrick C. Reading
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Justine D. Mintern
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia
- * E-mail:
| |
Collapse
|
89
|
Infection of Mouse Macrophages by Seasonal Influenza Viruses Can Be Restricted at the Level of Virus Entry and at a Late Stage in the Virus Life Cycle. J Virol 2015; 89:12319-29. [PMID: 26423941 DOI: 10.1128/jvi.01455-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/22/2015] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED Airway epithelial cells are susceptible to infection with seasonal influenza A viruses (IAV), resulting in productive virus replication and release. Macrophages (MΦ) are also permissive to IAV infection; however, virus replication is abortive. Currently, it is unclear how productive infection of MΦ is impaired or the extent to which seasonal IAV replicate in MΦ. Herein, we compared mouse MΦ and epithelial cells for their ability to support genomic replication and transcription, synthesis of viral proteins, assembly of virions, and release of infectious progeny following exposure to genetically defined IAV. We confirm that seasonal IAV differ in their ability to utilize cell surface receptors for infectious entry and that this represents one level of virus restriction. Following virus entry, we demonstrate synthesis of all eight segments of genomic viral RNA (vRNA) and mRNA, as well as seven distinct IAV proteins, in IAV-infected mouse MΦ. Although newly synthesized hemagglutinin (HA) and neuraminidase (NA) glycoproteins are incorporated into the plasma membrane and expressed at the cell surface, electron microscopy confirmed that virus assembly was defective in IAV-infected MΦ, defining a second level of restriction late in the virus life cycle. IMPORTANCE Seasonal influenza A viruses (IAV) and highly pathogenic avian influenza viruses (HPAI) infect macrophages, but only HPAI replicate productively in these cells. Herein, we demonstrate that impaired virus uptake into macrophages represents one level of restriction limiting infection by seasonal IAV. Following uptake, seasonal IAV do not complete productive replication in macrophages, representing a second level of restriction. Using murine macrophages, we demonstrate that productive infection is blocked late in the virus life cycle, such that virus assembly is defective and newly synthesized virions are not released. These studies represent an important step toward identifying host-encoded factors that block replication of seasonal IAV, but not HPAI, in macrophages.
Collapse
|
90
|
Narayana SK, Helbig KJ, McCartney EM, Eyre NS, Bull RA, Eltahla A, Lloyd AR, Beard MR. The Interferon-induced Transmembrane Proteins, IFITM1, IFITM2, and IFITM3 Inhibit Hepatitis C Virus Entry. J Biol Chem 2015; 290:25946-59. [PMID: 26354436 DOI: 10.1074/jbc.m115.657346] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Indexed: 01/15/2023] Open
Abstract
The interferon-induced transmembrane (IFITM) family of proteins have recently been identified as important host effector molecules of the type I interferon response against viruses. IFITM1 has been identified as a potent antiviral effector against hepatitis C virus (HCV), whereas the related family members IFITM2 and IFITM3 have been described to have antiviral effects against a broad range of RNA viruses. Here, we demonstrate that IFITM2 and IFITM3 play an integral role in the interferon response against HCV and act at the level of late entry stages of HCV infection. We have established that in hepatocytes, IFITM2 and IFITM3 localize to the late and early endosomes, respectively, as well as the lysosome. Furthermore, we have demonstrated that S-palmitoylation of all three IFITM proteins is essential for anti-HCV activity, whereas the conserved tyrosine residue in the N-terminal domain of IFITM2 and IFITM3 plays a significant role in protein localization. However, this tyrosine was found to be dispensable for anti-HCV activity, with mutation of the tyrosine resulting in an IFITM1-like phenotype with the retention of anti-HCV activity and co-localization of IFITM2 and IFITM3 with CD81. In conclusion, we propose that the IFITM proteins act in a coordinated manner to restrict HCV infection by targeting the endocytosed HCV virion for lysosomal degradation and demonstrate that the actions of the IFITM proteins are indeed virus and cell-type specific.
Collapse
Affiliation(s)
- Sumudu K Narayana
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Karla J Helbig
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Erin M McCartney
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Nicholas S Eyre
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Rowena A Bull
- the Inflammation and Infection Research Centre, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Auda Eltahla
- the Inflammation and Infection Research Centre, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Andrew R Lloyd
- the Inflammation and Infection Research Centre, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Michael R Beard
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| |
Collapse
|
91
|
Amin R, Marfak A, Pangault C, Oblet C, Chanut A, Tarte K, Denizot Y, Cogné M. The class-specific BCR tonic signal modulates lymphomagenesis in a c-myc deregulation transgenic model. Oncotarget 2015; 5:8995-9006. [PMID: 25229630 PMCID: PMC4253413 DOI: 10.18632/oncotarget.2297] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Deregulation of c-myc by translocation onto immunoglobulin (Ig) loci can promote B cell malignant proliferations with phenotypes as diverse as acute lymphoid leukemia, Burkitt lymphoma, diffuse large B cell lymphoma, myeloma… The B cell receptor (BCR) normally providing tonic signals for cell survival and mitogenic responses to antigens, can also contribute to lymphomagenesis upon sustained ligand binding or activating mutations. BCR signaling varies among cell compartments and BCR classes. For unknown reasons, some malignancies associate with expression of either IgM or class-switched Ig. We explored whether an IgA BCR, with strong tonic signaling, would affect lymphomagenesis in c-myc IgH 3′RR transgenic mice prone to lymphoproliferations. Breeding c-myc transgenics in a background where IgM expression was replaced with IgA delayed lymphomagenesis. By comparison to single c-myc transgenics, lymphomas from double mutant animals were more differentiated and less aggressive, with an altered transcriptional program. Larger tumor cells more often expressed CD43 and CD138, which culminated in a plasma cell phenotype in 10% of cases. BCR class-specific signals thus appear to modulate lymphomagenesis and may partly explain the observed association of specific Ig classes with human B cell malignancies of differential phenotype, progression and prognosis.
Collapse
Affiliation(s)
- Rada Amin
- Centre National de la Recherche Scientifique, Limoges, France. Université de Limoges, Limoges, France. INSERM UMR U917, Rennes, France
| | | | | | - Christelle Oblet
- Centre National de la Recherche Scientifique, Limoges, France. Université de Limoges, Limoges, France
| | - Aurélie Chanut
- Centre National de la Recherche Scientifique, Limoges, France. Université de Limoges, Limoges, France
| | | | - Yves Denizot
- Centre National de la Recherche Scientifique, Limoges, France. Université de Limoges, Limoges, France
| | - Michel Cogné
- Centre National de la Recherche Scientifique, Limoges, France. Université de Limoges, Limoges, France
| |
Collapse
|
92
|
Blondel D, Maarifi G, Nisole S, Chelbi-Alix MK. Resistance to Rhabdoviridae Infection and Subversion of Antiviral Responses. Viruses 2015; 7:3675-702. [PMID: 26198243 PMCID: PMC4517123 DOI: 10.3390/v7072794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 12/13/2022] Open
Abstract
Interferon (IFN) treatment induces the expression of hundreds of IFN-stimulated genes (ISGs). However, only a selection of their products have been demonstrated to be responsible for the inhibition of rhabdovirus replication in cultured cells; and only a few have been shown to play a role in mediating the antiviral response in vivo using gene knockout mouse models. IFNs inhibit rhabdovirus replication at different stages via the induction of a variety of ISGs. This review will discuss how individual ISG products confer resistance to rhabdoviruses by blocking viral entry, degrading single stranded viral RNA, inhibiting viral translation or preventing release of virions from the cell. Furthermore, this review will highlight how these viruses counteract the host IFN system.
Collapse
Affiliation(s)
- Danielle Blondel
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS UMR 9198, Université Paris-Sud, Gif-sur-Yvette 91190, France.
| | - Ghizlane Maarifi
- INSERM UMR-S 1124, Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris (FR 3567, CNRS), 75270 Paris Cedex 6, France.
| | - Sébastien Nisole
- INSERM UMR-S 1124, Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris (FR 3567, CNRS), 75270 Paris Cedex 6, France.
| | - Mounira K Chelbi-Alix
- INSERM UMR-S 1124, Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris (FR 3567, CNRS), 75270 Paris Cedex 6, France.
| |
Collapse
|
93
|
Chesarino NM, McMichael TM, Yount JS. Regulation of the trafficking and antiviral activity of IFITM3 by post-translational modifications. Future Microbiol 2015; 9:1151-63. [PMID: 25405885 DOI: 10.2217/fmb.14.65] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IFITM3 restricts cellular infection by multiple important viral pathogens, and is particularly critical for the innate immune response against influenza virus. Expression of IFITM3 expands acidic endolysosomal compartments and prevents fusion of endocytosed viruses, leading to their degradation. This small, 133 amino acid, antiviral protein is controlled by at least four distinct post-translational modifications. Positive regulation of IFITM3 antiviral activity is provided by S-palmitoylation, while negative regulatory mechanisms include lysine ubiquitination, lysine methylation and tyrosine phosphorylation. Herein, we describe specific insights into IFITM3 trafficking and activity that were provided by studies of IFITM3 post-translational modifications, and discuss evidence suggesting that IFITM3 adopts multiple membrane topologies involving at least one intramembrane domain in its antivirally active conformation.
Collapse
Affiliation(s)
- Nicholas M Chesarino
- Department of Microbial Infection & Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
94
|
Suo S, Ren X. Importance of interferon inducible trans-membrane proteins and retinoic acid inducible gene I for influenza virus replication: A review. Crit Rev Microbiol 2015; 42:75-82. [PMID: 25811941 DOI: 10.3109/1040841x.2014.886553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Understanding the interplay between Influenza viruses and host cells is key to elucidating the pathogenesis of these viruses. Several host factors have been identified that exert antiviral functions; however, influenza viruses continue to replicate utilizing host cell machinery. Herein, we review the mechanisms of action of two host-derived proteins on conferring cellular resistance to the influenza virus; (1) the interferon inducible trans-membrane proteins, 1, 2 and 3, a recently identified family of early restriction factors; and (2) retinoic acid inducible gene I, a key mediator of antiviral immunity. These data may contribute to the design of novel and efficient anti-influenza treatments.
Collapse
Affiliation(s)
- Siqingaowa Suo
- a College of Veterinary Medicine, Northeast Agricultural University , Xiangfang , Harbin , China
| | - Xiaofeng Ren
- a College of Veterinary Medicine, Northeast Agricultural University , Xiangfang , Harbin , China
| |
Collapse
|
95
|
Jia R, Ding S, Pan Q, Liu SL, Qiao W, Liang C. The C-terminal sequence of IFITM1 regulates its anti-HIV-1 activity. PLoS One 2015; 10:e0118794. [PMID: 25738301 PMCID: PMC4349745 DOI: 10.1371/journal.pone.0118794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/11/2015] [Indexed: 11/28/2022] Open
Abstract
The interferon-inducible transmembrane (IFITM) proteins inhibit a wide range of viruses. We previously reported the inhibition of human immunodeficiency virus type 1 (HIV-1) strain BH10 by human IFITM1, 2 and 3. It is unknown whether other HIV-1 strains are similarly inhibited by IFITMs and whether there exists viral countermeasure to overcome IFITM inhibition. We report here that the HIV-1 NL4-3 strain (HIV-1NL4-3) is not restricted by IFITM1 and its viral envelope glycoprotein is partly responsible for this insensitivity. However, HIV-1NL4-3 is profoundly inhibited by an IFITM1 mutant, known as Δ(117–125), which is deleted of 9 amino acids at the C-terminus. In contrast to the wild type IFITM1, which does not affect HIV-1 entry, the Δ(117–125) mutant diminishes HIV-1NL4-3 entry by 3-fold. This inhibition correlates with the predominant localization of Δ(117–125) to the plasma membrane where HIV-1 entry occurs. In spite of strong conservation of IFITM1 among most species, mouse IFITM1 is 19 amino acids shorter at its C-terminus as compared to human IFITM1 and, like the human IFITM1 mutant Δ(117–125), mouse IFITM1 also inhibits HIV-1 entry. This is the first report illustrating the role of viral envelope protein in overcoming IFITM1 restriction. The results also demonstrate the importance of the C-terminal region of IFITM1 in modulating the antiviral function through controlling protein subcellular localization.
Collapse
Affiliation(s)
- Rui Jia
- Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin, 300071, China
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
| | - Shilei Ding
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
- Department of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Qinghua Pan
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
| | - Shan-Lu Liu
- Department of Molecular Microbiology & Immunology, School of Medicine, Bond Life Sciences Center, University of Missouri, Columbia, Missouri, 65211–7310, United States of America
| | - Wentao Qiao
- Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
- Department of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
- * E-mail:
| |
Collapse
|
96
|
Benfield CTO, Smith SE, Wright E, Wash RS, Ferrara F, Temperton NJ, Kellam P. Bat and pig IFN-induced transmembrane protein 3 restrict cell entry by influenza virus and lyssaviruses. J Gen Virol 2015; 96:991-1005. [PMID: 25614588 PMCID: PMC4631062 DOI: 10.1099/vir.0.000058] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/14/2015] [Indexed: 12/15/2022] Open
Abstract
IFN-induced transmembrane protein 3 (IFITM3) is a restriction factor that blocks cytosolic entry of numerous viruses that utilize acidic endosomal entry pathways. In humans and mice, IFITM3 limits influenza-induced morbidity and mortality. Although many IFITM3-sensitive viruses are zoonotic, whether IFITMs function as antiviral restriction factors in mammalian species other than humans and mice is unknown. Here, IFITM3 orthologues in the microbat (Myotis myotis) and pig (Sus scrofa domesticus) were identified using rapid amplification of cDNA ends. Amino acid residues known to be important for IFITM3 function were conserved in the pig and microbat orthologues. Ectopically expressed pig and microbat IFITM3 co-localized with transferrin (early endosomes) and CD63 (late endosomes/multivesicular bodies). Pig and microbat IFITM3 restricted cell entry mediated by multiple influenza haemagglutinin subtypes and lyssavirus glycoproteins. Expression of pig or microbat IFITM3 in A549 cells reduced influenza virus yields and nucleoprotein expression. Conversely, small interfering RNA knockdown of IFITM3 in pig NPTr cells and primary microbat cells enhanced virus replication, demonstrating that these genes are functional in their species of origin at endogenous levels. In summary, we showed that IFITMs function as potent broad-spectrum antiviral effectors in two mammals - pigs and bats - identified as major reservoirs for emerging viruses.
Collapse
Affiliation(s)
- Camilla T O Benfield
- Department of Pathology and Pathogen Biology, The Royal Veterinary College, Hatfield, UK
| | - Sarah E Smith
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Edward Wright
- Viral Pseudotype Unit (Fitzrovia), Faculty of Science and Technology, University of Westminster, London, UK
| | - Rachael S Wash
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Francesca Ferrara
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham Maritime, Kent, UK
| | - Nigel J Temperton
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham Maritime, Kent, UK
| | - Paul Kellam
- MRC/UCL Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London, UK.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| |
Collapse
|
97
|
Li K, Jia R, Li M, Zheng YM, Miao C, Yao Y, Ji HL, Geng Y, Qiao W, Albritton LM, Liang C, Liu SL. A sorting signal suppresses IFITM1 restriction of viral entry. J Biol Chem 2014; 290:4248-59. [PMID: 25527505 DOI: 10.1074/jbc.m114.630780] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The interferon-induced transmembrane proteins (IFITMs) broadly inhibit virus infections, particularly at the viral entry level. However, despite this shared ability to inhibit fusion, IFITMs differ in the potency and breadth of viruses restricted, an anomaly that is not fully understood. Here, we show that differences in the range of viruses restricted by IFITM1 are regulated by a C-terminal non-canonical dibasic sorting signal KRXX that suppresses restriction of some viruses by governing its intracellular distribution. Replacing the two basic residues with alanine (KR/AA) increased restriction of jaagsiekte sheep retrovirus and 10A1 amphotropic murine leukemia virus. Deconvolution microscopy revealed an altered subcellular distribution for KR/AA, with fewer molecules in LAMP1-positive lysosomes balanced by increased levels in CD63-positive multivesicular bodies, where jaagsiekte sheep retrovirus pseudovirions are colocalized. IFITM1 binds to cellular adaptor protein complex 3 (AP-3), an association that is lost when the dibasic motif is altered. Although knockdown of AP-3 itself decreases some virus entry, expression of parental IFITM1, but not its KR/AA mutant, potentiates inhibition of viral infections in AP-3 knockdown cells. By using the substituted cysteine accessibility method, we provide evidence that IFITM1 adopts more than one membrane topology co-existing in cellular membranes. Because the C-terminal dibasic sorting signal is unique to human IFITM1, our results provide novel insight into understanding the species- and virus-specific antiviral effect of IFITMs.
Collapse
Affiliation(s)
- Kun Li
- From the Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211
| | - Rui Jia
- the Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Minghua Li
- From the Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211
| | - Yi-Min Zheng
- From the Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211
| | - Chunhui Miao
- From the Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211
| | - Yunfang Yao
- the Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hong-Long Ji
- the Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas Health Science Center, Tyler, Texas 75708
| | - Yunqi Geng
- the Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Wentao Qiao
- the Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lorraine M Albritton
- the Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Chen Liang
- the McGill AIDS Centre, Lady Davis Institute, Montreal, Quebec H3T 1E2, Canada, and the Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Shan-Lu Liu
- From the Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211,
| |
Collapse
|
98
|
Compton AA, Bruel T, Porrot F, Mallet A, Sachse M, Euvrard M, Liang C, Casartelli N, Schwartz O. IFITM proteins incorporated into HIV-1 virions impair viral fusion and spread. Cell Host Microbe 2014; 16:736-47. [PMID: 25464829 PMCID: PMC7104936 DOI: 10.1016/j.chom.2014.11.001] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/16/2014] [Accepted: 10/15/2014] [Indexed: 10/28/2022]
Abstract
The interferon-induced transmembrane (IFITM) proteins protect cells from diverse virus infections by inhibiting virus-cell fusion. IFITM proteins also inhibit HIV-1 replication through mechanisms only partially understood. We show that when expressed in uninfected lymphocytes, IFITM proteins exert protective effects during cell-free virus infection, but this restriction can be overcome upon HIV-1 cell-to-cell spread. However, when present in virus-producing lymphocytes, IFITM proteins colocalize with viral Env and Gag proteins and incorporate into nascent HIV-1 virions to limit entry into new target cells. IFITM in viral membranes is associated with impaired virion fusion, offering additional and more potent defense against virus spread. Thus, IFITM proteins act additively in both productively infected cells and uninfected target cells to inhibit HIV-1 spread, potentially conferring these proteins with greater breadth and potency against enveloped viruses.
Collapse
Affiliation(s)
- Alex A Compton
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France.
| | - Timothée Bruel
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France
| | - Françoise Porrot
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France
| | - Adeline Mallet
- Institut Pasteur, Imagopole, Ultrastructural Microscopy Platform, Paris 75015, France
| | - Martin Sachse
- Institut Pasteur, Imagopole, Ultrastructural Microscopy Platform, Paris 75015, France
| | - Marine Euvrard
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; École Normale Supérieure, Department of Biology, Cachan 94230, France
| | - Chen Liang
- Lady Davis Institute, McGill AIDS Centre, Montreal, QC H3T 1E2, Canada
| | - Nicoletta Casartelli
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France
| | - Olivier Schwartz
- Institut Pasteur, Department of Virology, Virus & Immunity Unit, Paris 75015, France; CNRS URA 3015, Paris 75015, France; Vaccine Research Institute, Creteil 94010, France.
| |
Collapse
|
99
|
Qian J, Le Duff Y, Wang Y, Pan Q, Ding S, Zheng YM, Liu SL, Liang C. Primate lentiviruses are differentially inhibited by interferon-induced transmembrane proteins. Virology 2014; 474:10-8. [PMID: 25463599 PMCID: PMC4581848 DOI: 10.1016/j.virol.2014.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 09/29/2014] [Accepted: 10/16/2014] [Indexed: 01/09/2023]
Abstract
Interferon-induced transmembrane (IFITM) proteins inhibit the entry of a large number of viruses. Not surprisingly, many viruses are refractory to this inhibition. In this study, we report that different strains of HIV and SIV are inhibited by human IFITM proteins to various degrees, with SIV of African green monkeys (SIVAGM) being mostly restricted by human IFITM2. Interestingly, SIVAGM is as much inhibited by human IFITM2 as by IFITM3 of its own host African green monkeys. Our data further demonstrate that the entry of SIVAGM is impaired by human IFITM2 and that this inhibition is overcome by the cholesterol-binding compound amphotericin B that also overcomes IFITM inhibition of influenza A viruses. These results suggest that IFITM proteins exploit similar mechanisms to inhibit the entry of both pH-independent primate lentiviruses and the pH-dependent influenza A viruses. HIV and SIV strains are differentially inhibited by IFITM proteins. Human IFITM2 protein potently inhibits SIVAGM by impeding virus entry. IFITM3 of African green monkeys inhibits both HIV-1 and SIVAGM. The cholesterol-binding agent amphotericin B overcomes IFITM3 inhibition of SIVAGM.
Collapse
Affiliation(s)
- Jin Qian
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2; Department of Medicine, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Yann Le Duff
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2
| | - Yimeng Wang
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2; Department of Medicine, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Qinghua Pan
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2
| | - Shilei Ding
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2; Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Yi-Min Zheng
- Department of Molecular Microbiology & Immunology, School of Medicine, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310, USA
| | - Shan-Lu Liu
- Department of Molecular Microbiology & Immunology, School of Medicine, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310, USA
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2; Department of Medicine, McGill University, Montreal, Quebec, Canada H3A 2B4; Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada H3A 2B4.
| |
Collapse
|
100
|
Abstract
Current dogma is that mouse primordial germ cells (PGCs) segregate within the allantois, or source of the umbilical cord, and translocate to the gonads, differentiating there into sperm and eggs. In light of emerging data on the posterior embryonic-extraembryonic interface, and the poorly studied but vital fetal-umbilical connection, we have reviewed the past century of experiments on mammalian PGCs and their relation to the allantois. We demonstrate that, despite best efforts and valuable data on the pluripotent state, what is and is not a PGC in vivo is obscure. Furthermore, sufficient experimental evidence has yet to be provided either for an extragonadal origin of mammalian PGCs or for their segregation within the posterior region. Rather, most evidence points to an alternative hypothesis that PGCs in the mouse allantois are part of a stem/progenitor cell pool that exhibits all known PGC "markers" and that builds/reinforces the fetal-umbilical interface, common to amniotes. We conclude by suggesting experiments to distinguish the mammalian germ line from the soma.
Collapse
|