Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 8(11): e80723, 2013.
Article in English | MEDLINE | ID: mdl-24278312

ABSTRACT

The interferon-inducible transmembrane (IFITM) family of proteins has been shown to restrict a broad range of viruses in vitro and in vivo by halting progress through the late endosomal pathway. Further, single nucleotide polymorphisms (SNPs) in its sequence have been linked with risk of developing severe influenza virus infections in humans. The number of viruses restricted by this host protein has continued to grow since it was first demonstrated as playing an antiviral role; all of which enter cells via the endosomal pathway. We therefore sought to test the limits of antimicrobial restriction by Ifitm3 using a knockout mouse model. We showed that Ifitm3 does not impact on the restriction or pathogenesis of bacterial (Salmonella typhimurium, Citrobacter rodentium, Mycobacterium tuberculosis) or protozoan (Plasmodium berghei) pathogens, despite in vitro evidence. However, Ifitm3 is capable of restricting respiratory syncytial virus (RSV) in vivo either through directly restricting RSV cell infection, or by exerting a previously uncharacterised function controlling disease pathogenesis. This represents the first demonstration of a virus that enters directly through the plasma membrane, without the need for the endosomal pathway, being restricted by the IFITM family; therefore further defining the role of these antiviral proteins.


Subject(s)
Bacteria/metabolism , Membrane Proteins/metabolism , Plasmodium berghei/physiology , Respiratory Syncytial Viruses/physiology , Animals , Citrobacter rodentium/growth & development , Citrobacter rodentium/physiology , Homeostasis , Kinetics , Malaria/parasitology , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/physiology , Phenotype , Plasmodium berghei/growth & development , Respiratory Syncytial Viruses/growth & development , Salmonella typhimurium/physiology
2.
Cell Rep ; 5(4): 895-908, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24268777

ABSTRACT

The IFITMs inhibit influenza A virus (IAV) replication in vitro and in vivo. Here, we establish that the antimycotic heptaen, amphotericin B (AmphoB), prevents IFITM3-mediated restriction of IAV, thereby increasing viral replication. Consistent with its neutralization of IFITM3, a clinical preparation of AmphoB, AmBisome, reduces the majority of interferon's protective effect against IAV in vitro. Mechanistic studies reveal that IFITM1 decreases host-membrane fluidity, suggesting both a possible mechanism for IFITM-mediated restriction and its negation by AmphoB. Notably, we reveal that mice treated with AmBisome succumbed to a normally mild IAV infection, similar to animals deficient in Ifitm3. Therefore, patients receiving antifungal therapy with clinical preparations of AmphoB may be functionally immunocompromised and thus more vulnerable to influenza, as well as other IFITM3-restricted viral infections.


Subject(s)
Amphotericin B/adverse effects , Antifungal Agents/adverse effects , Immunocompromised Host , Influenza A Virus, H1N1 Subtype/immunology , Membrane Proteins/genetics , Orthomyxoviridae Infections/immunology , Virus Internalization/drug effects , Acetylcholine/pharmacology , Amphotericin B/administration & dosage , Animals , Anti-Bacterial Agents/pharmacology , Antifungal Agents/administration & dosage , Antigens, Differentiation/metabolism , Biological Transport/drug effects , COS Cells , Cell Fusion , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Chlorocebus aethiops , HeLa Cells , Humans , Influenza, Human/immunology , Interferons/immunology , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Nystatin/pharmacology , RNA Interference , RNA, Small Interfering , Sodium/metabolism , Tetraethylammonium/pharmacology , Virus Replication/drug effects
3.
J Virol ; 87(14): 7837-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23658454

ABSTRACT

The interferon-induced transmembrane protein 3 (IFITM3) gene is an interferon-stimulated gene that inhibits the replication of multiple pathogenic viruses in vitro and in vivo. IFITM3 is a member of a large protein superfamily, whose members share a functionally undefined area of high amino acid conservation, the CD225 domain. We performed mutational analyses of IFITM3 and identified multiple residues within the CD225 domain, consisting of the first intramembrane domain (intramembrane domain 1 [IM1]) and a conserved intracellular loop (CIL), that are required for restriction of both influenza A virus (IAV) and dengue virus (DENV) infection in vitro. Two phenylalanines within IM1 (F75 and F78) also mediate a physical association between IFITM proteins, and the loss of this interaction decreases IFITM3-mediated restriction. By extension, similar IM1-mediated associations may contribute to the functions of additional members of the CD225 domain family. IFITM3's distal N-terminal domain is also needed for full antiviral activity, including a tyrosine (Y20), whose alteration results in mislocalization of a portion of IFITM3 to the cell periphery and surface. Comparative analyses demonstrate that similar molecular determinants are needed for IFITM3's restriction of both IAV and DENV. However, a portion of the CIL including Y99 and R87 is preferentially needed for inhibition of the orthomyxovirus. Several IFITM3 proteins engineered with rare single-nucleotide polymorphisms demonstrated reduced expression or mislocalization, and these events were associated with enhanced viral replication in vitro, suggesting that possessing such alleles may impact an individual's risk for viral infection. On the basis of this and other data, we propose a model for IFITM3-mediated restriction.


Subject(s)
Dengue Virus/physiology , Influenza A virus/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Virus Replication/physiology , Amino Acid Sequence , Animals , Cell Culture Techniques , Cloning, Molecular , Conserved Sequence/genetics , DNA Mutational Analysis , DNA, Complementary/genetics , Dogs , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Madin Darby Canine Kidney Cells , Mass Spectrometry , Microscopy, Confocal , Models, Biological , Molecular Sequence Data , Polymorphism, Single Nucleotide/genetics , Protein Structure, Tertiary/genetics , Virus Replication/genetics
4.
Nature ; 484(7395): 519-23, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446628

ABSTRACT

The 2009 H1N1 influenza pandemic showed the speed with which a novel respiratory virus can spread and the ability of a generally mild infection to induce severe morbidity and mortality in a subset of the population. Recent in vitro studies show that the interferon-inducible transmembrane (IFITM) protein family members potently restrict the replication of multiple pathogenic viruses. Both the magnitude and breadth of the IFITM proteins' in vitro effects suggest that they are critical for intrinsic resistance to such viruses, including influenza viruses. Using a knockout mouse model, we now test this hypothesis directly and find that IFITM3 is essential for defending the host against influenza A virus in vivo. Mice lacking Ifitm3 display fulminant viral pneumonia when challenged with a normally low-pathogenicity influenza virus, mirroring the destruction inflicted by the highly pathogenic 1918 'Spanish' influenza. Similar increased viral replication is seen in vitro, with protection rescued by the re-introduction of Ifitm3. To test the role of IFITM3 in human influenza virus infection, we assessed the IFITM3 alleles of individuals hospitalized with seasonal or pandemic influenza H1N1/09 viruses. We find that a statistically significant number of hospitalized subjects show enrichment for a minor IFITM3 allele (SNP rs12252-C) that alters a splice acceptor site, and functional assays show the minor CC genotype IFITM3 has reduced influenza virus restriction in vitro. Together these data reveal that the action of a single intrinsic immune effector, IFITM3, profoundly alters the course of influenza virus infection in mouse and humans.


Subject(s)
Influenza A virus/pathogenicity , Membrane Proteins/metabolism , Orthomyxoviridae Infections/mortality , RNA-Binding Proteins/metabolism , Alleles , Amino Acid Sequence , Animals , Cytokines/immunology , England/epidemiology , Gene Deletion , Humans , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A virus/classification , Influenza A virus/growth & development , Influenza B virus/classification , Influenza B virus/growth & development , Influenza B virus/pathogenicity , Influenza, Human/complications , Influenza, Human/epidemiology , Influenza, Human/mortality , Influenza, Human/virology , Leukocytes/immunology , Lung/pathology , Lung/virology , Membrane Proteins/chemistry , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/pathology , Pneumonia, Viral/etiology , Pneumonia, Viral/pathology , Pneumonia, Viral/prevention & control , Polymorphism, Single Nucleotide/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Scotland/epidemiology , Virus Replication
5.
Blood ; 119(6): 1370-9, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22184403

ABSTRACT

Stem cell differentiation and lineage specification depend on coordinated programs of gene expression, but our knowledge of the chromatin-modifying factors regulating these events remains incomplete. Ubiquitination of histone H2A (H2A-K119u) is a common chromatin modification associated with gene silencing, and controlled by the ubiquitin-ligase polycomb repressor complex 1 (PRC1) and H2A-deubiquitinating enzymes (H2A-DUBs). The roles of H2A-DUBs in mammalian development, stem cells, and hematopoiesis have not been addressed. Here we characterized an H2A-DUB targeted mouse line Mysm1(tm1a/tm1a) and demonstrated defects in BM hematopoiesis, resulting in lymphopenia, anemia, and thrombocytosis. Development of lymphocytes was impaired from the earliest stages of their differentiation, and there was also a depletion of erythroid cells and a defect in erythroid progenitor function. These phenotypes resulted from a cell-intrinsic requirement for Mysm1 in the BM. Importantly, Mysm1(tm1a/tm1a) HSCs were functionally impaired, and this was associated with elevated levels of reactive oxygen species, γH2AX DNA damage marker, and p53 protein in the hematopoietic progenitors. Overall, these data establish a role for Mysm1 in the maintenance of BM stem cell function, in the control of oxidative stress and genetic stability in hematopoietic progenitors, and in the development of lymphoid and erythroid lineages.


Subject(s)
Cell Differentiation/genetics , Endopeptidases/genetics , Hematopoiesis/genetics , Lymphocytes/metabolism , Animals , Blood Cell Count , Blotting, Western , Endopeptidases/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Genotype , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Proteases
SELECTION OF CITATIONS
SEARCH DETAIL
...